1
|
Zhang H, Ji Y, Yi Z, Zhao J, Liu J, Zhang X. Identification and Validation of Glycosylation‑Related Genes in Ischemic Stroke Based on Bioinformatics and Machine Learning. J Mol Neurosci 2025; 75:60. [PMID: 40299100 DOI: 10.1007/s12031-025-02352-5] [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: 03/11/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
Ischemic stroke (IS) constitutes a severe neurological disorder with restricted treatment alternatives. Recent investigations have disclosed that glycosylation is closely associated with the occurrence and outcome of IS. Nevertheless, data on the transcriptomic dynamics of glycosylation in IS are lacking. The objective of this study was to undertake a comprehensive exploration of glycosylation-related genes (GRGs) in IS via bioinformatics and to assess their immune characteristics. In this study, through the intersection of genes from weighted gene co-expression network analysis, GRGs from five glycosylation pathways, and DEGs from differential expression analysis, 20 candidate GRGs were identified. Subsequently, through LASSO, Random Forest, and SVM-RFE, 3 hub GRGs (F5, PPP6C, and UBE2J1) were identified. Additional, a gene diagnostic model linked to glycosylation was developed and validated. The findings indicated that the diagnostic model could effectively distinguish between IS patients and healthy individuals in the training, validation, and merging datasets, indicating clinical relevance. Subsequently, by employing unsupervised clustering analysis, IS patients were classified into three clusters, and significant disparities were witnessed in immune cell infiltration among distinct clusters. In summary, this study successfully identified hub GRGs in IS and investigated the roles of these hub genes in the immune microenvironment, indicating potential clinical applications for IS.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Yanan Ji
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Zhongquan Yi
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Jing Zhao
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Jianping Liu
- Department of Neurology, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China.
| | - Xianxian Zhang
- Department of Neurology, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China.
| |
Collapse
|
2
|
Huang K, Zhang Q, Wan H, Ban X, Chen X, Wan X, Lu R, He Y, Xiong K. TAK1 at the crossroads of multiple regulated cell death pathways: from molecular mechanisms to human diseases. FEBS J 2025. [DOI: 10.1111/febs.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 02/14/2025] [Indexed: 05/03/2025]
Abstract
Regulated cell death (RCD), the form of cell death that can be genetically controlled by multiple signaling pathways, plays an important role in organogenesis, tissue remodeling, and maintenance of organism homeostasis and is closely associated with various human diseases. Transforming growth factor‐beta‐activated kinase 1 (TAK1) is a member of the serine/threonine protein kinase family, which can respond to different internal and external stimuli and participate in inflammatory and immune responses. Emerging evidence suggests that TAK1 is an important regulator at the crossroad of multiple RCD pathways, including apoptosis, necroptosis, pyroptosis, and PANoptosis. The regulation of TAK1 affects disease progression through multiple signaling pathways, and therapeutic strategies targeting TAK1 have been proposed for inflammatory diseases, central nervous system diseases, and cancers. In this review, we provide an overview of the downstream signaling pathways regulated by TAK1 and its binding proteins. Their critical regulatory roles in different forms of cell death are also summarized. In addition, we discuss the potential of targeting TAK1 in the treatment of human diseases, with a specific focus on neurological disorders and cancer.
