1
|
Li MQ, He YQ, Zhang MN, Tang W, Tan Y, Cheng Y, Yang M, Zhao N, Li L, Yu SR, Li RL, Pan Q, Wu MY, Chai J. Dronedarone hydrochloride (DH) induces pancreatic cancer cell death by triggering mtDNA-mediated pyroptosis. Cell Death Dis 2024; 15:725. [PMID: 39358349 PMCID: PMC11447222 DOI: 10.1038/s41419-024-07102-w] [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: 02/06/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Pancreatic cancer is one of the leading causes of cancer-associated mortality, with a poor treatment approach. Previous study has shown that inducing pyroptosis in pancreatic ductal adenocarcinoma (PDAC) slows the growth of PDACs, implying that pyroptosis inducers are potentially effective for PDAC therapy. Here, we found that Dronedarone hydrochloride (DH), an antiarrhythmic drug, induces pyroptosis in pancreatic cancer cells and inhibits PDAC development in mice. In PANC-1 cells, DH caused cell death in a dosage- and time-dependent manner, with only pyroptosis inhibitors and GSDMD silencing rescuing the cell death, indicating that DH triggered GSDMD-dependent pyroptosis. Further work revealed that DH increased mitochondrial stresses and caused mitochondrial DNA (mtDNA) leakage, activating the cytosolic STING-cGAS and pyroptosis pathways. Finally, we assessed the anti-cancer effects of DH in a pancreatic cancer mouse model and found that DH treatment suppressed pancreatic tumor development in vivo. Collectively, our investigation demonstrates that DH triggers pyroptosis in PDAC and proposes its potential effects on anti-PDAC growth.
Collapse
Affiliation(s)
- Ming-Qiao Li
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yu-Qi He
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Meng-Ni Zhang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Wan Tang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ya Tan
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yue Cheng
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Mei Yang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Nan Zhao
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ling Li
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Si-Rui Yu
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ruo-Lan Li
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Qiong Pan
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ming-Yue Wu
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| | - Jin Chai
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD), the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| |
Collapse
|
2
|
Kelly T, Bhandari S, Carew M, Rubino R, Nicol C, Yang X. A Novel Bioluminescent Biosensor Quantifying Intramolecular Interaction and Levels of Pyroptosis Effector GSDMD. Cells 2024; 13:1606. [PMID: 39404370 PMCID: PMC11475789 DOI: 10.3390/cells13191606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Gasdermin D (GSDMD) is a key executor of pyroptosis, a form of inflammation-induced programmed cell death. Recently, GSDMD has been shown to play important roles in the development of various inflammatory-related human diseases including heart failure and cancer, suggesting that it is a promising therapeutic target for these diseases. While extensive studies on GSDMD's role in pyroptosis have been reported, it is challenging to study its function due to the lack of enzymatic activity of GSDMD. In this study, we used the NanoBiT technology to develop a novel GSDMD bioluminescent biosensor (GSDMD-BS) that detects the amount of non-cleaved GSDMD. This sensor allows us to quantify GSDMD's intramolecular interactions, the amounts of uncleaved GSDMD after caspase-1 cleavage, and expression levels in living cells. In vitro experiments using purified GSDMD-BS also confirmed the sensor's accuracy in reporting GSDMD levels and cleavage. Moreover, the potential for in vivo application was demonstrated in a xenograft mouse model. In conclusion, we have developed a GSDMD biosensor that is a valuable tool for real-time monitoring of GSDMD dynamics and pyroptosis. This biosensor will significantly expedite pyroptosis research and can be used for high-throughput screening for drugs targeting GSDMD for the therapy of many inflammation-related diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (T.K.); (S.B.); (M.C.); (R.R.); (C.N.)
| |
Collapse
|
3
|
Rodrigues AM, Paula Zen Petisco Fiore A, Guardia GDA, Tomasin R, Azevedo Reis Teixeira A, Giordano RJ, Schechtman D, Pagano M, Galante PAF, Bruni-Cardoso A. Identification of a novel alternative splicing isoform of the Hippo kinase STK3/MST2 with impaired kinase and cell growth suppressing activities. Oncogene 2024; 43:2938-2950. [PMID: 39174858 DOI: 10.1038/s41388-024-03104-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/27/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024]
Abstract
Mammalian Ste-20-like Kinases 1 and 2 (MST1/2) are core serine-threonine kinases of the Hippo pathway regulating several cellular processes, including cell cycle arrest and cell death. Here, we discovered a novel alternative splicing variant of the MST2 encoding gene, STK3, in malignant cells and tumor datasets. This variant, named STK3∆7 or MST2∆7 (for mRNA or protein, respectively), resulted from the skipping of exon 7. MST2∆7 exhibited increased ubiquitylation and interaction with the E3 ubiquitin-protein ligase CHIP compared to the full-length protein (MST2FL). Exon 7 in STK3 encodes a segment within the kinase domain, and its exclusion compromised MST2 interaction with and phosphorylation of MOB, a major MST1/2 substrate. Nevertheless, MST2∆7 was capable of interacting with MST1 and MST2FL. Unlike MST2FL, overexpression of MST2∆7 did not lead to increased cell death and growth arrest. Strikingly, we observed the exclusion of STK3 exon 7 in 3.2-15% of tumor samples from patients of several types of cancer, while STK3∆7 was seldomly found in healthy tissues. Our study identified a novel STK3 splicing variant with loss of function and the potential to disturb tissue homeostasis by impacting on MST2 activities in the regulation of cell death and quiescence.
Collapse
Affiliation(s)
- Ana Maria Rodrigues
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Paula Zen Petisco Fiore
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
- Department of Biology, New York University, New York, NY, USA
| | | | - Rebeka Tomasin
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | - Ricardo Jose Giordano
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Deborah Schechtman
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Pedro A F Galante
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Alexandre Bruni-Cardoso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
4
|
Wang S, He H, Qu L, Shen Q, Dai Y. Dual roles of inflammatory programmed cell death in cancer: insights into pyroptosis and necroptosis. Front Pharmacol 2024; 15:1446486. [PMID: 39257400 PMCID: PMC11384570 DOI: 10.3389/fphar.2024.1446486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/16/2024] [Indexed: 09/12/2024] Open
Abstract
Programmed cell death (PCD) is essential for cellular homeostasis and defense against infections, with inflammatory forms like pyroptosis and necroptosis playing significant roles in cancer. Pyroptosis, mediated by caspases and gasdermin proteins, leads to cell lysis and inflammatory cytokine release. It has been implicated in various diseases, including cancer, where it can either suppress tumor growth or promote tumor progression through chronic inflammation. Necroptosis, involving RIPK1, RIPK3, and MLKL, serves as a backup mechanism when apoptosis is inhibited. In cancer, necroptosis can enhance immune responses or contribute to tumor progression. Both pathways have dual roles in cancer, acting as tumor suppressors or promoting a pro-tumorigenic environment depending on the context. This review explores the molecular mechanisms of pyroptosis and necroptosis, their roles in different cancers, and their potential as therapeutic targets. Understanding the context-dependent effects of these pathways is crucial for developing effective cancer therapies.
Collapse
Affiliation(s)
- Shuai Wang
- Collage of Medicine, Xinyang Normal University, Xinyang, China
| | - Huanhuan He
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lailiang Qu
- Collage of Medicine, Xinyang Normal University, Xinyang, China
| | - Qianhe Shen
- Collage of Medicine, Xinyang Normal University, Xinyang, China
| | - Yihang Dai
- Collage of Medicine, Xinyang Normal University, Xinyang, China
| |
Collapse
|
5
|
An X, Yu W, Liu J, Tang D, Yang L, Chen X. Oxidative cell death in cancer: mechanisms and therapeutic opportunities. Cell Death Dis 2024; 15:556. [PMID: 39090114 PMCID: PMC11294602 DOI: 10.1038/s41419-024-06939-5] [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: 03/08/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen-containing molecules generated as natural byproducts during cellular processes, including metabolism. Under normal conditions, ROS play crucial roles in diverse cellular functions, including cell signaling and immune responses. However, a disturbance in the balance between ROS production and cellular antioxidant defenses can lead to an excessive ROS buildup, causing oxidative stress. This stress damages essential cellular components, including lipids, proteins, and DNA, potentially culminating in oxidative cell death. This form of cell death can take various forms, such as ferroptosis, apoptosis, necroptosis, pyroptosis, paraptosis, parthanatos, and oxeiptosis, each displaying distinct genetic, biochemical, and signaling characteristics. The investigation of oxidative cell death holds promise for the development of pharmacological agents that are used to prevent tumorigenesis or treat established cancer. Specifically, targeting key antioxidant proteins, such as SLC7A11, GCLC, GPX4, TXN, and TXNRD, represents an emerging approach for inducing oxidative cell death in cancer cells. This review provides a comprehensive summary of recent progress, opportunities, and challenges in targeting oxidative cell death for cancer therapy.
Collapse
Affiliation(s)
- Xiaoqin An
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Wenfeng Yu
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Jinbao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Li Yang
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China.
| | - Xin Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
6
|
Wang X, Ding B, Liu W, Qi L, Li J, Zheng X, Song Y, Li Q, Wu J, Zhang M, Chen H, Wang Y, Li Y, Sun B, Ma P. Dual Starvations Induce Pyroptosis for Orthotopic Pancreatic Cancer Therapy through Simultaneous Deprivation of Glucose and Glutamine. J Am Chem Soc 2024; 146:17854-17865. [PMID: 38776361 DOI: 10.1021/jacs.4c03478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Pancreatic cancer is a highly fatal disease, and existing treatment methods are ineffective, so it is urgent to develop new effective treatment strategies. The high dependence of pancreatic cancer cells on glucose and glutamine suggests that disrupting this dependency could serve as an alternative strategy for pancreatic cancer therapy. We identified the vital genes glucose transporter 1 (GLUT1) and alanine-serine-cysteine transporter 2 (ASCT2) through bioinformatics analysis, which regulate glucose and glutamine metabolism in pancreatic cancer, respectively. Human serum albumin nanoparticles (HSA NPs) for delivery of GLUT1 and ASCT2 inhibitors, BAY-876/V-9302@HSA NPs, were prepared by a self-assembly process. This nanodrug inhibits glucose and glutamine uptake of pancreatic cancer cells through the released BAY-876 and V-9302, leading to nutrition deprivation and oxidative stress. The inhibition of glutamine leads to the inhibition of the synthesis of the glutathione, which further aggravates oxidative stress. Both of them lead to a significant increase in reactive oxygen species, activating caspase 1 and GSDMD and finally inducing pyroptosis. This study provides a new effective strategy for orthotopic pancreatic cancer treatment by dual starvation-induced pyroptosis. The study for screening metabolic targets using bioinformatics analysis followed by constructing nanodrugs loaded with inhibitors will inspire future targeted metabolic therapy for pancreatic cancer.
Collapse
Affiliation(s)
- Xinlong Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Wei Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Jiating Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xin Zheng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yiqin Song
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Qiyuan Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jiawen Wu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Meng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Hua Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yongwei Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
7
|
Argentiero A, Andriano A, Caradonna IC, de Martino G, Desantis V. Decoding the Intricate Landscape of Pancreatic Cancer: Insights into Tumor Biology, Microenvironment, and Therapeutic Interventions. Cancers (Basel) 2024; 16:2438. [PMID: 39001498 PMCID: PMC11240778 DOI: 10.3390/cancers16132438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant oncological challenges due to its aggressive nature and poor prognosis. The tumor microenvironment (TME) plays a critical role in progression and treatment resistance. Non-neoplastic cells, such as cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), contribute to tumor growth, angiogenesis, and immune evasion. Although immune cells infiltrate TME, tumor cells evade immune responses by secreting chemokines and expressing immune checkpoint inhibitors (ICIs). Vascular components, like endothelial cells and pericytes, stimulate angiogenesis to support tumor growth, while adipocytes secrete factors that promote cell growth, invasion, and treatment resistance. Additionally, perineural invasion, a characteristic feature of PDAC, contributes to local recurrence and poor prognosis. Moreover, key signaling pathways including Kirsten rat sarcoma viral oncogene (KRAS), transforming growth factor beta (TGF-β), Notch, hypoxia-inducible factor (HIF), and Wnt/β-catenin drive tumor progression and resistance. Targeting the TME is crucial for developing effective therapies, including strategies like inhibiting CAFs, modulating immune response, disrupting angiogenesis, and blocking neural cell interactions. A recent multi-omic approach has identified signature genes associated with anoikis resistance, which could serve as prognostic biomarkers and targets for personalized therapy.
