1
|
Sun C, Gui J, Sheng Y, Huang L, Zhu X, Huang K. Specific signaling pathways mediated programmed cell death in tumor microenvironment and target therapies. Discov Oncol 2025; 16:776. [PMID: 40377777 PMCID: PMC12084487 DOI: 10.1007/s12672-025-02592-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Increasing evidence has shown that programmed cell death (PCD) plays a crucial role in tumorigenesis and cancer progression. The components of PCD are complex and include various mechanisms such as apoptosis, necroptosis, alkaliptosis, oxeiptosis, and anoikis, all of which are interrelated in their functions and regulatory pathways. Given the significance of these processes, it is essential to conduct a comprehensive study on PCD to elucidate its multifaceted nature. Key signaling pathways, particularly the caspase signaling pathway, the RIPK1/RIPK3/MLKL pathway, and the mTOR signaling pathway, are pivotal in regulating PCD and influencing tumor progression. In this review, we briefly describe the generation mechanisms of different PCD components and focus on the regulatory mechanisms of these three major signaling pathways within the context of global PCD. Furthermore, we discuss various tumor therapeutic compounds that target different signaling axes of these pathways, which may provide novel strategies for effective tumor therapy and help improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Chengpeng Sun
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Jiawei Gui
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Yilei Sheng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Le Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China
| | - Xingen Zhu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Kai Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
2
|
Sharma A, Pettee D, Mella C, Hord C, Brockwell M, Hardy S, Ball HC, Safadi FF, Kuerbitz SJ. Epigenetic Inactivation of RIPK3-Dependent Necroptosis Augments Cisplatin Chemoresistance in Human Osteosarcoma. Int J Mol Sci 2025; 26:3863. [PMID: 40332549 PMCID: PMC12027565 DOI: 10.3390/ijms26083863] [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: 02/13/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy in children and adolescents. Unfortunately, drug resistance limits the efficacy of chemotherapeutic treatment and compromises therapeutic outcomes in a substantial proportion of cases. Aberrant CpG island methylation-associated transcriptional silencing contributes to chemoresistance in pediatric solid tumors. Here, using whole-genome DNA methylation screening on 16 human primary OS specimens, we identify receptor interacting protein kinase-3 (RIPK3), a molecular regulator of the necroptosis programmed cell death pathway, as a gene target of aberrant CpG methylation and demonstrate its role in human OS chemoresistance. We validated these findings via enforced expression and DsiRNA silencing, and evaluated the role of RIPK3 in cisplatin chemosensitivity and necroptosis activation through MLKL phosphorylation. We found that CpG island methylation results in RIPK3 silencing in primary human OS samples and cell lines. Enforced RIPK3 expression significantly enhanced cisplatin cytotoxicity in OS cells and DsiRNA knockdown reversed the cisplatin-sensitive phenotype. In cells with enforced RIPK3 expression, cisplatin treatment significantly increased phosphorylation of both RIPK3 and its target, MLKL, indicative of induction of necroptosis. Here, we identify RIPK3 as an important mediator of chemoresistance in OS and a potential pharmacologic target to improve chemotherapy efficacy in drug-resistant tumors.
Collapse
Affiliation(s)
- Aditya Sharma
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
| | - Daniel Pettee
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
| | - Christine Mella
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
| | - Catherine Hord
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
| | - Maximilian Brockwell
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
| | - Samantha Hardy
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
| | - Hope C. Ball
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
| | - Fayez F. Safadi
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
- University Hospitals Health Systems, 11100 Euclid Ave, Cleveland, OH 44106, USA
| | - Steven J. Kuerbitz
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
| |
Collapse
|
3
|
Li G, Guo Y, Zhang Y. Role of necroptosis and immune infiltration in essential thrombocytosis. Hereditas 2025; 162:62. [PMID: 40229837 PMCID: PMC11995491 DOI: 10.1186/s41065-025-00428-1] [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: 01/17/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Necroptosis, a recently identified form of programmed cell death involved in the pathogenesis of a variety of tumor and non-tumor diseases. Nevertheless, the function of necroptosis in essential thrombocytosis (ET) remains unclear, which is a classic myeloproliferative tumor. MATERIALS AND METHODS The role of necroptosis in ET was determined via bioinformatics combined with qRT-PCR analysis of clinical samples. GSE57793 and GSE26049 datasets were recruited to identify necroptosis differentially expressed genes based on differential gene identification, necroptosis gene sets and data machine learning. Enrichment analysis (GSEA) was used to evaluate the gene enrichment signaling pathway of ET, immune infiltration analysis was used to explore the abundance of immune cell infiltration in ET, and the correlation between necroptosis differential genes and immune cell infiltration was studied. RESULTS Five necroptosis genes were recognized to be remarkably enriched in the necroptosis pathway, including CHMP1B, FTH1, HSP90AB1, IL1A, and RBCK1. The imbalance of invasion of Th1/Th17 cells was identified in ET, and the differential necroptosis gene was positively correlated with the infiltration of multiple immune cells. There is significant necroptosis in ET, which is enriched in the necrotizing apoptotic pathway, and is associated with immune infiltration. CONCLUSIONS Necroptosis might drive the progression of ET via stimulating immune infiltration and immune responses. The findings bring new insights into the treatment mechanism and treatment strategy of ET in the future.