Collapse
Affiliation(s)
- Kun Huang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Xiangya School of Medicine Central South University Changsha China
| | - Qi Zhang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Department of Ophthalmology Stanford University School of Medicine Palo Alto CA USA
- Key Laboratory of Emergency and Trauma of Ministry of Education, College of Emergency and Trauma Hainan Medical University Haikou China
| | - Hao Wan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xiao‐Xia Ban
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xin‐Yu Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xin‐Xing Wan
- Department of Endocrinology Third Xiangya Hospital, Central South University Changsha China
| | - Rui Lu
- Department of Molecular and Cellular Physiology Stanford University Stanford CA USA
| | - Ye He
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Changsha Aier Eye Hospital China
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Key Laboratory of Emergency and Trauma of Ministry of Education, College of Emergency and Trauma Hainan Medical University Haikou China
- Hunan Key Laboratory of Ophthalmology Changsha China
| |
Collapse
|
3
|
Tu A, Wu H, Wang J, Hou X, Wang M, Jiang M, Zhou X. Hypermethylation of miR-129-2-3p inhibits esophageal cancer proliferation and migration by down-regulating PPP6C expression. Am J Transl Res 2025; 17:1459-1469. [PMID: 40092081 PMCID: PMC11909538 DOI: 10.62347/wjgt6717] [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: 01/18/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVE MicroRNAs (miRNAs) play crucial roles in gene regulation, and their dysregulation is associated with various diseases, including cancer. Abnormal DNA methylation can alter gene expression and influence carcinogenesis. DNA methylation-based biomarkers are emerging as promising tools for early cancer diagnosis. This study aimed to investigate the role of miR-129-2-3p in esophageal cancer (EC) and explore its potential as a diagnostic biomarker. METHODS To achieve these objectives, we employed multi-sample MethylTarget technology to assess the methylation status of miR-129-2-3p in EC tissues. The diagnostic value of miR-129-2-3p was evaluated using logistic regression and receiver operating characteristic (ROC) curve analysis. Functional assays were conducted to examine the effects of miR-129-2-3p overexpression on EC cell proliferation and migration. Luciferase reporter assays were performed to confirm Protein Phosphatase 6 Catalytic Subunit (PPP6C) as a direct target of miR-129-2-3p. Finally, the impact of PPP6C overexpression on the inhibitory effects induced by miR-129-2-3p was assessed. RESULTS We found that miR-129-2-3p is hypermethylated in EC tissues. Diagnostic analysis revealed that miR-129-2-3p had a sensitivity of 0.884, a specificity of 0.659, and an area under the curve (AUC) of 0.799. Overexpression of miR-129-2-3p significantly suppressed EC cell proliferation and migration. Furthermore, PPP6C was identified as a direct target of miR-129-2-3p, and its expression was suppressed. The elevation of PPP6C counteracted the inhibitory effects of miR-129-2-3p on EC cell proliferation and migration. CONCLUSION Hypermethylated miR-129-2-3p inhibits EC cell proliferation and migration by downregulating PPP6C expression, suggesting that miR-129-2-3p may serve as a potential diagnostic biomarker for EC.
Collapse
Affiliation(s)
- Ailing Tu
- Department of Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Han Wu
- Department of Biochemistry and Molecular Biology, Medical College, Soochow UniversitySuzhou, Jiangsu, China
| | - Junjie Wang
- Department of Biochemistry and Molecular Biology, Medical College, Soochow UniversitySuzhou, Jiangsu, China
| | - Xinyang Hou
- Department of Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Minghua Wang
- Department of Biochemistry and Molecular Biology, Medical College, Soochow UniversitySuzhou, Jiangsu, China
| | - Meng Jiang
- Department of Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Xiumin Zhou
- Department of Oncology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| |
Collapse
|
4
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [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: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
5
|
Wu J, Ai T, He P, Shi Q, Li Y, Zhang Z, Chen M, Huang Z, Wu S, Chen W, Han J. Cecal necroptosis triggers lethal cardiac dysfunction in TNF-induced severe SIRS. Cell Rep 2024; 43:114778. [PMID: 39325617 DOI: 10.1016/j.celrep.2024.114778] [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: 03/11/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Tumor necrosis factor (TNF) induces systemic inflammatory response syndrome (SIRS), and severe SIRS can serve as a model for studying animal death caused by organ failure. Through strategic cecectomy, we demonstrate that necroptosis in the cecum initiates the death process in TNF-treated mice, but it is not the direct cause of death. Instead, we show that it is the cardiac dysfunction downstream of cecum damage that ultimately leads to the death of TNF-treated mice. By in vivo and ex vivo physiological analyses, we reveal that TNF and the damage-associated molecular patterns (DAMPs) released from necroptotic cecal cells jointly target cardiac endothelial cells, triggering caspase-8 activation and subsequent cardiac endothelial damage. Cardiac endothelial damage is a primary cause of the deterioration of diastolic function in the heart of TNF-treated mice. Our research provides insights into the pathophysiological process of TNF-induced lethality.