Collapse
Affiliation(s)
| | - Alessandro Andriano
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, Medical School, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Ingrid Catalina Caradonna
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, Medical School, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giulia de Martino
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Vanessa Desantis
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, Medical School, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
8
|
Wang J, Wu Z, Zhu M, Zhao Y, Xie J. ROS induced pyroptosis in inflammatory disease and cancer. Front Immunol 2024; 15:1378990. [PMID: 39011036 PMCID: PMC11246884 DOI: 10.3389/fimmu.2024.1378990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
Pyroptosis, a form of caspase-1-dependent cell death, also known as inflammation-dependent death, plays a crucial role in diseases such as stroke, heart disease, or tumors. Since its elucidation, pyroptosis has attracted widespread attention from various sectors. Reactive oxygen species (ROS) can regulate numerous cellular signaling pathways. Through further research on ROS and pyroptosis, the level of ROS has been revealed to be pivotal for the occurrence of pyroptosis, establishing a close relationship between the two. This review primarily focuses on the molecular mechanisms of ROS and pyroptosis in tumors and inflammatory diseases, exploring key proteins that may serve as drug targets linking ROS and pyroptosis and emerging fields targeting pyroptosis. Additionally, the potential future development of compounds and proteins that influence ROS-regulated cell pyroptosis is anticipated, aiming to provide insights for the development of anti-tumor and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Ziyong Wu
- Department of Pharmacy, Ezhou Central Hospital, Ezhou, Hubei, China
| | - Min Zhu
- Department of Pharmacy, Xuchang Central Hospital, Xuchang, Henan, China
| | - Yang Zhao
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Jingwen Xie
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, China
- Department of Health, Chongqing Industry & Trade Polytechnic, Chongqing, China
| |
Collapse
|
9
|
Lin G, Xia A, Qiao J, Zhang H, Chen P, Zhou P, Hu Q, Xiang Z, Zhang S, Li L, Yang S. Identification of a new class of activators of the Hippo pathway with antitumor activity in vitro and in vivo. Biochem Pharmacol 2024; 224:116217. [PMID: 38641306 DOI: 10.1016/j.bcp.2024.116217] [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: 11/24/2023] [Revised: 03/05/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
The Hippo pathway is a key regulator of tissue growth, organ size, and tumorigenesis. Activating the Hippo pathway by gene editing or pharmaceutical intervention has been proven to be a new therapeutic strategy for treatment of the Hippo pathway-dependent cancers. To now, a number of compounds that directly target the downstream effector proteins of Hippo pathway, including YAP and TEADs, have been disclosed, but very few Hippo pathway activators are reported. Here, we discovered a new class of Hippo pathway activator, YL-602, which inhibited CTGF expression in cells irrespective of cell density and the presence of serum. Mechanistically, YL-602 activates the Hippo pathway via MST1/2, which is different from known activators of Hippo pathway. In vitro, YL-602 significantly induced tumor cell apoptosis and inhibited colony formation of tumor cells. In vivo, oral administration of YL-602 substantially suppressed the growth of cancer cells by activation of Hippo pathway. Overall, YL-602 could be a promising lead compound, and deserves further investigation for its mechanism of action and therapeutic applications.
Collapse
Affiliation(s)
- Guifeng Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Anjie Xia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingxin Qiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hailin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pei Chen
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Pei Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyu Xiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiyu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Zhou J, Li L, Wu B, Feng Z, Lu Y, Wang Z. MST1/2: Important regulators of Hippo pathway in immune system associated diseases. Cancer Lett 2024; 587:216736. [PMID: 38369002 DOI: 10.1016/j.canlet.2024.216736] [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: 11/17/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 02/20/2024]
Abstract
The Hippo signaling pathway is first found in Drosophila and is highly conserved in evolution. Previous studies on this pathway in mammals have revealed its key role in cell proliferation and differentiation, organ size control, and carcinogenesis. Apart from these, recent findings indicate that mammalian Ste20-like kinases 1 and 2 (MST1/2) have significant effects on immune regulation. In this review, we summarize the updated understanding of how MST1/2 affect the regulation of the immune system and the specific mechanism. The effect of MST1/2 on immune cells and its role in the tumor immune microenvironment can alter the body's response to tumor cells. The relationship between MST1/2 and the immune system suggests new directions in the manipulation of immune responses for clinical immunotherapy, especially for tumor treatment.
Collapse
Affiliation(s)
- Jingjing Zhou
- Department of Gastroenterology, Shanghai Xuhui Central Hospital and Department of Anatomy and Histoembrvology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lanfang Li
- Department of Gastroenterology, Shanghai Xuhui Central Hospital and Department of Anatomy and Histoembrvology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Baojin Wu
- Department of Plastic Surgery, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Jing'an District, Shanghai, 200040, China
| | - Zhen Feng
- Department of Gastroenterology, Shanghai Xuhui Central Hospital and Department of Anatomy and Histoembrvology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, China.
| | - Zuoyun Wang
- Department of Gastroenterology, Shanghai Xuhui Central Hospital and Department of Anatomy and Histoembrvology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
11
|
Lozon L, Ramadan WS, Kawaf RR, Al-Shihabi AM, El-Awady R. Decoding cell death signalling: Impact on the response of breast cancer cells to approved therapies. Life Sci 2024; 342:122525. [PMID: 38423171 DOI: 10.1016/j.lfs.2024.122525] [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: 11/25/2023] [Revised: 02/04/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Breast cancer is a principal cause of cancer-related mortality in female worldwide. While many approved therapies have shown promising outcomes in treating breast cancer, understanding the intricate signalling pathways controlling cell death is crucial for optimizing the treatment outcome. A growing body of evidence has unveiled the aberrations in multiple cell death pathways across diverse cancer types, highlighting these pathways as appealing targets for therapeutic interventions. In this review, we provide a comprehensive overview of the current state of knowledge on the cell death signalling mechanisms with a particular focus on their impact on the response of breast cancer cells to approved therapies. Additionally, we discuss the potentials of combination therapies that exploit the synergy between approved drugs and therapeutic agents targeting modulators of cell death pathways.
Collapse
Affiliation(s)
- Lama Lozon
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Wafaa S Ramadan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Rawan R Kawaf
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Aya M Al-Shihabi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Raafat El-Awady
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
12
|
Wang G, Chen J, Dai S, Zhang J, Gao Y, Yin L, Jiang K, Miao Y, Lu Z. High pyroptosis activity in pancreatic adenocarcinoma: poor prognosis and oxaliplatin resistance. Apoptosis 2024; 29:344-356. [PMID: 37848674 DOI: 10.1007/s10495-023-01901-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Pyroptosis, as a type of inflammatory programmed cell death, has been studied in inflammatory diseases and numerous cancers but its role in pancreatic ductal adenocarcinoma (PDAC) remains further exploration. METHODS A TCGA-PDAC cohort was enrolled for bioinformatics analysis to investigate the effect of pyroptosis on the prognosis and drug sensitivity of patients. PA-TU-8988T and CFPAC-1 cells were selected for investigating the role of GSDMC in PDAC. RESULTS A distinct classification pattern of PDAC mediated by 21 pyroptosis-related genes (PRGs) was identified. It was suggested that higher pyroptosis activity was associated with poor prognosis of patients and higher tumor proliferation rates. We further established a prognostic model based on three PRGs (GSDMC, CASP4 and NLRP1) and the TCGA-PDAC cohort was classified into low and high-risk subgroups. It is noteworthy that the high-risk group showed significantly higher tumor proliferation rates and was proved to be highly correlated with oxaliplatin resistance. Further experiments suggested that overexpression of GSDMC promoted the proliferation and oxaliplatin resistance of PA-TU-8988T cells in vitro and vivo, while downregulation of GSDMC showed opposite effects in CFPAC-1 cells. Finally, we found that the activation of pentose phosphate pathway (PPP) was the mechanism by which GSDMC overexpression promoted the proliferation and oxaliplatin resistance of pancreatic cancer cells. CONCLUSIONS In this study, we found that higher pyroptosis activity is associated with worse prognosis and oxaliplatin resistance of PDAC patients. In addition, as a core effector of pyroptosis, GSDMC promoted proliferation and oxaliplatin resistance of pancreatic cancer cells, which will provide new therapeutic target for PDAC patients.
Collapse
Affiliation(s)
- Guangfu Wang
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Jin Chen
- Department of Gynecological Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Shangnan Dai
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Jinfan Zhang
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yong Gao
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Lingdi Yin
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yi Miao
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Pancreas Institute, Nanjing Medical University, Nanjing, China.
- Pancreas Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China.
| | - Zipeng Lu
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Pancreas Institute, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Wang A, Wang Y, Du C, Yang H, Wang Z, Jin C, Hamblin MR. Pyroptosis and the tumor immune microenvironment: A new battlefield in ovarian cancer treatment. Biochim Biophys Acta Rev Cancer 2024; 1879:189058. [PMID: 38113952 DOI: 10.1016/j.bbcan.2023.189058] [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: 08/19/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
Ovarian cancer is a less common tumor in women compared to cervical or breast cancer, however it is more malignant and has worse outcomes. Ovarian cancer patients still have a five-year survival rate < 50% despite advances in therapy. Due to recent developments in immune checkpoint inhibitors (ICIs), cancer immunotherapy has attracted increased interest. Pyroptosis is a highly inflammatory form of cell death, which is essential for bridging innate and adaptive immunity, and is involved in immune regulation within the tumor microenvironment (TME). Recent research has shown that pyroptosis can promote immunotherapy of ovarian cancer, including treatment with chimeric antigen receptor T-cells (CAR-T) or ICIs. Moreover, inflammasomes, various signaling pathways and lncRNAs can all affect pyroptosis in ovarian cancer. Here we discuss how pyroptosis affects the development and progression of ovarian cancer as well as the TME. We also provide a summary of small molecule drugs that could target pyroptotic cell death processes and may be useful in ovarian cancer therapy.
Collapse
Affiliation(s)
- Aihong Wang
- Department of Gynecologic Oncology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, PR China
| | - Yin Wang
- Department of Gynecologic Oncology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, PR China
| | - Chenxiang Du
- Department of Gynecologic Oncology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, PR China
| | - Huilun Yang
- Department of Gynecologic Oncology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, PR China
| | - Zhengping Wang
- Department of Gynecologic Oncology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, PR China
| | - Canhui Jin
- Department of Gynecologic Oncology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, PR China.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| |
Collapse
|
14
|
Tan M, Mao J, Zheng J, Meng Y, Li J, Hao J, Shen H. Mammalian STE20-like kinase 1 inhibits synoviocytes activation in rheumatoid arthritis through mitochondrial dysfunction mediated by SIRT3/mTOR axis. Inflamm Res 2024; 73:415-432. [PMID: 38265688 DOI: 10.1007/s00011-023-01846-5] [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: 11/07/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Mammalian STE20-like kinase 1 (MST1) is involved in the occurrence of cancer and autoimmune diseases by regulating cell proliferation, differentiation, apoptosis and other functions. However, its role and downstream targets in rheumatoid arthritis (RA) remain unclear. METHODS The model of RA fibroblast-like synoviocytes (RA-FLSs) overexpressing MST1 was constructed by lentiviral transfection in vitro and analyzed the effects of MST1 on apoptosis, migration, invasion, and inflammation of RA-FLSs. The effect of MST1 on joint synovial membrane inflammation and bone destruction was observed in vivo by establishing a rat model of arthritis with complete Freund's adjuvant. RESULTS MST1 is down-regulated in RA-FLSs, and up-regulation of MST1 inhibits the survival, migration, invasion and inflammation of RA-FLSs. Mechanistically, MST1 inhibits SIRT3/mTOR-signaling pathway, inducing decreased mitochondrial autophagy and increased mitochondrial fission, resulting in mitochondrial morphological abnormalities and dysfunction, and ultimately increased apoptosis. We have observed that activation of MST1 alleviates synovial inflammation and bone erosion in vivo. CONCLUSIONS MST1 reduces the survival, migration, invasion and inflammation of FLSs by inhibiting the SIRT3/mTOR axis to reduce mitochondrial autophagy and promote mitochondrial division, thereby achieving the potential role of relieving rheumatoid arthritis.