Collapse
Affiliation(s)
- Guangming Li
- Surgery Base Training, Shanghai Fengxian District Central Hospital, Shanghai, 201499, China
| | - Ying Guo
- Department of Hematology, Daqing Oilfield General Hospital, Daqing City, Heilongjiang Province, 163001, China
| | - Yuanyuan Zhang
- Internal Medicine Base Training, Shanghai Fengxian District Central Hospital, No.6600, Nanfeng Highway, Nanqiao Town, Fengxian District, Shanghai, 201499, China.
| |
Collapse
|
4
|
Yao GS, Dai JS, Fu LM, Lin J, Tan ZP, Dai L, Chen W, Luo JH, Wei JH. Development and validation of hierarchical signature for precision individualized therapy based on the landscape associated with necroptosis in clear cell renal cell carcinoma. Front Pharmacol 2025; 16:1470145. [PMID: 40255563 PMCID: PMC12006085 DOI: 10.3389/fphar.2025.1470145] [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: 07/25/2024] [Accepted: 03/04/2025] [Indexed: 04/22/2025] Open
Abstract
Background Increasing evidence is showing that necroptosis has unique clinical significance in the occurrence and development of multiple diseases. Here, we systematically evaluate the role of necroptosis in clear cell renal cell carcinoma (ccRCC) and analyze its regulatory patterns. Methods First, we evaluated the expression and enrichment of necroptotic factors in ccRCC using gene set enrichment analysis (GSEA) and survival analysis in the expression profile from The Cancer Genome Atlas (TCGA) to demonstrate the overall mutation of necroptotic pathway genes. Then, we used unsupervised clustering to divide the samples into two subtypes related to necroptosis with significant differences in overall survival (OS) and subsequently detected the differentially expressed genes (DEGs) between them. Based on this, we constructed the necroptosis scoring system (NSS), which also performed outstandingly in hierarchical data. Finally, we analyzed the association between NSS and clinical parameters, immune infiltration, and the efficacy of immunotherapy containing immune checkpoint inhibitors (ICIs), and we suggested potential therapeutic strategies. Results We screened 97 necroptosis-related genes and demonstrated that they were dysregulated in ccRCC. Using Cox analysis and least absolute shrinkage and selection operator (LASSO) regression, a prognostic prediction signature of seven genes was built. Receiver operating characteristic (ROC) curves and Kaplan-Meier (KM) analyses both showed that the model was accurate, and univariate/multivariate Cox analysis showed that as an independent prognostic factor, the higher the risk score, the poorer the survival outcome. Furthermore, the predicted scores based on the signature were observably associated with immune cell infiltration and the mutation of specific genes. In addition, the risk score could potentially predict patients' responsiveness to different chemotherapy regimens. Specifically, Nivolumab is more effective for patients with higher scores. Conclusion The necroptosis-related signature we constructed can accurately predict the prognosis of ccRCC patients and further provide clues for targeted, individualized therapy.