Collapse
Affiliation(s)
- Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Ai
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Peng He
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Qilin Shi
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangxin Li
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Ziguan Zhang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Minwei Chen
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Zhengrong Huang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Suqin Wu
- Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Wanze Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518000, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
6
|
Akanyibah FA, Zhu Y, Jin T, Ocansey DKW, Mao F, Qiu W. The Function of Necroptosis and Its Treatment Target in IBD. Mediators Inflamm 2024; 2024:7275309. [PMID: 39118979 PMCID: PMC11306684 DOI: 10.1155/2024/7275309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/22/2024] [Accepted: 07/13/2024] [Indexed: 08/10/2024] Open
Abstract
Inflammatory bowel disease (IBD), which encompasses Crohn's disease (CD) and ulcerative colitis (UC), is a complicated illness whose exact cause is yet unknown. Necroptosis is associated with IBD pathogenesis, leading to intestinal barrier abnormalities and uncontrolled inflammation. Molecules involved in necroptosis, however, exhibit different expression levels in IBD and its associated colorectal cancer. Multiple studies have shown that inhibiting these molecules alleviates necroptosis-induced IBD. Moreover, due to the severe scarcity of clinical medications for treating IBD caused by necroptosis, we review the various functions of crucial necroptosis molecules in IBD, the stimuli regulating necroptosis, and the current emerging therapeutic strategies for treating IBD-associated necroptosis. Eventually, understanding the pathogenesis of necroptosis in IBD will enable the development of additional therapeutic approaches for the illness.
Collapse
Affiliation(s)
- Francis Atim Akanyibah
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceDepartment of Laboratory MedicineSchool of MedicineJiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Yi Zhu
- The People's Hospital of DanyangAffiliated Danyang Hospital of Nantong University, Zhenjiang 212300, Jiangsu, China
| | - Tao Jin
- Department of Gastrointestinal and EndoscopyThe Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceDepartment of Laboratory MedicineSchool of MedicineJiangsu University, Zhenjiang 212013, Jiangsu, China
- Directorate of University Health ServicesUniversity of Cape Coast, Cape Coast CC0959347, Ghana
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceDepartment of Laboratory MedicineSchool of MedicineJiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Wei Qiu
- Nanjing Jiangning Hospital, Nanjing 211100, Jiangsu, China
| |
Collapse
|
7
|
Bayat M, Nahand JS. Let's make it personal: CRISPR tools in manipulating cell death pathways for cancer treatment. Cell Biol Toxicol 2024; 40:61. [PMID: 39075259 PMCID: PMC11286699 DOI: 10.1007/s10565-024-09907-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024]
Abstract
Advancements in the CRISPR technology, a game-changer in experimental research, have revolutionized various fields of life sciences and more profoundly, cancer research. Cell death pathways are among the most deregulated in cancer cells and are considered as critical aspects in cancer development. Through decades, our knowledge of the mechanisms orchestrating programmed cellular death has increased substantially, attributed to the revolution of cutting-edge technologies. The heroic appearance of CRISPR systems have expanded the available screening platform and genome engineering toolbox to detect mutations and create precise genome edits. In that context, the precise ability of this system for identification and targeting of mutations in cell death signaling pathways that result in cancer development and therapy resistance is an auspicious choice to transform and accelerate the individualized cancer therapy. The concept of personalized cancer therapy stands on the identification of molecular characterization of the individual tumor and its microenvironment in order to provide a precise treatment with the highest possible outcome and minimum toxicity. This study explored the potential of CRISPR technology in precision cancer treatment by identifying and targeting specific cell death pathways. It showed the promise of CRISPR in finding key components and mutations involved in programmed cell death, making it a potential tool for targeted cancer therapy. However, this study also highlighted the challenges and limitations that need to be addressed in future research to fully realize the potential of CRISPR in cancer treatment.
Collapse
Affiliation(s)
- Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 15731, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 15731, Iran.
| |
Collapse
|
8
|
Du J, Wang Z. Regulation of RIPK1 Phosphorylation: Implications for Inflammation, Cell Death, and Therapeutic Interventions. Biomedicines 2024; 12:1525. [PMID: 39062098 PMCID: PMC11275223 DOI: 10.3390/biomedicines12071525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/04/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Receptor-interacting protein kinase 1 (RIPK1) plays a crucial role in controlling inflammation and cell death. Its function is tightly controlled through post-translational modifications, enabling its dynamic switch between promoting cell survival and triggering cell death. Phosphorylation of RIPK1 at various sites serves as a critical mechanism for regulating its activity, exerting either activating or inhibitory effects. Perturbations in RIPK1 phosphorylation status have profound implications for the development of severe inflammatory diseases in humans. This review explores the intricate regulation of RIPK1 phosphorylation and dephosphorylation and highlights the potential of targeting RIPK1 phosphorylation as a promising therapeutic strategy for mitigating human diseases.