Collapse
Affiliation(s)
- Min Tan
- Department of Rheumatology, Lanzhou University Second Hospital, No. 82, Cui Ying Men Street, Lanzhou City, 730030, Gansu Province, People's Republic of China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Jing Mao
- Department of Rheumatology, Lanzhou University Second Hospital, No. 82, Cui Ying Men Street, Lanzhou City, 730030, Gansu Province, People's Republic of China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Jianxiong Zheng
- Department of Rheumatology, Lanzhou University Second Hospital, No. 82, Cui Ying Men Street, Lanzhou City, 730030, Gansu Province, People's Republic of China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Yu Meng
- Department of Pain, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Jun Li
- Department of Rheumatology, Lanzhou University Second Hospital, No. 82, Cui Ying Men Street, Lanzhou City, 730030, Gansu Province, People's Republic of China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Jiayao Hao
- Department of Rheumatology, Lanzhou University Second Hospital, No. 82, Cui Ying Men Street, Lanzhou City, 730030, Gansu Province, People's Republic of China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Haili Shen
- Department of Rheumatology, Lanzhou University Second Hospital, No. 82, Cui Ying Men Street, Lanzhou City, 730030, Gansu Province, People's Republic of China.
| |
Collapse
|
15
|
Xiong Y, Kong X, Mei H, Wang J, Zhou S. Bioinformatics-based analysis of the relationship between disulfidptosis and prognosis and treatment response in pancreatic cancer. Sci Rep 2023; 13:22218. [PMID: 38097783 PMCID: PMC10721597 DOI: 10.1038/s41598-023-49752-4] [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: 09/02/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023] Open
Abstract
Tumor formation is closely associated with disulfidptosis, a new form of cell death induced by disulfide stress-induced. The exact mechanism of action of disulfidptosis in pancreatic cancer (PCa) is not clear. This study analyzed the impact of disulfidptosis-related genes (DRGs) on the prognosis of PCa and identified clusters of DRGs, and based on this, a risk score (RS) signature was developed to assess the impact of RS on the prognosis, immune and chemotherapeutic response of PCa patients. Based on transcriptomic data and clinical information from PCa tissue and normal pancreatic tissue samples obtained from the TCGA and GTEx databases, differentially expressed and differentially surviving DRGs in PCa were identified from among 15 DRGs. Two DRGs clusters were identified by consensus clustering by merging the PCa samples in the GSE183795 dataset. Analysis of DRGs clusters about the PCa tumor microenvironment and differential analysis to obtain differential genes between the two DRG clusters. Patients were then randomized into the training and testing sets, and a prognostic prediction signature associated with disulfidptosis was constructed in the training set. Then all samples were divided into high-disulfidptosis-risk (HDR) and low-disulfidptosis-risk (LDR) subgroups based on the RS calculated from the signature. The predictive efficacy of the signature was assessed by survival analysis, nomograms, correlation analysis of clinicopathological characteristics, and the receiver operating characteristic (ROC) curves. To assess differences between different risk subgroups in immune cell infiltration, expression of immune checkpoint molecules, somatic gene mutations, and effectiveness of immunotherapy and chemotherapy. The GSE57495 dataset was used as external validation, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression levels of DRGs. A total of 12 DRGs with differential expression and prognosis in PCa were identified, based on which a risk-prognosis signature containing five differentially expressed genes (DEGs) was developed. The signature was a good predictor and an independent risk factor. The nomogram and calibration curve shows the signature's excellent clinical applicability. Functional enrichment analysis showed that RS was associated with tumor and immune-related pathways. RS was strongly associated with the tumor microenvironment, and analysis of response to immunotherapy and chemotherapy suggests that the signature can be used to assess the sensitivity of treatments. External validation further demonstrated the model's efficacy in predicting the prognosis of PCa patients, with RT-qPCR and immunohistochemical maps visualizing the expression of each gene in PCa cell lines and the tissue. Our study is the first to apply the subtyping model of disulfidptosis to PCa and construct a signature based on the disulfidptosis subtype, which can provide an accurate assessment of prognosis, immunotherapy, and chemotherapy response in PCa patients, providing new targets and directions for the prognosis and treatment of PCa.
Collapse
Affiliation(s)
- Yuanpeng Xiong
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiaoyu Kong
- School of Public Health, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Haoran Mei
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jie Wang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shifa Zhou
- Department of Emergency Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
16
|
Hajibabaie F, Abedpoor N, Mohamadynejad P. Types of Cell Death from a Molecular Perspective. BIOLOGY 2023; 12:1426. [PMID: 37998025 PMCID: PMC10669395 DOI: 10.3390/biology12111426] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
The former conventional belief was that cell death resulted from either apoptosis or necrosis; however, in recent years, different pathways through which a cell can undergo cell death have been discovered. Various types of cell death are distinguished by specific morphological alterations in the cell's structure, coupled with numerous biological activation processes. Various diseases, such as cancers, can occur due to the accumulation of damaged cells in the body caused by the dysregulation and failure of cell death. Thus, comprehending these cell death pathways is crucial for formulating effective therapeutic strategies. We focused on providing a comprehensive overview of the existing literature pertaining to various forms of cell death, encompassing apoptosis, anoikis, pyroptosis, NETosis, ferroptosis, autophagy, entosis, methuosis, paraptosis, mitoptosis, parthanatos, necroptosis, and necrosis.
Collapse
Affiliation(s)
- Fatemeh Hajibabaie
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran;
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran
| | - Navid Abedpoor
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan 81551-39998, Iran
| | - Parisa Mohamadynejad
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran;
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord 88137-33395, Iran
| |
Collapse
|
17
|
Guo Y, Jiang Z, Chen Q, Xie D, Zhou Y, Yin W, Wang Z, Wang B, Ren C, Jiang X. Construction and experimental validation of a signature for predicting prognosis and immune infiltration analysis of glioma based on disulfidptosis-related lncRNAs. Front Immunol 2023; 14:1291385. [PMID: 38022537 PMCID: PMC10655028 DOI: 10.3389/fimmu.2023.1291385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Backgrounds Disulfidptosis, a newly discovered mechanism of programmed cell death, is believed to have a unique role in elucidating cancer progression and guiding cancer therapy strategies. However, no studies have yet explored this mechanism in glioma. Methods We downloaded data on glioma patients from online databases to address this gap. Subsequently, we identified disulfidptosis-related genes from published literature and verified the associated lncRNAs. Results Through univariate, multivariate, and least absolute shrinkage and selection operator (LASSO) regression algorithms analyses, we identified 10 lncRNAs. These were then utilized to construct prognostic prediction models, culminating in a risk-scoring signature. Reliability and validity tests demonstrated that the model effectively discerns glioma patients' prognosis outcomes. We also analyzed the relationship between the risk score and immune characteristics, and identified several drugs that may be effective for high-risk patients. In vitro experiments revealed that LINC02525 could enhances glioma cells' migration and invasion capacities. Additionally, knocking down LINC02525 was observed to promote glioma cell disulfidptosis. Conclusion This study delves into disulfidptosis-related lncRNAs in glioma, offering novel insights into glioma therapeutic strategies.
Collapse
Affiliation(s)
- Youwei Guo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhipeng Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Quan Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dongcheng Xie
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Yin
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zihan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Binbin Wang
- Department of Neurosurgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Caiping Ren
- Cancer Research Institute, Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Health Commission (NHC) Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xingjun Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
18
|
Zhong PC, Liu ZW, Xing QC, Chen J, Yang RP. Neferine inhibits the development of lung cancer cells by downregulating TGF-β to regulate MST1/ROS-induced pyroptosis. Kaohsiung J Med Sci 2023; 39:1106-1118. [PMID: 37698291 DOI: 10.1002/kjm2.12752] [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: 02/21/2023] [Revised: 07/04/2023] [Accepted: 07/31/2023] [Indexed: 09/13/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for ~85% of all lung cancer cases. Neferine is used as a traditional Chinese medicine with many pharmacological effects, including antitumor properties; however, it has not been reported whether neferine plays an anticancer role by causing pyroptosis in NSCLC cells. We used two typical lung cancer cell lines, A549 and H1299, and 42 lung cancer tissue samples to investigate the regulatory effects of neferine on TGF-β and MST1. We also treated lung cancer cells with different concentrations of neferine to study its effects on lung cancer cell survival, migration, invasion, and epithelial-mesenchymal transition (EMT) as well as on pyroptosis. Lentivirus-mediated gain-of-function studies of TGF-β and MST1 were applied to validate the roles of TGF-β and MST1 in lung cancer. Next, we used murine transplanted tumor models to evaluate the effect of neferine treatment on the metastatic capacity of lung cancer tissues. With increasing neferine concentration, the viability, migration, invasion, and EMT capacity of A549 and H1299 cells decreased, whereas pyroptosis increased. Neferine repressed TGF-β expression to modulate the induction of reactive oxygen species (ROS) by MST1. Overexpression of TGF-β in either in vitro or mouse-transplanted A549 cells restored the inhibitory effect of neferine on tumor development. Overexpression of MST1 clearly enhanced pyroptosis. Neferine contributed to pyroptosis by regulating MST1 expression through downregulation of TGF-β to induce ROS formation. Therefore, our study shows that neferine can serve as an adjuvant therapy for NSCLC patients.
Collapse
Affiliation(s)
- Peng-Cheng Zhong
- Department of Integrated Traditional Chinese and Western Medicine, Xiangtan Central Hospital, Xiangtan, China
| | - Zhi-Wen Liu
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, Hong Kong, China
| | - Qi-Chang Xing
- Department of Pharmaceutical, Xiangtan Central Hospital, Xiangtan, China
| | - Jia Chen
- Department of Pharmaceutical, Xiangtan Central Hospital, Xiangtan, China
| | - Rui-Pei Yang
- Laboratory of Traditional Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
19
|
Arrè V, Scialpi R, Centonze M, Giannelli G, Scavo MP, Negro R. The 'speck'-tacular oversight of the NLRP3-pyroptosis pathway on gastrointestinal inflammatory diseases and tumorigenesis. J Biomed Sci 2023; 30:90. [PMID: 37891577 PMCID: PMC10612184 DOI: 10.1186/s12929-023-00983-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023] Open
Abstract
The NLRP3 inflammasome is an intracellular sensor and an essential component of the innate immune system involved in danger recognition. An important hallmark of inflammasome activation is the formation of a single supramolecular punctum, known as a speck, per cell, which is the site where the pro-inflammatory cytokines IL-1β and IL-18 are converted into their bioactive form. Speck also provides the platform for gasdermin D protein activation, whose N-terminus domain perforates the plasma membrane, allowing the release of mature cytokines alongside with a highly inflammatory form of cell death, namely pyroptosis. Although controlled NLRP3 inflammasome-pyroptosis pathway activation preserves mucosal immunity homeostasis and contributes to host defense, a prolonged trigger is deleterious and could lead, in genetically predisposed subjects, to the onset of inflammatory bowel disease, including Crohn's disease and ulcerative colitis, as well as to gastrointestinal cancer. Experimental evidence shows that the NLRP3 inflammasome has both protective and pathogenic abilities. In this review we highlight the impact of the NLRP3-pyroptosis axis on the pathophysiology of the gastrointestinal tract at molecular level, focusing on newly discovered features bearing pro- and anti-inflammatory and neoplastic activity, and on targeted therapies tested in preclinical and clinical trials.