Collapse
Affiliation(s)
- Gao-Sheng Yao
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jun-Shang Dai
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
- Department of Obstetrics and Gynecology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Liang-Min Fu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
| | - Juan Lin
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhi-Ping Tan
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Lei Dai
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Wei Chen
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jun-Hang Luo
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
- Institute of Precision Medicine, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jin-Huan Wei
- Department of Urology, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Kim DH, Yu JE, Lee DH, Kim MJ, Jeon SH, Yun J, Son DJ, Kim B, Yong YJ, Lim YS, Kim TH, Khalid AM, Han SB, Lee YS, Hong JT. Anti-arthritis Effect of Anti-chitinase-3-like 1 Antibody Through Inhibition of MMP3. Immune Netw 2025; 25:e5. [PMID: 40342842 PMCID: PMC12056290 DOI: 10.4110/in.2025.25.e5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/30/2024] [Accepted: 01/05/2025] [Indexed: 05/11/2025] Open
Abstract
Chitinase-3-like 1 (CHI3L1) is a key factor in regulating inflammatory processes and development of rheumatoid arthritis (RA) since is highly produced by synoviocytes and macrophages in the development RA. Collagen-induced arthritis (CIA) model is the most widely used because its pathogenesis is similar to human RA. Thus, we aimed to investigate if anti-CHI3L1 antibody could reduce RA development in the CIA model. To induce CIA, DBA1/J mice were immunized with a type II bovine collagen emulsion in complete Freund's adjuvant, and boosted type II bovine collagen. THP-1 and MH7A cells were used for pro-inflammation responses. Anti-CHI3L1 Ab treatment reduced the RA clinical score and paw thickness of mice. Inflammation-induced matrix metalloproteinase 3 (MMP3) expression was reduced by inhibiting CHI3L1, and MMP3 knockdown suppressed the expression of RA-related inflammatory cytokines in LPS-treated THP-1 and MH7A cells. Our findings suggest that anti-CHI3L1 Ab showed significant anti-arthritic effects by inhibiting MMP3 expression.
Collapse
Affiliation(s)
- Dae Hwan Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Ji Eun Yu
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Dong Hun Lee
- Department of Biological Sciences, Chonnam National University, Gwangju 61186, Korea
| | - Min Ji Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Seong Hee Jeon
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | | | | | | | | | | | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Yong Sun Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea
| |
Collapse
|
6
|
Bao X, Chen Y, Chang J, Du J, Yang C, Wu Y, Sha Y, Li M, Chen S, Yang M, Liu SB. Machine learning-based bulk RNA analysis reveals a prognostic signature of 13 cell death patterns and potential therapeutic target of SMAD3 in acute myeloid leukemia. BMC Cancer 2025; 25:273. [PMID: 39955536 PMCID: PMC11830216 DOI: 10.1186/s12885-025-13658-3] [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: 04/02/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Dysregulation or abnormality of the programmed cell death (PCD) pathway is closely related to the occurrence and development of many tumors, including acute myeloid leukemia (AML). Studying the abnormal characteristics of PCD pathway-related molecular markers can provide a basis for prognosis prediction and targeted drug design in AML patients. METHODS A total of 1394 genes representing 13 different PCD pathways were examined in AML patients and healthy donors. The upregulated genes were analyzed for their ability to predict overall survival (OS) individually, and these prognostic genes were subsequently combined to construct a PCD-related prognostic signature via an integrated approach consisting of 101 models based on ten machine learning algorithms. RNA transcriptome and clinical data from multiple AML cohorts (TCGA-AML, GSE106291, GSE146173 and Beat AML) were obtained to develop and validate the AML prognostic model. RESULTS A total of 214 upregulated PCD-related genes were identified in AML patients, 39 of which were proven to be prognostic genes in the training cohort. On the basis of the average C-index and number of model genes identified from the machine learning combinations, a PCD index was developed and validated for predicting AML OS. A prognostic nomogram was then generated and validated on the basis of the PCD index, age and ELN risk stratification in the Beat AML cohort and the GSE146173 cohort, revealing satisfactory predictive power (AUC values ≥ 0.7). With different mutation patterns, a higher PCD index was associated with a worse OS. The PCD index was significantly related to higher scores for immunosuppressive cells and mature leukemia cell subtypes. As the gene most closely related to the PCD index, the expression of SMAD3 was further validated in vitro. AML cells harboring KMT2A rearrangements were more sensitive to the SMAD3 inhibitor SIS3, and the expression of the autophagy-related molecular marker LC3 was increased in KMT2A-rearranged cell lines after SIS3 monotherapy and combined treatment. CONCLUSION The PCD index and SMAD3 gene expression levels have potential prognostic value and can be used in targeted therapy for AML, and these findings can lead to the development of effective strategies for the combined treatment of high-risk AML patients.
Collapse
Affiliation(s)
- Xiebing Bao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yao Chen
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Jie Chang
- School of Public Health, Medical College of Soochow University, Suzhou, 215123, China
| | - Jiahui Du
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Chen Yang
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Yijie Wu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Yu Sha
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Ming Li
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Minfeng Yang
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong, 226019, China.