Collapse
Affiliation(s)
- Jingchun Du
- Department of Clinical Immunology, Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 510182, China
| | - Zhigao Wang
- Center for Regenerative Medicine, Heart Institute, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 560 Channelside Drive, Tampa, FL 33602, USA
| |
Collapse
|
9
|
Bynigeri RR, Malireddi RKS, Mall R, Connelly JP, Pruett-Miller SM, Kanneganti TD. The protein phosphatase PP6 promotes RIPK1-dependent PANoptosis. BMC Biol 2024; 22:122. [PMID: 38807188 PMCID: PMC11134900 DOI: 10.1186/s12915-024-01901-5] [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: 11/15/2023] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The innate immune system serves as the first line of host defense. Transforming growth factor-β-activated kinase 1 (TAK1) is a key regulator of innate immunity, cell survival, and cellular homeostasis. Because of its importance in immunity, several pathogens have evolved to carry TAK1 inhibitors. In response, hosts have evolved to sense TAK1 inhibition and induce robust lytic cell death, PANoptosis, mediated by the RIPK1-PANoptosome. PANoptosis is a unique innate immune inflammatory lytic cell death pathway initiated by an innate immune sensor and driven by caspases and RIPKs. While PANoptosis can be beneficial to clear pathogens, excess activation is linked to pathology. Therefore, understanding the molecular mechanisms regulating TAK1 inhibitor (TAK1i)-induced PANoptosis is central to our understanding of RIPK1 in health and disease. RESULTS In this study, by analyzing results from a cell death-based CRISPR screen, we identified protein phosphatase 6 (PP6) holoenzyme components as regulators of TAK1i-induced PANoptosis. Loss of the PP6 enzymatic component, PPP6C, significantly reduced TAK1i-induced PANoptosis. Additionally, the PP6 regulatory subunits PPP6R1, PPP6R2, and PPP6R3 had redundant roles in regulating TAK1i-induced PANoptosis, and their combined depletion was required to block TAK1i-induced cell death. Mechanistically, PPP6C and its regulatory subunits promoted the pro-death S166 auto-phosphorylation of RIPK1 and led to a reduction in the pro-survival S321 phosphorylation. CONCLUSIONS Overall, our findings demonstrate a key requirement for the phosphatase PP6 complex in the activation of TAK1i-induced, RIPK1-dependent PANoptosis, suggesting this complex could be therapeutically targeted in inflammatory conditions.
Collapse
Affiliation(s)
- Ratnakar R Bynigeri
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Raghvendra Mall
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Current affiliation: Biotechnology Research Center, Technology Innovation Institute, Abu Dhabi, United Arab Emirates
| | - Jon P Connelly
- Center for Advanced Genome Engineering (CAGE), St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering (CAGE), St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | |
Collapse
|
10
|
Kondo A, Tanaka H, Rai S, Shima H, Matsumura I, Watanabe T. Depletion of Ppp6c in hematopoietic and vascular endothelial cells causes embryonic lethality and decreased hematopoietic potential. Exp Hematol 2024; 133:104205. [PMID: 38490577 DOI: 10.1016/j.exphem.2024.104205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
Protein phosphatase 6 (PP6) is a serine/threonine (Ser/Thr) protein phosphatase, and its catalytic subunit is Ppp6c. PP6 forms the PP2A subfamily with PP2A and PP4. The diverse phenotypes observed following small interfering RNA (siRNA)-based knockdown of Ppp6c in cultured mammalian cells suggest that PP6 plays roles in cell growth and DNA repair. There is also evidence that PP6 regulates nuclear factor kappa B (NF-κB) signaling and mitogen-activated protein kinases and inactivates transforming growth factor-β-activated kinase 1 (TAK1). Loss of Ppp6c causes several abnormalities, including those of T cell and regulatory T cell function, neurogenesis, oogenesis, and spermatogenesis. PP2A has been reported to play an important role in erythropoiesis. However, the roles of PP6 in other hematopoietic cells have not been investigated. We generated Ppp6cfl/fl;Tie2-Cre (Ppp6cTKO) mice, in which Ppp6c was specifically deleted in hematopoietic and vascular endothelial cells. Ppp6cTKO mice displayed embryonic lethality. Ppp6c deficiency increased the number of dead cells and decreased the percentages of erythroid and monocytic cells during fetal hematopoiesis. By contrast, the number of Lin-Sca-1+c-Kit+ cells, which give rise to all hematopoietic cells, was slightly increased, but their colony-forming cell activity was markedly decreased. Ppp6c deficiency also increased phosphorylation of extracellular signal-regulated kinase 1/2 and c-Jun amino (N)-terminal kinase in fetal liver hematopoietic cells.