Collapse
Affiliation(s)
- Valentina Arrè
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Rosanna Scialpi
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Matteo Centonze
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Maria Principia Scavo
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Roberto Negro
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy.
| |
Collapse
|
20
|
Barar E, Shi J. Genome, Metabolism, or Immunity: Which Is the Primary Decider of Pancreatic Cancer Fate through Non-Apoptotic Cell Death? Biomedicines 2023; 11:2792. [PMID: 37893166 PMCID: PMC10603981 DOI: 10.3390/biomedicines11102792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid tumor characterized by poor prognosis and resistance to treatment. Resistance to apoptosis, a cell death process, and anti-apoptotic mechanisms, are some of the hallmarks of cancer. Exploring non-apoptotic cell death mechanisms provides an opportunity to overcome apoptosis resistance in PDAC. Several recent studies evaluated ferroptosis, necroptosis, and pyroptosis as the non-apoptotic cell death processes in PDAC that play a crucial role in the prognosis and treatment of this disease. Ferroptosis, necroptosis, and pyroptosis play a crucial role in PDAC development via several signaling pathways, gene expression, and immunity regulation. This review summarizes the current understanding of how ferroptosis, necroptosis, and pyroptosis interact with signaling pathways, the genome, the immune system, the metabolism, and other factors in the prognosis and treatment of PDAC.
Collapse
Affiliation(s)
- Erfaneh Barar
- Liver and Pancreatobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - Jiaqi Shi
- Department of Pathology & Clinical Labs, Rogel Cancer Center, Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
21
|
Liu Y, Zhang X, Zhang P, He T, Zhang W, Ma D, Li P, Chen J. A high-throughput Gaussia luciferase reporter assay for screening potential gasdermin E activators against pancreatic cancer. Acta Pharm Sin B 2023; 13:4253-4272. [PMID: 37799380 PMCID: PMC10548051 DOI: 10.1016/j.apsb.2023.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/20/2023] [Accepted: 06/15/2023] [Indexed: 10/07/2023] Open
Abstract
It is discovered that activated caspase-3 tends to induce apoptosis in gasdermin E (GSDME)-deficient cells, but pyroptosis in GSDME-sufficient cells. The high GSDME expression and apoptosis resistance of pancreatic ductal adenocarcinoma (PDAC) cells shed light on another attractive strategy for PDAC treatment by promoting pyroptosis. Here we report a hGLuc-hGSDME-PCA system for high-throughput screening of potential GSDME activators against PDAC. This screening system neatly quantifies the oligomerization of GSDME-N to characterize whether pyroptosis occurs under the stimulation of chemotherapy drugs. Based on this system, ponatinib and perifosine are screened out from the FDA-approved anti-cancer drug library containing 106 compounds. Concretely, they exhibit the most potent luminescent activity and cause drastic pyroptosis in PDAC cells. Further, we demonstrate that perifosine suppresses pancreatic cancer by promoting pyroptosis via caspase-3/GSDME pathway both in vitro and in vivo. Collectively, this study reveals the great significance of hGLuc-hGSDME-PCA in identifying compounds triggering GSDME-dependent pyroptosis and developing promising therapeutic agents for PDAC.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaowei Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ping Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tingting He
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Weitao Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Dingyuan Ma
- Department of Prenatal Diagnosis, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing 210004, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
22
|
Huang ZL, Abdallah AS, Shen GX, Suarez M, Feng P, Yu YJ, Wang Y, Zheng SH, Hu YJ, Xiao X, Liu Y, Liu SR, Chen ZP, Li XN, Xia YF. Silencing GMPPB Inhibits the Proliferation and Invasion of GBM via Hippo/MMP3 Pathways. Int J Mol Sci 2023; 24:14707. [PMID: 37834154 PMCID: PMC10572784 DOI: 10.3390/ijms241914707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive malignancy and represents the most common brain tumor in adults. To better understand its biology for new and effective therapies, we examined the role of GDP-mannose pyrophosphorylase B (GMPPB), a key unit of the GDP-mannose pyrophosphorylase (GDP-MP) that catalyzes the formation of GDP-mannose. Impaired GMPPB function will reduce the amount of GDP-mannose available for O-mannosylation. Abnormal O-mannosylation of alpha dystroglycan (α-DG) has been reported to be involved in cancer metastasis and arenavirus entry. Here, we found that GMPPB is highly expressed in a panel of GBM cell lines and clinical samples and that expression of GMPPB is positively correlated with the WHO grade of gliomas. Additionally, expression of GMPPB was negatively correlated with the prognosis of GBM patients. We demonstrate that silencing GMPPB inhibits the proliferation, migration, and invasion of GBM cells both in vitro and in vivo and that overexpression of GMPPB exhibits the opposite effects. Consequently, targeting GMPPB in GBM cells results in impaired GBM tumor growth and invasion. Finally, we identify that the Hippo/MMP3 axis is essential for GMPPB-promoted GBM aggressiveness. These findings indicate that GMPPB represents a potential novel target for GBM treatment.
Collapse
Affiliation(s)
- Zi-Lu Huang
- State Key Laboratory of Oncology in Southern China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China or (Z.-L.H.); (P.F.); (Y.W.); (S.-H.Z.); (Y.-J.H.); (X.X.); (Y.L.)
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (A.S.A.); (M.S.)
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Aalaa Sanad Abdallah
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (A.S.A.); (M.S.)
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Guang-Xin Shen
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou 528031, China;
| | - Milagros Suarez
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (A.S.A.); (M.S.)
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ping Feng
- State Key Laboratory of Oncology in Southern China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China or (Z.-L.H.); (P.F.); (Y.W.); (S.-H.Z.); (Y.-J.H.); (X.X.); (Y.L.)
| | - Yan-Jiao Yu
- State Key Laboratory of Oncology in Southern China, Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (Y.-J.Y.); (Z.-P.C.)
| | - Ying Wang
- State Key Laboratory of Oncology in Southern China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China or (Z.-L.H.); (P.F.); (Y.W.); (S.-H.Z.); (Y.-J.H.); (X.X.); (Y.L.)
| | - Shuo-Han Zheng
- State Key Laboratory of Oncology in Southern China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China or (Z.-L.H.); (P.F.); (Y.W.); (S.-H.Z.); (Y.-J.H.); (X.X.); (Y.L.)
| | - Yu-Jun Hu
- State Key Laboratory of Oncology in Southern China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China or (Z.-L.H.); (P.F.); (Y.W.); (S.-H.Z.); (Y.-J.H.); (X.X.); (Y.L.)
| | - Xiang Xiao
- State Key Laboratory of Oncology in Southern China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China or (Z.-L.H.); (P.F.); (Y.W.); (S.-H.Z.); (Y.-J.H.); (X.X.); (Y.L.)
| | - Ya Liu
- State Key Laboratory of Oncology in Southern China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China or (Z.-L.H.); (P.F.); (Y.W.); (S.-H.Z.); (Y.-J.H.); (X.X.); (Y.L.)
| | - Song-Ran Liu
- State Key Laboratory of Oncology in Southern China, Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China;
| | - Zhong-Ping Chen
- State Key Laboratory of Oncology in Southern China, Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (Y.-J.Y.); (Z.-P.C.)
| | - Xiao-Nan Li
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (A.S.A.); (M.S.)
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yun-Fei Xia
- State Key Laboratory of Oncology in Southern China, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China or (Z.-L.H.); (P.F.); (Y.W.); (S.-H.Z.); (Y.-J.H.); (X.X.); (Y.L.)
| |
Collapse
|
23
|
Saadh MJ, Baher H, Li Y, Chaitanya M, Arias-Gonzáles JL, Allela OQB, Mahdi MH, Carlos Cotrina-Aliaga J, Lakshmaiya N, Ahjel S, Amin AH, Gilmer Rosales Rojas G, Ameen F, Ahsan M, Akhavan-Sigari R. The bioengineered and multifunctional nanoparticles in pancreatic cancer therapy: Bioresponisive nanostructures, phototherapy and targeted drug delivery. ENVIRONMENTAL RESEARCH 2023; 233:116490. [PMID: 37354932 DOI: 10.1016/j.envres.2023.116490] [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: 05/24/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
The multidisciplinary approaches in treatment of cancer appear to be essential in term of bringing benefits of several disciplines and their coordination in tumor elimination. Because of the biological and malignant features of cancer cells, they have ability of developing resistance to conventional therapies such as chemo- and radio-therapy. Pancreatic cancer (PC) is a malignant disease of gastrointestinal tract in which chemotherapy and radiotherapy are main tools in its treatment, and recently, nanocarriers have been emerged as promising structures in its therapy. The bioresponsive nanocarriers are able to respond to pH and redox, among others, in targeted delivery of cargo for specific treatment of PC. The loading drugs on the nanoparticles that can be synthetic or natural compounds, can help in more reduction in progression of PC through enhancing their intracellular accumulation in cancer cells. The encapsulation of genes in the nanoparticles can protect against degradation and promotes intracellular accumulation in tumor suppression. A new kind of therapy for cancer is phototherapy in which nanoparticles can stimulate both photothermal therapy and photodynamic therapy through hyperthermia and ROS overgeneration to trigger cell death in PC. Therefore, synergistic therapy of phototherapy with chemotherapy is performed in accelerating tumor suppression. One of the important functions of nanotechnology is selective targeting of PC cells in reducing side effects on normal cells. The nanostructures are capable of being surface functionalized with aptamers, proteins and antibodies to specifically target PC cells in suppressing their progression. Therefore, a specific therapy for PC is provided and future implications for diagnosis of PC is suggested.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | - Hala Baher
- Department of Radiology and Ultrasonography Techniques, College of Medical Techniques, Al-Farahidi University, Baghdad, Iraq
| | - Yuanji Li
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, 066004, China
| | - Mvnl Chaitanya
- Department of Pharmacognosy, School of Pharmacy, Lovely Professional University, Phagwara, Punjab, 144001, India
| | - José Luis Arias-Gonzáles
- Department of Social Sciences, Faculty of Social Studies, University of British Columbia, Vancouver, Canada
| | | | | | | | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Salam Ahjel
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | | | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Muhammad Ahsan
- Department of Measurememts and Control Systems, Silesian University of Technology, Gliwice, 44-100, Poland.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
24
|
Lin SC, Tsou SH, Kuo CY, Chen WL, Wu KW, Lin CL, Huang CN. Denosumab Attenuates Glucolipotoxicity-Induced β-Cell Dysfunction and Apoptosis by Attenuating RANK/RANKL Signals. Int J Mol Sci 2023; 24:10289. [PMID: 37373436 DOI: 10.3390/ijms241210289] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Obesity is strongly associated with insulin sensitivity in type 2 diabetes (T2D), mainly because free fatty acids (FFAs) are released from excess fat tissue. Long-term exposure to high levels of FFAs and glucose leads to glucolipotoxicity, causing damage to pancreatic β-cells, thus accelerating the progression of T2D. Therefore, the prevention of β-cell dysfunction and apoptosis is essential to prevent the development of T2D. Unfortunately, there are currently no specific clinical strategies for protecting β-cells, highlighting the need for effective therapies or preventive approaches to improve the survival of β-cells in T2D. Interestingly, recent studies have shown that the monoclonal antibody denosumab (DMB), used in osteoporosis, displays a positive effect on blood glucose regulation in patients with T2D. DMB acts as an osteoprotegerin (OPG) by inhibiting the receptor activator of the NF-κB ligand (RANKL), preventing the maturation and function of osteoclasts. However, the exact mechanism by which the RANK/RANKL signal affects glucose homeostasis has not been fully explained. The present study used human 1.4 × 107 β-cells to simulate the T2D metabolic condition of high glucose and free fatty acids (FFAs), and it investigated the ability of DMB to protect β-cells from glucolipotoxicity. Our results show that DMB effectively attenuated the cell dysfunction and apoptosis caused by high glucose and FFAs in β-cells. This may be caused by blocking the RANK/RANKL pathway that reduced mammalian sterile 20-like kinase 1 (MST1) activation and indirectly increased pancreatic and duodenal homeobox 1 (PDX-1) expression. Furthermore, the increase in inflammatory cytokines and ROS caused by the RANK/RANKL signal also played an important role in glucolipotoxicity-induced cytotoxicity, and DMB can also protect β-cells by reducing the mechanisms mentioned above. These findings provide detailed molecular mechanisms for the future development of DMB as a potential protective agent of β-cells.