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
| | - Song-Bai Liu
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China.
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China.
| |
Collapse
|
7
|
Wang Y, Zhang TJ, Zhang LC, Xu ZJ, Chu MQ, Zhao YJ, Lin J, Qian J, Zhou JD. Overexpression and oncogenic role of RIPK3 in acute myeloid leukemia associated with specific subtypes and treatment outcome. BMC Cancer 2025; 25:253. [PMID: 39948488 PMCID: PMC11827379 DOI: 10.1186/s12885-025-13613-2] [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: 02/28/2024] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Receptor-interacting protein kinase 3 (RIPK3) has been implicated in the pathogenesis of diverse human cancers. However, the role of RIPK3 in acute myeloid leukemia (AML) is not fully understood, which needs further research and clarification. METHODS We first identified the expression and clinical prognostic value of RIPK3 in AML through a public database and further validated in our research cohort. In addition, the biological function of RIPK3 in leukemic development was further verified through in vitro experiments. RESULTS Based on the GEPIA database, we screened that RIPK3 overexpression among RIPK family was associated with poor prognosis in AML. Afterwards, another independent cohort from our research center further confirmed the expression pattern of RIPK3 in AML patients. Clinically, increased RIPK3 expression was closely related to specific subtypes of AML, such as FAB-M4/M5, normal karyotype and NPM1 mutation. The significant association of RIPK3 overexpression with FAB-M4/M5 was further validated in AML cell lines. Notably, AML patients with RIPK3 overexpression received transplantation presented a markedly longer survival than those just receiving chemotherapy, whereas those with RIPK3 underexpression showed similar survival between transplantation and chemotherapy group. Bioinformatics analysis showed the significant association of RIPK3 expression with diverse oncogenes/tumor suppressor genes and tumor-related biological processes in AML. Subsequently, we further performed functional experiments in vitro confirmed the potential oncogenic role of RIPK3 in AML. CONCLUSIONS Overexpression of RIPK3 was associated with specific subtypes of AML, such as FAB-M4/M5, normal karyotype and NPM1 mutation, and may facilitate the leukemic development. Moreover, RIPK3 overexpression was associated poor prognosis, and may guide treatment choice in AML.
Collapse
MESH Headings
- Humans
- Receptor-Interacting Protein Serine-Threonine Kinases/genetics
- Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Nucleophosmin
- Prognosis
- Male
- Female
- Treatment Outcome
- Cell Line, Tumor
- Middle Aged
- Mutation
- Biomarkers, Tumor/genetics
- Adult
Collapse
Affiliation(s)
- Yun Wang
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, 212002, China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, 212002, China
| | - Ting-Juan Zhang
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, 212002, China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, 212002, China
| | - Liu-Chao Zhang
- Medical Laboratory, The Affiliated Qidong Hospital of Nantong University Qidong People's Hospital Qidong Liver Cancer Institute, Qidong, Jiangsu, 226200, China
| | - Zi-Jun Xu
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, 212002, China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, 212002, China
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Ming-Qiang Chu
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, 212002, China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, 212002, China
| | - Yang-Jing Zhao
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, 212002, China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, 212002, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jiang Lin
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, 212002, China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, 212002, China
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Jun Qian
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212002, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, 212002, China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, 212002, China
| | - Jing-Dong Zhou
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China.
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212002, China.
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, 212002, China.
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, 212002, China.
| |
Collapse
|
8
|
You Y, Guo Z, Wolter T, Hu Q. Intracellular metal ion-based chemistry for programmed cell death. Chem Soc Rev 2025; 54:1552-1582. [PMID: 39744985 DOI: 10.1039/d4cs00930d] [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: 02/04/2025]
Abstract
Intracellular metal ions play essential roles in multiple physiological processes, including catalytic action, diverse cellular processes, intracellular signaling, and electron transfer. It is crucial to maintain intracellular metal ion homeostasis which is achieved by the subtle balance of storage and release of metal ions intracellularly along with the influx and efflux of metal ions at the interface of the cell membrane. Dysregulation of intracellular metal ions has been identified as a key mechanism in triggering programmed cell death (PCD). Despite the importance of metal ions in initiating PCD, the molecular mechanisms of intracellular metal ions within these processes are infrequently discussed. An in-depth understanding and review of the role of metal ions in triggering PCD may better uncover novel tools for cancer diagnosis and therapy. Specifically, the essential roles of calcium (Ca2+), iron (Fe2+/3+), copper (Cu+/2+), and zinc (Zn2+) ions in triggering PCD are primarily explored in this review, and other ions like manganese (Mn2+/3+/4+), cobalt (Co2+/3+) and magnesium ions (Mg2+) are briefly discussed. Further, this review elaborates on the underlying chemical mechanisms and summarizes these metal ions triggering PCD in cancer therapy. This review bridges chemistry, immunology, and biology to foster the rational regulation of metal ions to induce PCD for cancer therapy.