Collapse
Affiliation(s)
- Ayumi Kondo
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Nara, Japan
| | - Hirokazu Tanaka
- Department of Hematology and Rheumatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Shinya Rai
- Department of Hematology and Rheumatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Hiroshi Shima
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Toshio Watanabe
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Nara, Japan.
| |
Collapse
|
11
|
Ito M, Tanuma N, Kotani Y, Murai K, Kondo A, Sumiyoshi M, Shima H, Matsuda S, Watanabe T. Oncogenic K-Ras G12V cannot overcome proliferation failure caused by loss of Ppp6c in mouse embryonic fibroblasts. FEBS Open Bio 2024; 14:545-554. [PMID: 38318686 PMCID: PMC10988750 DOI: 10.1002/2211-5463.13775] [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: 09/28/2023] [Revised: 12/11/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Protein phosphatase 6 is a Ser/Thr protein phosphatase and its catalytic subunit is Ppp6c. Ppp6c is thought to be indispensable for proper growth of normal cells. On the other hand, loss of Ppp6c accelerates growth of oncogenic Ras-expressing cells. Although it has been studied in multiple contexts, the role(s) of Ppp6c in cell proliferation remains controversial. It is unclear how oncogenic K-Ras overcomes cell proliferation failure induced by Ppp6c deficiency; therefore, in this study, we attempted to shed light on how oncogenic K-Ras modulates tumor cell growth. Contrary to our expectations, loss of Ppp6c decreased proliferation, anchorage-independent growth in soft agar, and tumor formation of oncogenic Ras-expressing mouse embryonic fibroblasts (MEFs). These findings show that oncogenic K-RasG12V cannot overcome proliferation failure caused by loss of Ppp6c in MEFs.
Collapse
Affiliation(s)
- Mai Ito
- Department of Biological Science, Graduate School of Humanities and SciencesNara Women's UniversityJapan
| | - Nobuhiro Tanuma
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Yui Kotani
- Department of Biological Science, Graduate School of Humanities and SciencesNara Women's UniversityJapan
| | - Kokoro Murai
- Department of Biological Science, Graduate School of Humanities and SciencesNara Women's UniversityJapan
| | - Ayumi Kondo
- Department of Biological Science, Graduate School of Humanities and SciencesNara Women's UniversityJapan
| | - Mami Sumiyoshi
- Department of Cell Signaling, Institute of Biomedical ScienceKansai Medical UniversityHirakataJapan
| | - Hiroshi Shima
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Satoshi Matsuda
- Department of Cell Signaling, Institute of Biomedical ScienceKansai Medical UniversityHirakataJapan
| | - Toshio Watanabe
- Department of Biological Science, Graduate School of Humanities and SciencesNara Women's UniversityJapan
| |
Collapse
|
12
|
KITAMURA N, OHAMA T, SATO K. Protein phosphatase 6 promotes transforming growth factor-β signaling in mouse embryonic fibroblasts. J Vet Med Sci 2023; 85:1319-1323. [PMID: 37880139 PMCID: PMC10788163 DOI: 10.1292/jvms.23-0380] [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: 09/07/2023] [Accepted: 10/15/2023] [Indexed: 10/27/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) is a multifunctional cytokine that controls various cellular processes. Protein phosphatase 6 (PP6) is an evolutionarily conserved serine/threonine protein phosphatase with diverse functions in cell signaling. However, it has not been linked to TGF-β signaling. We found that TGF-β treatment increased PP6 protein levels via transcriptional and post-translational regulation. Loss of the Ppp6c gene suppressed TGF-β-induced canonical Smad3 phosphorylation and its transcriptional activity. PP6 knockout also inhibited non-canonical p38 mitogen-activated protein kinase (MAPK) pathway. Moreover, PP6 depletion suppressed cell migration induced by TGF-β. These findings uncovered the role of PP6 as a positive regulator for TGF-β signaling.