Collapse
Affiliation(s)
- Sheng-Chieh Lin
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Orthopaedics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Sing-Hua Tsou
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Chien-Yin Kuo
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Wei-Liang Chen
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Kuan-Wen Wu
- Department of Orthopaedic Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chih-Li Lin
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Chien-Ning Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| |
Collapse
|
25
|
Huang Q, Peng X, Li Q, Zhu J, Xue J, Jiang H. Construction and comprehensive analysis of a novel prognostic signature associated with pyroptosis molecular subtypes in patients with pancreatic adenocarcinoma. Front Immunol 2023; 14:1111494. [PMID: 36817451 PMCID: PMC9935619 DOI: 10.3389/fimmu.2023.1111494] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Background Treatment of cancer with pyroptosis is an emerging strategy. Molecular subtypes based on pyroptosis-related genes(PRGs) seem to be considered more conducive to individualized therapy. It is meaningful to construct a pyroptosis molecular subtypes-related prognostic signature (PMSRPS) to predict the overall survival (OS) of patients with pancreatic adenocarcinoma(PAAD) and guide treatment. Methods Based on the transcriptome data of 23 PRGs, consensus clustering was applied to divide the TCGA and GSE102238 combined cohort into three PRGclusters. Prognosis-related differentially expressed genes(DEGs) among PRGclusters were subjected to LASSO Cox regression analysis to determine a PMSRPS. External cohort and in vitro experiments were conducted to verify this PMSRPS. The CIBERSORT algorithm, the ESTIMATE algorithm and the Immunophenoscore (IPS) were used to analyze the infiltrating abundance of immune cells, the tumor microenvironment (TME), and the response to immunotherapy, respectively. Wilcoxon analysis was used to compare tumor mutational burden (TMB) and RNA stemness scores (RNAss) between groups. RT-qPCR and in vitro functional experiments were used for evaluating the expression and function of SFTA2. Results Based on three PRGclusters, 828 DEGs were obtained and a PMSRPS was subsequently constructed. In internal and external validation, patients in the high-risk group had significantly lower OS than those in the low-risk group and PMSRPS was confirmed to be an independent prognostic risk factor for patients with PAAD with good predictive performance. Immune cell infiltration abundance and TME scores indicate patients in the high-risk group have typical immunosuppressive microenvironment characteristics. Analysis of IPS suggests patients in the high-risk group responded better to novel immune checkpoint inhibitors (ICIs) than PD1/CTLA4. The high-risk group had higher TMB and RNAss. In addition, 10 potential small-molecule compounds were screened out. Finally, we found that the mRNA expression of SFTA2 gene with the highest risk coefficient in PMSRPS was significantly higher in PAAD than in paracancerous tissues, and knockdown of it significantly delayed the progression of PAAD. Conclusions PMSRPS can well predict the prognosis, TME and immunotherapy response of patients with PAAD, identify potential drugs, and provide treatment guidance based on individual needs.
Collapse
Affiliation(s)
- Qian Huang
- Department of General Practice, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China,Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xingyu Peng
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingqing Li
- Department of General Practice, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China,Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinfeng Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ju Xue
- Department of Pathology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Hua Jiang
- Department of General Practice, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China,Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China,*Correspondence: Hua Jiang,
| |
Collapse
|
26
|
Zhao L, You Z, Bai Z, Xie J. Machine learning-based construction of a ferroptosis and necroptosis associated lncRNA signature for predicting prognosis and immunotherapy response in hepatocellular cancer. Front Oncol 2023; 13:1171878. [PMID: 37152064 PMCID: PMC10157233 DOI: 10.3389/fonc.2023.1171878] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/06/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Liver hepatocellular carcinoma (LIHC), one of the most common malignancies worldwide, occurs with high incidence and mortality. Ferroptosis and necroptosis are critically associated with LIHC prognosis. Some long non-coding RNAs (lncRNAs) have been found to induce ferroptosis and necroptosis in hepatocellular carcinoma cells. Methods Cox regression analysis was used to construct a risk model for LIHC based on differentially expressed ferroptosis and necroptosis related lncRNAs (F-NLRs), and their expression in SMMC7721, HepG2 and WRL68 cells was detected by qPCR. Results Five F-NLRs were associated with LIHC prognosis, including KDM4A-AS1, ZFPM2-AS1, AC099850.3, MKLN1-AS, and BACE1-AS. Kaplan-Meier survival analysis indicated that patients with LIHC in the high-risk group were associated with poor prognosis. The combined F-NLR signature model demonstrated a prognostic AUC value of 0.789 and was more accurate than standard clinical variables for predicting LIHC prognosis. T cell functions and immunotherapy responses differed significantly between patients in the low- and high-risk groups. Additionally, immune checkpoints and m6A-related genes were differentially expressed between patients in the two risk groups. Furthermore, proteins encoded by the five F-NLRs were overexpressed in four liver cancer cell lines compared to that in human liver cell line WRL68. Pan-cancer examination revealed that expression levels of the five F-NLRs differed between most common tumor types and normal tissues. Conclusion F-NLRs identified in this study provide a predictive signature representing ferroptosis and necroptosis in LIHC, which correlated well with patient prognosis, clinicopathological characteristics, and immunotherapy responses. The study findings help to elucidate the mechanisms of F-NLRs in LIHC and provide further guidance for the selection and development of immunotherapeutic agents for LIHC.
Collapse
Affiliation(s)
- Lei Zhao
- The Third Clinical lnstitute, Guangzhou Medical University, Guangzhou, China
| | - Zhixuan You
- The Third Clinical lnstitute, Guangzhou Medical University, Guangzhou, China
| | - Zhixun Bai
- Department of Nephrology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Zhixun Bai, ; Jian Xie,
| | - Jian Xie
- Department of Medical Genetics, Zunyi Medical University, Zunyi, China
- *Correspondence: Zhixun Bai, ; Jian Xie,
| |
Collapse
|
27
|
Ding M, Liu J, Lv H, Zhu Y, Chen Y, Peng H, Fan S, Chen X. Knocking down GALNT6 promotes pyroptosis of pancreatic ductal adenocarcinoma cells through NF-κB/NLRP3/GSDMD and GSDME signaling pathway. Front Oncol 2023; 13:1097772. [PMID: 36925932 PMCID: PMC10013470 DOI: 10.3389/fonc.2023.1097772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC), the most prevalent type of pancreatic cancer, is a highly lethal malignancy with poor prognosis. Polypeptide N-acetylgalactosaminyltransferase-6 (GALNT6) is frequently overexpressed in PDAC. However, the role of GALNT6 in the PDAC remains unclear. Methods The expression of GALNT6 in pancreatic cancer and normal tissues were analyzed by bioinformatic analyses and immunohistochemistry. CCK8 and colony formation were used to detect cell proliferation. Flow cytometry was applied to detect cell cycle.The pyroptosis was detected by scanning electron microscopy. The mRNA expression was detected by qRT-PCR. The protein expression and localization were detected by western blot and immunofluorescence assay. ELISA was used to detect the levels of inflammatory factors. Results The expression of GALNT6 was associated with advanced tumor stage, and had an area under curve (AUC) value of 0.919 in pancreatic cancer based on the cancer genome atlas (TCGA) dataset. Knockdown of GALNT6 inhibited cell proliferation, migration, invasion and cell cycle arrest of PDAC cells. Meanwhile, knockdown of GALNT6 increased the expression levels of interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and interleukin-18 (IL-18), the release of inflammasome and an increasing of Gasdermin D (GSDMD), N-terminal of GSDMD (GSDMD-N), Gasdermin E (GSDME) and N-terminal of GSDME (GSDME-N) in PDAC cells. GALNT6 suppressed the expression of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) and GSDMD by glycosylation of NF-κB and inhibiting the nucleus localization of NF-κB. Additionally, GALNT6 promotes the degradation of GSDME by O-glycosylation. Conclusion We found that GALNT6 is highly expressed in pancreatic cancer and plays a carcinogenic role. The results suggested that GALNT6 regulates the pyroptosis of PDAC cells through NF-κB/NLRP3/GSDMD and GSDME signaling. Our study might provides novel insights into the roles of GALNT6 in PDAC progression.
Collapse
Affiliation(s)
- Mengyang Ding
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Glycobiological Engineering, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingyu Liu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Glycobiological Engineering, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Honghui Lv
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Glycobiological Engineering, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yanlin Zhu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Glycobiological Engineering, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yumiao Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Glycobiological Engineering, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui Peng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Glycobiological Engineering, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sairong Fan
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Glycobiological Engineering, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaoming Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Glycobiological Engineering, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
28
|
Prediction of Survival and Tumor Microenvironment Infiltration Based on Pyroptosis-Related lncRNAs in Pancreatic Cancer. DISEASE MARKERS 2022; 2022:5634887. [PMID: 36618967 PMCID: PMC9822759 DOI: 10.1155/2022/5634887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/16/2022] [Accepted: 12/07/2022] [Indexed: 01/01/2023]
Abstract
Pancreatic cancer (PC) is a fatal tumor with high mortality. Pyroptosis plays a tumor suppressor role as a novel cell death. However, the influences of the pyroptosis-related lncRNAs (PRlncRNAs) on the prognosis and tumor microenvironment (TME) infiltration have not been fully studied in PC. Using coexpression analysis and univariate Cox regression analysis, we identified seventeen prognostic PRlncRNAs from The Cancer Genome Atlas (TCGA) dataset, which were all expressed differently in normal and tumor samples. A seven-PRlncRNA risk signature was constructed and validated using the least absolute shrinkage and selection operator (LASSO) regression. Furthermore, we verified its independence and created a nomogram to validate the clinical viability of the risk signature. We then identified its relationship with clinical factors and evaluated its values in TME infiltration, functional enrichment, tumor mutation, and therapeutic responses in PC. Lower ImmuneScore, ESTIMATEScore, and advanced tumor stage were connected with high-risk score. The low-risk group was characterized by better OS, elevated immune activation, and higher susceptibility of pazopanib and sunitinib. The high-risk group possessed a worse immune infiltration and poor survival, with higher tumor mutations and lapatinib and paclitaxel that may be better choices in this group. In conclusion, we developed an original seven-PRlncRNA risk signature to predict prognosis, TME infiltration, tumor mutation, and therapeutic options for PC patients.