Collapse
Affiliation(s)
- Yawen You
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zhaochen Guo
- Department of Biochemistry, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Tyler Wolter
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Institute for Clinical and Translational Research, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
9
|
Taha SR, Karimi M, Mahdavi B, Yousefi Tehrani M, Bemani A, Kabirian S, Mohammadi J, Jabbari S, Hushmand M, Mokhtar A, Pourhanifeh MH. Crosstalk between non-coding RNAs and programmed cell death in colorectal cancer: implications for targeted therapy. Epigenetics Chromatin 2025; 18:3. [PMID: 39810224 PMCID: PMC11734566 DOI: 10.1186/s13072-024-00560-8] [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: 09/17/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the most common causes of cancer-related mortality worldwide. Its progression is influenced by complex interactions involving genetic, epigenetic, and environmental factors. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have been identified as key regulators of gene expression, affecting diverse biological processes, notably programmed cell death (PCD). OBJECTIVE This review aims to explore the relationship between ncRNAs and PCD in CRC, focusing on how ncRNAs influence cancer cell survival, proliferation, and treatment resistance. METHODS A comprehensive literature analysis was conducted to examine recent findings on the role of ncRNAs in modulating various PCD mechanisms, including apoptosis, autophagy, necroptosis, and pyroptosis, and their impact on CRC development and therapeutic response. RESULTS ncRNAs were found to significantly regulate PCD pathways, impacting tumor growth, metastasis, and treatment sensitivity in CRC. Their influence on these pathways highlights the potential of ncRNAs as biomarkers for early CRC detection and as targets for innovative therapeutic interventions. CONCLUSION Understanding the involvement of ncRNAs in PCD regulation offers new insights into CRC biology. The targeted modulation of ncRNA-PCD interactions presents promising avenues for personalized cancer treatment, which may improve patient outcomes by enhancing therapeutic effectiveness and reducing resistance.
Collapse
Affiliation(s)
- Seyed Reza Taha
- Department of Pathology and Immunology, Washington University School of Medicine, St. LouisWashington, MO, USA
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kiev, Ukraine.
| | - Bahar Mahdavi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Ali Bemani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahriar Kabirian
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Mohammadi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sina Jabbari
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Meysam Hushmand
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mokhtar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- PAKAN Institute, Tehran, Iran.
| |
Collapse
|
10
|
Wen D, Yan R, Zhang L, Zhang H, Chen X, Zhou J. Screening of necroptosis-related genes and evaluating the prognostic capacity, clinical value, and the effect of their copy number variations in acute myeloid leukemia. BMC Cancer 2025; 25:71. [PMID: 39806277 PMCID: PMC11727709 DOI: 10.1186/s12885-025-13439-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: 04/23/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is an aggressive hematological neoplasm. Little improvement in survival rates has been achieved over the past few decades. Necroptosis has relationship with certain types of malignancies outcomes. Here, we evaluated the diagnostic ability, prognostic capacity of necroptosis-related genes (NRGs) and the effect of their copy number variations (CNVs) in AML. METHODS Necroptosis-related differentially expressed genes (NRDEGs) were identified after intersecting differentially expressed genes (DEGs) from the Gene Expression Omnibus(GEO) database with NRGs from GeneCards, the Molecular Signatures Database (MSigDB) and literatures. Machine learning was applied to obtain hub-NRDEGs. The expression levels of the hub-NRDEGs were validated in vitro. The mRNA-miRNA and mRNA-TF interaction networks with the hub-NRDEGs were screened using Cytoscape@. Single-sample gene set enrichment analysis (ssGSEA) was utilized to calculate correlations between the hub-NRDEGs and immune cells. CNV analysis of the hub-NRDEGs was carried out on the TCGA-LAML datasets from the TCGA database. Kaplan-Meier (K-M) survival analyses were utilized to evaluate the prognostic values along with Cox model. RESULTS Six hub-NRDEGs (SLC25A5, PARP1, CTSS, ZNF217, NFKB1, and PYGL) were obtained and their expression changes derived from CNVs in AML were visualized. In total, 65 mRNA-miRNA and 80 mRNA-TF interaction networks with hub-NRDEGs were screened. The ssGSEA result showed the expression of RAPR1 was inversely related to CD56dim natural killer cells and the expression of CTSS was positive related to Myeloid-derived suppressor cells (MDSCs) in AML. The K-M results demonstrated that ZNF217 had significant difference in the duration of survival in AML patients. Cox regression models revealed that the hub-NRDEGs had better predictive power at year-1 and year-5. CONCLUSION These screened NRDEGs can be exploited as clinical prognostic predictions in AML patients, as well as potential biomarkers for diagnosis and therapeutic targeting.