Collapse
Affiliation(s)
- Nao KITAMURA
- Laboratory of Veterinary Pharmacology, Yamaguchi University
Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan
| | - Takashi OHAMA
- Laboratory of Veterinary Pharmacology, Yamaguchi University
Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan
| | - Koichi SATO
- Laboratory of Veterinary Pharmacology, Yamaguchi University
Joint Graduate School of Veterinary Medicine, Yamaguchi, Japan
| |
Collapse
|
13
|
Lu Y, Wang R, He S, Zhang Q, Wei J, Hu J, Ding Y. Downregulation of BUBR1 regulates the proliferation and cell cycle of breast cancer cells and increases the sensitivity of cells to cisplatin. In Vitro Cell Dev Biol Anim 2023; 59:778-789. [PMID: 38048028 DOI: 10.1007/s11626-023-00823-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: 09/06/2023] [Accepted: 11/01/2023] [Indexed: 12/05/2023]
Abstract
Breast cancer (BC) is a significant tissue for women's health worldwide. The spindle assembly checkpoint protein family includes BUBR1 (Bub1-related kinase or MAD3/Bub1b). High expression of BUBR1 promotes cell cycle disorders, leading to cell carcinogenesis and cancer progression. However, the underlying molecular mechanism and the role of BUBR1 in BC progression are unclear. The published dataset was analyzed to evaluate the clinical relevance of BUBR1. BUBR1 was knocked down in BC cells using shRNA. The CCK-8 assay was used to measure the cell viability, and mRNA and protein expression levels were detected by RT-qPCR and Western blot (WB). Cell apoptosis and cell cycle were detected by flow cytometry. Subcutaneous xenograft model was used to assess in vivo tumor growth. BUBR1 was found to be highly expressed in BC. The high expression of BUBR1 was associated with poor prognosis of BC patients. Upon BUBR1 knockdown using shRNA, the proliferation and metastatic ability of cells were decreased. Moreover, the cells with BUBR1 knockdown underwent cell cycle arrest. And the results showed that BUBR1 loss inhibited the phosphorylation of TAK1/JNK. In vitro and in vivo studies indicated the knockdown of BUBR1 rendered the BC cells more sensitive to cisplatin. In summary, BUBR1 may be a potential therapeutic target for BC and targeting BUBR1 may help overcome cisplatin resistance in BC patients.
Collapse
Affiliation(s)
- Yiran Lu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun City, Jilin, 130062, China
| | - Ruiqing Wang
- The Eye Center in the Second Hospital of Jilin University, Nanguan District, Ziqiang Street 218#, Changchun City, Jilin, 130041, China
| | - Song He
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun City, Jilin, 130062, China
| | - Qing Zhang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun City, Jilin, 130062, China
| | - Jiahui Wei
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun City, Jilin, 130062, China
| | - Jinping Hu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun City, Jilin, 130062, China
| | - Yu Ding
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun City, Jilin, 130062, China.
| |
Collapse
|
14
|
Mechanisms of TNF-independent RIPK3-mediated cell death. Biochem J 2022; 479:2049-2062. [PMID: 36240069 DOI: 10.1042/bcj20210724] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022]
Abstract
Apoptosis and necroptosis regulate many aspects of organismal biology and are involved in various human diseases. TNF is well known to induce both of these forms of cell death and the underlying mechanisms have been elaborately described. However, cells can also engage apoptosis and necroptosis through TNF-independent mechanisms, involving, for example, activation of the pattern recognition receptors Toll-like receptor (TLR)-3 and -4, or zDNA-binding protein 1 (ZBP1). In this context, cell death signaling depends on the presence of receptor-interacting serine/threonine protein kinase 3 (RIPK3). Whereas RIPK3 is required for TNF-induced necroptosis, it mediates both apoptosis and necroptosis upon TLR3/4 and ZBP1 engagement. Here, we review the intricate mechanisms by which TNF-independent cell death is regulated by RIPK3.
Collapse
|