Collapse
|
29
|
Ding Y, Ye B, Sun Z, Mao Z, Wang W. Reactive Oxygen Species‐Mediated Pyroptosis with the Help of Nanotechnology: Prospects for Cancer Therapy. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province Hangzhou Zhejiang 310009 China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease Zhejiang University Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province Hangzhou Zhejiang 310009 China
| | - Binglin Ye
- Department of Hepatobiliary and Pancreatic Surgery The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province Hangzhou Zhejiang 310009 China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease Zhejiang University Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province Hangzhou Zhejiang 310009 China
| | - Zhongquan Sun
- Department of Hepatobiliary and Pancreatic Surgery The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province Hangzhou Zhejiang 310009 China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease Zhejiang University Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province Hangzhou Zhejiang 310009 China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou Zhejiang 310009 China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province Hangzhou Zhejiang 310009 China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease Zhejiang University Hangzhou Zhejiang 310009 China
- The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province Hangzhou Zhejiang 310009 China
| |
Collapse
|
30
|
Sarwar A, Zhu M, Su Q, Zhu Z, Yang T, Chen Y, Peng X, Zhang Y. Targeting mitochondrial dysfunctions in pancreatic cancer evokes new therapeutic opportunities. Crit Rev Oncol Hematol 2022; 180:103858. [DOI: 10.1016/j.critrevonc.2022.103858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/07/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
|
31
|
Liu B, Dong C, Chen Q, Fan Z, Zhang Y, Wu Y, Cui T, Liu F. Circ_0007534 as new emerging target in cancer: Biological functions and molecular interactions. Front Oncol 2022; 12:1031802. [PMID: 36505874 PMCID: PMC9730518 DOI: 10.3389/fonc.2022.1031802] [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: 08/30/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Circular RNA (circRNAs), an important member of the non-coding RNA (ncRNA) family, are widely expressed in a variety of biological cells. Owing to their stable structures, sequence conservations, and cell- or tissue-specific expressions, these RNA have become a popular subject of scientific research. With the development of sequencing methods, it has been revealed that circRNAs exert their biological function by sponging microRNAs (miRNAs), regulating transcription, or binding to proteins. Humans have historically been significantly impacted by various types of cancer. Studies have shown that circRNAs are abnormally expressed in various cancers and are involved in the occurrence and development of malignant tumors, such as tumor cell proliferation, migration, and invasion. As one of its star molecules, circ_0007534 is upregulated in colorectal, cervical, and pancreatic cancers; is closely related to the occurrence, development, and prognosis of tumors; and is expected to become a novel tumor marker and therapeutic target. This article briefly reviews the expression and mechanism of circ_0007534 in malignant tumors based on the domestic and foreign literature.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fuquan Liu
- Department of Interventional Therapy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
A novel refined pyroptosis and inflammasome-related genes signature for predicting prognosis and immune microenvironment in pancreatic ductal adenocarcinoma. Sci Rep 2022; 12:18384. [PMID: 36319832 PMCID: PMC9626462 DOI: 10.1038/s41598-022-22864-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/20/2022] [Indexed: 01/01/2023] Open
Abstract
Pyroptosis is an inflammatory form of cell death, which plays a key role in the development of auto-inflammation and cancer. This study aimed to construct a pyroptosis and inflammasome-related genes for predicting prognosis of the pancreatic ductal adenocarcinoma (PDAC). This study was based primarily on the one-way analysis of variance, univariate Cox regression analysis, Least absolute shrinkage and selection operator (LASSO) Cox regression, a risk-prognostic signature, gene set variation analysis (GSVA), and immune microenvironment analysis, using PDAC data from The Cancer Genome Atlas and International Cancer Genome Consortium databases for the analysis of the role of 676 pyroptosis and inflammasome-related genes in PDAC retrieved from the Reactome and GeneCards databases. Lastly, we collected six paired PDAC and matched normal adjacent tissue samples to verify the expression of signature genes by quantitative real-time PCR (qRT-PCR). We identified 18 candidate pyroptosis and inflammasome-related genes that differed significantly between pathologic grades (stages) of PDAC patients. The univariate Cox and LASSO analyses pointed to six genes as the best variables for constructing a prognostic signature, including ACTA2, C1QTNF9, DNAH8, GATM, LBP, and NGF. The results of the risk prognostic model indicated that the AUCs at 1, 3, and 5 years were greater than 0.62. GSVA revealed that 'GLYCOLYSIS', 'P53 PATHWAY', 'KRAS SIGNALING UP', and 'INFLAMMATORY RESPONSE' hallmark gene sets were associated with the risk score. The high-risk group was associated with poor prognosis and was characterized by a lower infiltration of cells involved in anti-tumor immunity; whereas the low-risk group with higher T cells, NK cells, and macrophages showed relatively better survival and significantly higher upregulation of cytolytic scores and inflammation scores. Additionally, crucial pyroptosis and inflammasome-related genes were further validated by qRT-PCR. Our study revealed the prognostic role of the pyroptosis and inflammasome-related genes in PDAC for the first time. Simultaneously, the biological and prognostic heterogeneity of PDAC had been demonstrated, deepening our molecular understanding of this tumor.
Collapse
|
33
|
Pyroptosis-Related Gene Model Predicts Prognosis and Immune Microenvironment for Non-Small-Cell Lung Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1749111. [PMID: 36092153 PMCID: PMC9453043 DOI: 10.1155/2022/1749111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/12/2022] [Accepted: 08/09/2022] [Indexed: 12/13/2022]
Abstract
Non-small-cell lung cancer (NSCLC) has a high incidence and mortality worldwide. Moreover, it needs more accurate means for predicting prognosis and treatments. Pyroptosis is a novel form of cell death about inflammation which was highly related to the occurrence and development of tumors. Despite having some studies about pyroptosis-related genes (PRGs) and cancer, the correlation has not been explored enough between PRGs and immune in NSCLC. In this study, we constructed a PRG model by WGCNA to access the prognosis value PRGs have. The testing cohort (n = 464) with four datasets from the GEO database conducted a survival analysis to confirm the stability of the prognostic model. The risk score and age are examined as independent prognostic factors. Based on the PRGs, we found multiple pathways enriched in immune in NSCLC. Separating samples into three subtypes by consensus cluster analysis, Cluster 3 was identified as immune-inflamed phenotype with an optimistic prognostic outcome. A three-gene PRG signature (BNIP3, CASP9, and CAPN1) was identified, and BNIP3 was identified as the core gene. Knockdown of BNIP3 significantly inhibited the growth of H358 cells and induced pyroptosis. In conclusion, the model construction based on PRGs provides novel insights into the prediction of NSCLC prognosis, and BNIP3 can serve as a diagnostic biomarker for NSCLC.
Collapse
|
34
|
Laminarin Attenuates ROS-Mediated Cell Migration and Invasiveness through Mitochondrial Dysfunction in Pancreatic Cancer Cells. Antioxidants (Basel) 2022; 11:antiox11091714. [PMID: 36139787 PMCID: PMC9495390 DOI: 10.3390/antiox11091714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 12/26/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a notoriously aggressive type of cancer with a high metastasis rate. It is conventionally treated by surgical resection and neoadjuvant chemotherapy. However, continuous chemotherapy leads to relapse in most PDAC patients due to chemical resistance. Therefore, novel anticancer agents need to be identified and developed. The antitumor activities of laminarin extracted from brown algae against hepatocarcinoma, lung, and colon cancer have been established. However, its effects on pancreatic cancer have remained obscure. Purpose: Our study identified the anticancer effects of laminarin on pancreatic cancer cells and tried to explain its intracellular mechanisms. Methods: We assessed the cell viability of PANC-1 and MIA PaCa-2 cells using MTT assay. Hanging drop method was used for the spheroid formation. Flow cytometry was conducted to evaluate the several intracellular alterations including apoptosis, ROS production, mitochondrial membrane potential (MMP), and calcium concentration induced by laminarin. An invasion test was performed to assess the inhibitory effect of laminarin on cell migration and the invasive genes were evaluated by RT-qPCR. Signaling pathway related with anticancer effects of laminarin was analyzed by western blot. Results: We report that inhibiting laminarin increased the proliferation and viability of the representative pancreatic cancer cell lines, MIA PaCa-2 and PANC-1. Laminarin triggered apoptosis and mitochondrial impairment as evidenced by depolarized mitochondrial membranes, disrupted calcium, and suppressed cell migration caused by reactive oxygen species production and related intracellular signaling pathways. Moreover, laminarin showed synergistic effects when combined with 5-FU, a standard anticancer agent for PDAC. Conclusion: The present study is the first to report that laminarin exerts anticancer effect through ROS production in pancreatic cancer cells. Laminarin shows potential to serve as a new anticancer agent for treating PDAC.
Collapse
|
35
|
Abstract
Background Pyroptosis has been attracting much attention recently. We have briefly compared its differences and similarities with other programmed deaths and the process of its study. With further exploration of the caspase family, including caspase-1/3/4/5/8/11, new insights into the molecular pathways of action of pyroptosis have been gained. It is also closely related to the development of many cancers, which at the same time provides us with new ideas for the treatment of cancer. Scope of Review We describe what is known regarding the impact of pyroptosis on anticancer immunity and give insight into the potential of harnessing pyroptosis as a tool and applying it to novel or existing anticancer strategies. Major Conclusions Pyroptosis, a caspase-dependent cell death, causes pore formation, cell swelling, rupture of the plasma membrane, and release of all intracellular contents. The role of pyroptosis in cancer is an extremely complex issue. There is growing evidence that tumor pyroptosis has anti-tumor and pro-tumor roles. It should be discussed in different cancer periods according to the characteristics of cancer occurrence and development. In cancer treatment, pyroptosis provides us with some potential new targets. For the existing drugs, the study of pyroptosis also helps us make better use of existing drugs for anticancer treatment. Immunotherapy is a hot research direction in the field of cancer treatment.
Collapse
Affiliation(s)
- Chen Huang
- Department of Abdominal Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, China
| | - Jian Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Chenliang Zhang
- Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
36
|
Tao S, Tian L, Wang X, Shou Y. A pyroptosis-related gene signature for prognosis and immune microenvironment of pancreatic cancer. Front Genet 2022; 13:817919. [PMID: 36118860 PMCID: PMC9476319 DOI: 10.3389/fgene.2022.817919] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer is one of the most lethal tumors owing to its unspecific symptoms during the early stage and multiple treatment resistances. Pyroptosis, a newly discovered gasdermin-mediated cell death, facilitates anti- or pro-tumor effects in a variety of cancers, whereas the impact of pyroptosis in pancreatic cancer remains unclear. Therefore, we downloaded RNA expression and clinic data from the TCGA-PAAD cohort and were surprised to find that most pyroptosis-related genes (PRGs) are not only overexpressed in tumor tissue but also strongly associated with overall survival. For their remarkable prognostic value, cox regression analysis and lasso regression were used to establish a five-gene signature. All patients were divided into low- and high-risk groups based on the media value of the risk score, and we discovered that low-risk patients had better outcomes in both the testing and validation cohorts using time receiver operating characteristic (ROC), nomograms, survival, and decision analysis. More importantly, a higher somatic mutation burden and less immune cell infiltration were found in the high-risk group. Following that, we predicted tumor response to chemotherapy and immunotherapy in both low- and high-risk groups, which suggests patients with low risk were more likely to respond to both immunotherapy and chemotherapy. To summarize, our study established an effective model that can help clinicians better predict patients’ drug responses and outcomes, and we also present basic evidence for future pyroptosis related studies in pancreatic cancer.
Collapse
Affiliation(s)
- Sifan Tao
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Tian
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Xiaoyan Wang, ; Yajun Shou,
| | - Yajun Shou
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan, China
- *Correspondence: Xiaoyan Wang, ; Yajun Shou,
| |
Collapse
|
37
|
Emerging mechanisms of pyroptosis and its therapeutic strategy in cancer. Cell Death Dis 2022; 8:338. [PMID: 35896522 PMCID: PMC9329358 DOI: 10.1038/s41420-022-01101-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 12/20/2022]
Abstract
Pyroptosis, a type of inflammatory programmed cell death, is triggered by caspase cleavage of gasdermin family proteins. Based on accumulating evidence, pyroptosis is closely associated with tumour development, but the molecular mechanism underlying pyroptosis activation and the signalling pathways regulated by pyroptosis remain unclear. In this review, we first briefly introduce the definition, morphological characteristics, and activation pathways of pyroptosis and the effect of pyroptosis on anticancer immunity. Then we review recent progress concerning the complex role of pyroptosis in various tumours. Importantly, we summarise various FDA-approved chemotherapy drugs or natural compounds that exerted antitumor properties by inducing pyroptosis of cancer cells. Moreover, we also focus on the current application of nanotechnology-induced pyroptosis in tumour therapy. In addition, some unsolved problems and potential future research directions are also raised.