Collapse
Affiliation(s)
- Dake Wen
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, China
| | - Ru Yan
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, China
| | - Lin Zhang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, China
| | - Haoyang Zhang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, China
| | - Xuyang Chen
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, China
| | - Jian Zhou
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, China.
- Department of Pediatric Laboratory, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, 299-1, QingYang Road, Wuxi, 214023, China.
| |
Collapse
|
11
|
Zhang L, Yu Y, Li G, Li J, Ma X, Ren J, Liu N, Guo S, Li J, Cai J. Identification of PANoptosis-based signature for predicting the prognosis and immunotherapy response in AML. Heliyon 2024; 10:e40267. [PMID: 39634422 PMCID: PMC11616514 DOI: 10.1016/j.heliyon.2024.e40267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/21/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Background In recent years, the incidence of acute myeloid leukemia (AML) has increased rapidly with a suboptimal prognosis. In AML, cell death is independent of tumorigenesis, tumor invasion, and drug resistance. PANoptosis is a newly discovered form of cell death that combines pyroptosis, apoptosis, and necroptosis. However, no studies have explored the role of PANoptosis-based signatures in AML. Methods We screened for PANoptosis-related genes and established a PANoptosis-risk signature using the least absolute shrinkage and selection operator (LASSO) and Cox regression analysis. We combined TCGA, bulk RNA sequencing, and single-cell sequencing to investigate the correlation between candidate genes and the AML tumor microenvironment. Results The PANoptosis risk signature effectively predicted prognosis with good sensitivity and specificity. The risk score emerged as an independent prognostic factor. Functional enrichment analysis of PANoptosis-related differentially expressed genes suggested that the risk score may be related to cell immunity. Patients with high-risk scores exhibited increased immune cell infiltration, implying a hot tumor immune microenvironment. The risk score was positively correlated with the immune scores and expression levels of immune checkpoints. Therefore, we identified three model factors, BIRC3, PELI1, and PRKACG, as predictors for immunotherapy efficacy. Single-cell sequencing analysis demonstrated that PELI1 and BIRC3 may participate in the regulation of the AML immune microenvironment. Finally, we performed a drug sensitivity analysis to target BIRC3 and PELI1 using molecular docking and molecular dynamics simulations. Conclusion Our study established and verified a PANoptosis risk signature to predict the survival and immunological treatment response in AML.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261031, China
| | - Yanan Yu
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261031, China
| | - Guiqing Li
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Jiachun Li
- Department of Information Engineering, Weifang Vocational College of Food Science and Technology, Weifang, 262100, Shandong, China
| | - Xiaolin Ma
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Jiao Ren
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261031, China
| | - Na Liu
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261031, China
| | - Songyue Guo
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261031, China
| | - Jiaqiu Li
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261031, China
| | - Jinwei Cai
- Department of Oncology, People's Hospital of Kecheng District, Quzhou, 324000, Zhejiang, China
| |
Collapse
|
12
|
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
|
13
|
Zhang Y, Chen D, Ang B, Deng X, Li B, Bai Y, Zhang Y. A necroptosis-regulated model from single-cell analysis that predicts survival and identifies the Pivotal role of MAGEA6 in hepatocellular carcinoma. Heliyon 2024; 10:e37711. [PMID: 39315163 PMCID: PMC11417173 DOI: 10.1016/j.heliyon.2024.e37711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Objective Hepatocellular carcinoma (HCC) ranks as the third leading cause of cancer-related deaths, constituting 75%-85 % of all primary liver cancers. The objective of this study was to develop a necroptosis-related gene signature using single-cell and bulk RNA sequencing to predict HCC patient prognoses. Methods A total of 25 key necroptosis regulators were identified from previous literature. We evaluated the necroptosis scores of different cell types using single-cell sequencing data from HCC and analyzed 168 necroptosis-related genes. The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) dataset served as the training set for establishing a novel necroptosis-related gene risk signature, employing univariate and multivariate Cox regression analyses. Additionally, the study examined the model's relevance in immunity and immunotherapy, and predicted chemosensitivity in HCC patients based on the gene signature. The key genes were validated by the biological experiments. Results Compared to other cell types, hepatoma cells displayed the lowest necroptosis scores. A new six-gene necroptosis-related signature (S100A11, MAGEC2, MAGEA6, CTP2C9, SOX4, AKR1B10) was developed using the TCGA database and validated in the ICGC database. Patients in the high-risk category had poorer prognoses, with the risk score serving as an independent prognostic indicator beyond other clinical factors. These high-risk patients also exhibited greater immune infiltration but demonstrated a weaker anti-tumor response due to elevated expression of immune checkpoints. Pathways involving hypoxia, glycolysis, and P53, as well as the frequency of P53 somatic mutations, were notably heightened in the high-risk group. Additionally, the six genes in the model showed significantly different mRNA expression in hepatoma cell lines compared to normal hepatocytes, with the role of MAGEA6 in liver cancer being elucidated through critical experiments. Conclusions This study successfully developed a six-gene necroptosis-related signature to predict prognoses in HCC patients. It further explored the roles of necroptosis in hepatoma cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Youcheng Zhang
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192,China
- Department of Pediatric Surgery, Huai’an Maternal and Child Health Care Center, Huai'an, 223001, Jiangsu Province, China
| | - Dapeng Chen
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192,China
| | - Bing Ang
- Department of Oncology, Tianjin First Central Hospital Clinic Institute, Tianjin 300192, China
| | - Xiyue Deng
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192,China
| | - Bing Li
- Department of Pediatric Surgery, Huai’an Maternal and Child Health Care Center, Huai'an, 223001, Jiangsu Province, China
| | - Yi Bai
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yamin Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| |
Collapse
|
14
|
Wang W, Liu Y, Wang Z, Tan X, Jian X, Zhang Z. Exploring and validating the necroptotic gene regulation and related lncRNA mechanisms in colon adenocarcinoma based on multi-dimensional data. Sci Rep 2024; 14:22251. [PMID: 39333335 PMCID: PMC11437100 DOI: 10.1038/s41598-024-73168-3] [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: 02/27/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
Necroptosis is intimately associated with the initiation and progression of colon adenocarcinoma (COAD). However, studies on necroptosis-related genes (NRGs) and the regulating long non-coding RNAs (NRGlncRNAs) in the context of COAD are limited. We retrieved the cancer genome atlas (TCGA) to collect datasets of NRGs and NRGlncRNAs on COAD patients. The risk model constructed using Cox and least absolute shrinkage and selection operator (LASSO) regression was then employed to identify NRGs and NRGlncRNAs with prognostic significance. Subsequently, we validated the results using gene expression omnibus (GEO) datasets from different populations, conducted Mendelian randomization (MR) analysis to explore the potential causal relationships between prognostic NRGs and COAD, and conducted cell experiments to verify the expression of prognostic NRGlncRNAs in COAD. Furthermore, we explored potential pathways and regulatory mechanisms of these prognostic NRGlncRNAs and NRGs in COAD through enrichment analysis, immune cell correlation analysis, tumor microenvironment analysis, immune checkpoint analysis, tumor sample clustering, and so on. We identified eight NRGlncRNAs (AC245100.5, AP001619.1, LINC01614, AC010463.3, AL162595.1, ITGB1-DT, LINC01857, and LINC00513) used for constructing the prognostic model and nine prognostic NRGs (AXL, BACH2, CFLAR, CYLD, IPMK, MAP3K7, ATRX, BRAF, and OTULIN) with regulatory relationships with them, and their validation was performed using GEO and GWAS datasets, as well as cell experiments, which showed largely consistent results. These prognostic NRGlncRNAs and NRGs modulate various biological functions, including immune inflammatory response, oxidative stress, immune escape, telomere regulation, and cytokine response, influencing the development of COAD. Additionally, stratified analysis of the high-risk and low-risk groups based on the prognostic model revealed elevated expression of immune cells, increased expression of tumor microenvironment cells, and upregulation of immune checkpoint gene expression in the high-risk group. Finally, through cluster analysis, we identified tumor subtypes, and the results of cluster analysis were essentially consistent with the analysis between risk groups. The prognostic NGRlncRNAs and NRGs identified in our study serve as prognostic indicators and potential therapeutic targets for COAD, providing a theoretical basis for the clinical diagnosis and treatment of COAD and offering guidance for further research.