Collapse
|
38
|
A Predictive Model Based on Pyroptosis-Related Gene Features Can Effectively Predict Clear Cell Renal Cell Carcinoma Prognosis and May Be an Underlying Target for Immunotherapy. DISEASE MARKERS 2022; 2022:6402599. [PMID: 35845137 PMCID: PMC9286942 DOI: 10.1155/2022/6402599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/06/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022]
Abstract
Methods The clinical information and RNA-seq data of ccRCC patients were collected from the TCGA dataset to first explore differential pyroptosis-related genes (PRGs). Univariate Cox regression and consensus clustering were applied to identify ccRCC subtypes. The prognostic PRGs were subjected to LASSO regression analysis to establish a prognostic model and to investigate its value and function. Finally, the relationship of the model immunity checkpoints and immunity infiltration was assessed. Results The receiver operating characteristic (ROC) showed that the 1-year, 3-year, and 5-year prediction rates of the prognostic model were 0.715, 0.693, and 0.732, respectively. The high-risk group had lower overall survival and higher stage than the low-risk group. Functional enrichment analysis showed that PRGs were significantly enriched mainly in the PPAR pathway, inflammatory pathway, and immune activity. ccRCC patient prognosis correlates with immune components in the microenvironment, and immune checkpoint molecules are significantly expressed in the high-risk group. Immunotherapy may be effective in the high-risk group. Conclusion Pyroptosis-related gene has an important impact on the progression of ccRCC and can be used as an independent predictor of patient prognosis. In addition, immune checkpoint molecules are significantly upregulated in high-risk populations, which may be a potential target for immunotherapy.
Collapse
|
39
|
Four-Pyroptosis Gene-Based Nomogram as a Novel Strategy for Predicting the Effect of Immunotherapy in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2680110. [PMID: 35782053 PMCID: PMC9242783 DOI: 10.1155/2022/2680110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/28/2022] [Indexed: 11/20/2022]
Abstract
Background Immunotherapy has been considered as a promising cancer treatment for hepatocellular carcinoma (HCC). However, due to the particular immune environment of the liver, identifying patients who could benefit from immunotherapy is critical in clinical practice. Methods The pyroptosis gene expression database of 54 candidates from The Cancer Genome Atlas (TCGA) were collected to discover the critical prognostic-related pyroptosis genes. A novel pyroptosis gene model was established to calculate the risk score. Kaplan–Meier analysis and receiver operating characteristic curve (ROC) were used to verify its predictive ability. The International Cancer Genome Consortium (ICGC) data was collected as external validation data to verify the model's accuracy. We employed multiple bioinformatics tools and algorithms to evaluate the tumor immune microenvironment (TIME) and the response to immunotherapy. Results Our study found that most pyroptosis genes were expressed differently in normal and tumor tissues and that their expression was associated with the prognosis. Then, a precise four-pyroptosis gene model was generated. The one-year area under the curves (AUCs) among the training, internal, and external validation patients were 0.901, 0.727, and 0.671, respectively. An analysis of survival data revealed that individuals had a worse prognosis than patients with low risk. The analysis of TIME revealed that the low-risk group had more antitumor cells, fewer immunosuppressive cells, stronger immune function, less immune checkpoint gene expression, and better immunotherapy response than the high-risk group. Immunophenoscore (IPS) analysis also demonstrated that the low-risk score was related to superior immune checkpoint inhibitors therapy. Conclusion A nomogram based on the four-pyroptosis gene signature was a novel tool to predict the effectiveness of immunotherapy for HCC. Therefore, individualized treatment targeting the pyroptosis genes may influence TIME and play an essential role in improving the prognosis in HCC patients.
Collapse
|
40
|
Gao W, Wang X, Zhou Y, Wang X, Yu Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct Target Ther 2022; 7:196. [PMID: 35725836 PMCID: PMC9208265 DOI: 10.1038/s41392-022-01046-3] [Citation(s) in RCA: 362] [Impact Index Per Article: 181.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
In recent years, immunotherapy represented by immune checkpoint inhibitors (ICIs) has led to unprecedented breakthroughs in cancer treatment. However, the fact that many tumors respond poorly or even not to ICIs, partly caused by the absence of tumor-infiltrating lymphocytes (TILs), significantly limits the application of ICIs. Converting these immune “cold” tumors into “hot” tumors that may respond to ICIs is an unsolved question in cancer immunotherapy. Since it is a general characteristic of cancers to resist apoptosis, induction of non-apoptotic regulated cell death (RCD) is emerging as a new cancer treatment strategy. Recently, several studies have revealed the interaction between non-apoptotic RCD and antitumor immunity. Specifically, autophagy, ferroptosis, pyroptosis, and necroptosis exhibit synergistic antitumor immune responses while possibly exerting inhibitory effects on antitumor immune responses. Thus, targeted therapies (inducers or inhibitors) against autophagy, ferroptosis, pyroptosis, and necroptosis in combination with immunotherapy may exert potent antitumor activity, even in tumors resistant to ICIs. This review summarizes the multilevel relationship between antitumor immunity and non-apoptotic RCD, including autophagy, ferroptosis, pyroptosis, and necroptosis, and the potential targeting application of non-apoptotic RCD to improve the efficacy of immunotherapy in malignancy.
Collapse
Affiliation(s)
- Weitong Gao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, changsha, 410008, China
| | - Yang Zhou
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xueqian Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yan Yu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
41
|
Wu ZH, Wu B, Li C, Zhang YJ, Zhou T. Pyroptosis-Related Signature and Tumor Microenvironment Infiltration Characterization in Head and Neck Squamous Cell Carcinoma. Front Cell Dev Biol 2022; 10:702224. [PMID: 35712671 PMCID: PMC9194563 DOI: 10.3389/fcell.2022.702224] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 04/25/2022] [Indexed: 11/26/2022] Open
Abstract
Background: Head and neck squamous cell carcinoma (HNSCC) is the sixth most widespread and deadly cancer. Until now, very few studies have systematically evaluated the role of pyroptosis-related genes (PRGs) and lncRNAs in HNSCC patients. Methods: We integrated the genomic data to comprehensively assess the role of pyroptosis with the tumor microenvironment cell-infiltrating characteristics in HNSCC. In addition, we also constructed a set of the scoring system to calculate the pyroptosis dysfunction in each patient. Results: The analysis of the CNV alteration frequency displayed that CNV changes were common in 33 PRGs, and the frequency of copy number gain and loss was similar. CASP8 demonstrated the highest mutation frequency. Considering the individual heterogeneity, a scoring system to quantify the pyroptosis pattern in each patient was constructed based on these phenotypic-related genes, which we named as the PyroptosisScore. The results indicated that the low PyroptosisScore group experienced increased extensive TMB than the high group, with the most significant mutated genes being TP53 and TTN. Finally, we tried to find some useful pyroptosis-related lncRNAs, and 14 differentially expressed lncRNAs were selected as independent prognosis factors of HNSCC patients based on the multivariate Cox analysis. Conclusion: This work suggests the pyroptosis features and the potential mechanisms of the tumor microenvironment. The exploration may assist in identifying novel biomarkers and help patients predict prognosis, clinical diagnosis, and management.
Collapse
Affiliation(s)
- Zeng-Hong Wu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bian Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Li
- Department of Otolaryngology Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - You-Jing Zhang
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Tao Zhou,
| |
Collapse
|
42
|
Li L, Deng Z, Xiao Z, Zou W, Liu R. Analysis of Pyroptosis-Related Signature for Predicting Prognosis and Tumor Immune Microenvironment in Pancreatic Cancer. Front Oncol 2022; 12:770005. [PMID: 35712482 PMCID: PMC9192978 DOI: 10.3389/fonc.2022.770005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 05/03/2022] [Indexed: 12/02/2022] Open
Abstract
Pancreatic cancer (PC) has a poor prognosis, which is attributable to its high aggressiveness and lack of effective therapies. Although immunotherapy has been used for the treatment of various tumor, its efficacy in pancreatic cancer is not satisfactory. As a caspase-1-dependent programmed cell death, pyroptosis s involved in the pathological process of many tumors. Nevertheless, the vital role of the pyroptosis-related gene (PRG) in PC remains unknown. In this study, univariate COX regression was performed for 33 pyroptosis-related genes. Based on these prognosis-related PRGs, all PC patients in the Cancer Genome Atlas (TCGA) database were divided into four subtypes. Then, pyroptosis score (PP-score) was established to quantify pyroptosis level for individual PC patients using principal component analysis (PCA) algorithms. Assessment of pyroptosis level within individual PC patients may predict tumor classification and patient prognosis. Finally, a signature was constructed in TCGA and verified in ICGC. In addition, immunocheckpoint analysis revealed the possibility that the low-risk group would benefit more from immunocheckpoint therapy. Taken together, pyroptosis-related genes play a significant role in tumor immunotherapy and can be utilized to predict the prognosis of PC patients.
Collapse
Affiliation(s)
- Lincheng Li
- Medical School of Chinese PLA, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhaoda Deng
- Medical School of Chinese PLA, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhaohui Xiao
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wenbo Zou
- Medical School of Chinese PLA, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Rong Liu
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Rong Liu,
| |
Collapse
|
43
|
Xie W, Li X, Yang C, Li J, Shen G, Chen H, Xiao SY, Li Y. The Pyroptosis-Related Gene Prognostic Index Associated with Tumor Immune Infiltration for Pancreatic Cancer. Int J Mol Sci 2022; 23:6178. [PMID: 35682857 PMCID: PMC9180955 DOI: 10.3390/ijms23116178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 11/28/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most fatal malignancies. Pyroptosis, a type of inflammatory cell death, likely plays a critical role in the development and progression of tumors. However, the relationship between pyroptosis-related genes (PRGs) and prognosis and immunity to PC is not entirely clear. This study, aimed at identifying the key PRGs in PC, highlights their prognostic value, immune characteristics, and candidate drugs for therapies. We screened 47 differentially expressed PRGs between PC and normal pancreas tissues from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) datasets. Afterwards, a pyroptosis-related gene prognostic index (PRGPI) was constructed based on eight PRGs (AIM2, GBP1, HMGB1, IL18, IRF6, NEK7, NLRP1 and PLCG1) selected by univariate and multivariate Cox regression analysis and LASSO regression analysis, and verified in two external datasets from the International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO) databases. We found that the PC patients in the PRGPI-defined subgroups not only reflected significantly different levels of infiltration in a variety of immune cells, such as M1 macrophages, but also showed differential expression in genes of the human leukocyte antigen (HLA) family and immune checkpoints. Additionally, molecular characteristics and drug sensitivity also stayed close to the PRGPI risk scores. Therefore, PRGPI may serve as a valuable prognostic biomarker and may potentially provide guidance toward novel therapeutic options for PC patients.
Collapse
Affiliation(s)
- Wen Xie
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Xiaoyi Li
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Chunxiu Yang
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Jiahao Li
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Guoyan Shen
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Hongshan Chen
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| | - Shu-Yuan Xiao
- Department of Pathology, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Yueying Li
- Department of Pathology, Wuhan University Zhongnan Hospital, Wuhan 430000, China; (W.X.); (X.L.); (C.Y.); (J.L.); (G.S.); (H.C.)
- Wuhan University Center for Pathology and Molecular Diagnostics, Wuhan 430000, China
| |
Collapse
|
44
|
Su K, Peng Y, Yu H. Development of a Prognostic Model Based on Pyroptosis-Related Genes in Pancreatic Adenocarcinoma. DISEASE MARKERS 2022; 2022:9141117. [PMID: 35677632 PMCID: PMC9169203 DOI: 10.1155/2022/9141117] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022]
Abstract
Background The importance of pyroptosis in tumorigenesis and cancer progression is becoming increasingly apparent. However, the efficacy of using pyroptosis-related genes (PRGs) in predicting the prognosis of pancreatic adenocarcinoma (PAAD) patients is unknown. Methods This investigation used two databases to obtain expression data for PAAD patients. Differentially expressed PRGs (DEPRGs) were identified between PAAD and control samples. Several bioinformatic approaches were used to analyze the biological functions of DEPRGs and to identify prognostic DERPGs. A miRNA-prognostic DEPRG-transcription factor (TF) regulatory network was created via the miRNet online tool. A risk score model was created after each patient's risk score was calculated. The microenvironments of the low- and high-risk groups were assessed using xCell, the expression of immune checkpoints was determined, and gene set variation analysis (GSVA) was performed. Finally, the efficacy of certain potential drugs was predicted using the pRRophetic algorithm, and the results in the high- and low-risk groups were compared. Results A total of 13 DEPRGs were identified between PAAD and control samples. Functional enrichment analysis showed that the DEPRGs had a close relationship with inflammation. In univariate and multivariate Cox regression analyses, GSDMC, IRF1, and PLCG1 were identified as prognostic biomarkers in PAAD. The results of the miRNA-prognostic DEPRG-TF regulatory network showed that GSDMC, IRF1, and PLCG1 were regulated by both specific and common miRNAs and TFs. Based on the risk score and other independent prognostic indicators, a nomogram with a good ability to predict the survival of PAAD patients was developed. By evaluating the tumor microenvironment, we observed that the immune and metabolic microenvironments of the two groups were substantially different. In addition, individuals in the low-risk group were more susceptible to axitinib and camptothecin, whereas lapatinib might be preferred for patients in the high-risk group. Conclusion Our study revealed the prognostic value of PRGs in PAAD and created a reliable model for predicting the prognosis of PAAD patients. Our findings will benefit the prognostication and treatment of PAAD patients.
Collapse
Affiliation(s)
- Kaifeng Su
- Medical Faculty of Ludwig-Maximilians-University of Munich, University Hospital of LMU Munich, Munich, Germany
| | - Yang Peng
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haochen Yu
- Medical Faculty of Ludwig-Maximilians-University of Munich, University Hospital of LMU Munich, Munich, Germany
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
45
|
Cheng Y, Mao M, Lu Y. The biology of YAP in programmed cell death. Biomark Res 2022; 10:34. [PMID: 35606801 PMCID: PMC9128211 DOI: 10.1186/s40364-022-00365-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/18/2022] [Indexed: 02/08/2023] Open
Abstract
In the last few decades, YAP has been shown to be critical in regulating tumor progression. YAP activity can be regulated by many kinase cascade pathways and proteins through phosphorylation and promotion of cytoplasmic localization. Other factors can also affect YAP activity by modulating its binding to different transcription factors (TFs). Programmed cell death (PCD) is a genetically controlled suicide process present with the scope of eliminating cells unnecessary or detrimental for the proper development of the organism. In some specific states, PCD is activated and facilitates the selective elimination of certain types of tumor cells. As a candidate oncogene correlates with many regulatory factors, YAP can inhibit or induce different forms of PCD, including apoptosis, autophagy, ferroptosis and pyroptosis. Furthermore, YAP may act as a bridge between different forms of PCD, eventually leading to different outcomes regarding tumor development. Researches on YAP and PCD may benefit the future development of novel treatment strategies for some diseases. Therefore, in this review, we provide a general overview of the cellular functions of YAP and the relationship between YAP and PCD.
Collapse
Affiliation(s)
- Yifan Cheng
- Department of Gastrointestinal Surgery, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Misha Mao
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yong Lu
- Department of Gastrointestinal Surgery, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, Zhejiang, China.
| |
Collapse
|
46
|
Hsu SK, Chu YH, Syue WJ, Lin HYH, Chang WT, Chen JYF, Wu CY, Yen CH, Cheng KC, Chiu CC. The Role of Nonapoptotic Programmed Cell Death — Ferroptosis, Necroptosis, and Pyroptosis — in Pancreatic Ductal Adenocarcinoma Treatment. Front Oncol 2022; 12:872883. [PMID: 35664778 PMCID: PMC9160188 DOI: 10.3389/fonc.2022.872883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/19/2022] [Indexed: 11/26/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal cancer, with a dismal 5-year survival rate of less than 10%. It is estimated that approximately 80% of pancreatic ductal carcinoma (PDAC) patients are diagnosed at an advanced or metastatic stage. Hence, most patients are not appropriate candidates for surgical resection and therefore require systemic chemotherapy. However, it has been reported that most patients develop chemoresistance within several months, partly because of antiapoptotic mechanisms. Hence, inducing alternative programmed cell death (PCD), including ferroptosis, necroptosis or pyroptosis, seems to be a promising strategy to overcome antiapoptosis-mediated chemoresistance. In this review, we shed light on the molecular mechanisms of ferroptosis, necroptosis and pyroptosis and suggest several potential strategies (e.g., compounds and nanoparticles [NPs]) that are capable of triggering nonapoptotic PCD to suppress PDAC progression. In conclusion, these strategies might serve as adjuvants in combination with clinical first-line chemotherapies to improve patient survival rates.
Collapse
Affiliation(s)
- Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hsuan Chu
- Department of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wun-Jyun Syue
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hugo You-Hsien Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jeff Yi-Fu Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chia-Hung Yen
- The Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kai-Chun Cheng
- Department of Ophthalmology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Kai-Chun Cheng, ; Chien-Chih Chiu,
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Kai-Chun Cheng, ; Chien-Chih Chiu,
| |
Collapse
|
47
|
Guo J, Ye F, Xie W, Zhang X, Zeng R, Sheng W, Mi Y, Sheng X. The HOXC-AS2/miR-876-5p/HKDC1 axis regulates endometrial cancer progression in a high glucose-related tumor microenvironment. Cancer Sci 2022; 113:2297-2310. [PMID: 35485648 PMCID: PMC9277262 DOI: 10.1111/cas.15384] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
The tumor microenvironment (TME) is related to chronic inflammation and is currently identified as a risk factor for the occurrence and development of endometrial cancer (EC). Pyroptosis is a new proinflammatory form of programmed cell death that plays a critical role in the progression of multiple diseases. However, the important role of pyroptosis in high‐glucose (HG)‐related EC and the underlying molecular mechanisms remain elusive. In the present study, transcriptome high‐throughput sequencing revealed significantly higher hexokinase domain‐containing 1 (HKDC1) expression in EC patients with diabetes than in EC patients with normal glucose. Mechanistically, HKDC1 regulates HG‐induced cell pyroptosis by modulating the production of reactive oxygen species and pyroptosis‐induced cytokine release in EC. In addition, HKDC1 regulates TME formation by enhancing glycolysis, promoting a metabolic advantage in lactate‐rich environments to further accelerate EC progression. Subsequently, miR‐876‐5p was predicted to target the HKDC1 mRNA, and HOXC‐AS2 was identified to potentially inhibit the miR‐876‐5p/HKDC1 axis in regulating cell pyroptosis in HG‐related EC. Collectively, we elucidated the regulatory role of the HOXC‐AS2/miR‐876‐5p/HKDC1 signal transduction axis in EC cell pyroptosis at the molecular level, which may provide an effective therapeutic target for patients with diabetes who are diagnosed with EC.
Collapse
Affiliation(s)
- Jing Guo
- Department of Medical Oncology, Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, China
| | - Feng Ye
- Department of Medical Oncology, Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, China
| | - Wenli Xie
- Department of Gynecology, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Xinxin Zhang
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong, 250033, China
| | - Ru Zeng
- Department of Medical Oncology, Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, China
| | - Wang Sheng
- Department of Medical Oncology, Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, China
| | - Yanjun Mi
- Department of Medical Oncology, Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, China
| | - Xiugui Sheng
- Cancer Hospital of Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, Guangdong, 518116, China
| |
Collapse
|
48
|
Xing F, Hu Q, Qin Y, Xu J, Zhang B, Yu X, Wang W. The Relationship of Redox With Hallmarks of Cancer: The Importance of Homeostasis and Context. Front Oncol 2022; 12:862743. [PMID: 35530337 PMCID: PMC9072740 DOI: 10.3389/fonc.2022.862743] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/16/2022] [Indexed: 12/18/2022] Open
Abstract
Redox homeostasis is a lifelong pursuit of cancer cells. Depending on the context, reactive oxygen species (ROS) exert paradoxical effects on cancers; an appropriate concentration stimulates tumorigenesis and supports the progression of cancer cells, while an excessive concentration leads to cell death. The upregulated antioxidant system in cancer cells limits ROS to a tumor-promoting level. In cancers, redox regulation interacts with tumor initiation, proliferation, metastasis, programmed cell death, autophagy, metabolic reprogramming, the tumor microenvironment, therapies, and therapeutic resistance to facilitate cancer development. This review discusses redox control and the major hallmarks of cancer.
Collapse
Affiliation(s)
- Faliang Xing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- *Correspondence: Wei Wang, ; Xianjun Yu, ; Bo Zhang,
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- *Correspondence: Wei Wang, ; Xianjun Yu, ; Bo Zhang,
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- *Correspondence: Wei Wang, ; Xianjun Yu, ; Bo Zhang,
| |
Collapse
|
49
|
Yang Z, Chen Z, Wang Y, Wang Z, Zhang D, Yue X, Zheng Y, Li L, Bian E, Zhao B. A Novel Defined Pyroptosis-Related Gene Signature for Predicting Prognosis and Treatment of Glioma. Front Oncol 2022; 12:717926. [PMID: 35433410 PMCID: PMC9008739 DOI: 10.3389/fonc.2022.717926] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
Pyroptosis, a form of programmed cell death, that plays a significant role in the occurrence and progression of tumors, has been frequently investigated recently. However, the prognostic significance and therapeutic value of pyroptosis in glioma remain undetermined. In this research, we revealed the relationship of pyroptosis-related genes to glioma by analyzing whole transcriptome data from The Cancer Genome Atlas (TCGA) dataset serving as the training set and the Chinese Glioma Genome Atlas (CGGA) dataset serving as the validation set. We identified two subgroups of glioma patients with disparate prognostic and clinical features by performing consensus clustering analysis on nineteen pyroptosis-related genes that were differentially expressed between glioma and normal brain tissues. We further derived a risk signature, using eleven pyroptosis-related genes, that was demonstrated to be an independent prognostic factor for glioma. Furthermore, we used Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) to implement functional analysis of our gene set, and the results were closely related to immune and inflammatory responses in accordance with the characteristics of pyroptosis. Moreover, Gene Set Enrichment Analysis (GSEA) results showed that that the high-risk group exhibited enriched characteristics of malignant tumors in accordance with its poor prognosis. Next, we analyzed different immune cell infiltration between the two risk groups using ssGSEA. Finally, CASP1 was identified as a core gene, so we subsequently selected an inhibitor targeting CASP1 and simulated molecular docking. In addition, the inhibitory effect of belnacasan on glioma was verified at the cellular level. In conclusion, pyroptosis-related genes are of great significance for performing prognostic stratification and developing treatment strategies for glioma.
Collapse
Affiliation(s)
- Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Zhigang Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Yu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Zhiwei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Deran Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Xiaoyu Yue
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Yinfei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Lianxin Li
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Erbao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| |
Collapse
|
50
|
Cell pyroptosis in health and inflammatory diseases. Cell Death Dis 2022; 8:191. [PMID: 35411030 PMCID: PMC8995683 DOI: 10.1038/s41420-022-00998-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023]
Abstract
Inflammation is a defense mechanism that can protect the host against microbe invasion. A proper inflammatory response can maintain homeostasis, but continuous inflammation can cause many chronic inflammatory diseases. To properly treat inflammatory disorders, the molecular mechanisms underlying the development of inflammation need to be fully elucidated. Pyroptosis is an inflammation-related cell death program, that is different from other types of cell death. Pyroptosis plays crucial roles in host defense against infections through the release of proinflammatory cytokines and cell lysis. Accumulating evidence indicates that pyroptosis is associated with inflammatory diseases, such as arthritis, pneumonia, and colonitis. Furthermore, pyroptosis is also closely involved in cancers that develop as a result of inflammation, such as liver cancer, esophageal cancer, pancreatic cancer, and colon cancer. Here, we review the function and mechanism of pyroptosis in inflammatory disease development and provide a comprehensive description of the potential role of pyroptosis in inflammatory diseases.
Collapse
|