Collapse
Affiliation(s)
- Weili Wang
- Department of Oncology, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ziqi Wang
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaoning Tan
- Department of Oncology, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China.
| | - Xiaolan Jian
- Department of Oncology, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China.
| | - Zhen Zhang
- Department of Oncology, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China.
| |
Collapse
|
15
|
Yu L, Huang K, Liao Y, Wang L, Sethi G, Ma Z. Targeting novel regulated cell death: Ferroptosis, pyroptosis and necroptosis in anti-PD-1/PD-L1 cancer immunotherapy. Cell Prolif 2024; 57:e13644. [PMID: 38594879 PMCID: PMC11294428 DOI: 10.1111/cpr.13644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/02/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
Chemotherapy, radiotherapy, and immunotherapy represent key tumour treatment strategies. Notably, immune checkpoint inhibitors (ICIs), particularly anti-programmed cell death 1 (PD1) and anti-programmed cell death ligand 1 (PD-L1), have shown clinical efficacy in clinical tumour immunotherapy. However, the limited effectiveness of ICIs is evident due to many cancers exhibiting poor responses to this treatment. An emerging avenue involves triggering non-apoptotic regulated cell death (RCD), a significant mechanism driving cancer cell death in diverse cancer treatments. Recent research demonstrates that combining RCD inducers with ICIs significantly enhances their antitumor efficacy across various cancer types. The use of anti-PD-1/PD-L1 immunotherapy activates CD8+ T cells, prompting the initiation of novel RCD forms, such as ferroptosis, pyroptosis, and necroptosis. However, the functions and mechanisms of non-apoptotic RCD in anti-PD1/PD-L1 therapy remain insufficiently explored. This review summarises the emerging roles of ferroptosis, pyroptosis, and necroptosis in anti-PD1/PD-L1 immunotherapy. It emphasises the synergy between nanomaterials and PD-1/PD-L1 inhibitors to induce non-apoptotic RCD in different cancer types. Furthermore, targeting cell death signalling pathways in combination with anti-PD1/PD-L1 therapies holds promise as a prospective immunotherapy strategy for tumour treatment.
Collapse
Affiliation(s)
- Li Yu
- Health Science CenterYangtze UniversityJingzhouHubeiChina
- Department of UrologyJingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze UniversityJingzhouHubeiChina
| | - Ke Huang
- Health Science CenterYangtze UniversityJingzhouHubeiChina
| | - Yixiang Liao
- Department of UrologyJingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze UniversityJingzhouHubeiChina
| | - Lingzhi Wang
- Department of PharmacologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Cancer Science Institute of Singapore, National University of SingaporeSingaporeSingapore
- NUS Centre for Cancer Research (N2CR), National University of SingaporeSingaporeSingapore
| | - Gautam Sethi
- Department of PharmacologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- NUS Centre for Cancer Research (N2CR), National University of SingaporeSingaporeSingapore
| | - Zhaowu Ma
- Health Science CenterYangtze UniversityJingzhouHubeiChina
| |
Collapse
|
16
|
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
|
17
|
Guo L, Ma X, Li H, Yan S, Zhang K, Li J. Single‑cell RNA‑seq necroptosis‑related genes predict the prognosis of breast cancer and affect the differentiation of CD4 + T cells in tumor immune microenvironment. Mol Clin Oncol 2024; 21:49. [PMID: 38872949 PMCID: PMC11170320 DOI: 10.3892/mco.2024.2747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/30/2024] [Indexed: 06/15/2024] Open
Abstract
Breast cancer (BC) is one of the most prevalent types of malignancy and a major cause of cancer-related death. The purpose of the present study was to identify prognostic models of necroptosis-related genes (NRGs) in BC at the single-cell RNA-sequencing level and reveal the role of NRGs in tumour immune microenvironment (TIME). A risk model was constructed based on Cox regression and LASSO methods. Next, high-scoring cell populations were searched through AUCell scores, and cell subtypes were then analyzed by pseudotime analysis. Finally, the expression level of the model genes was verified by reverse transcription-quantitative (RT-qPCR). A new prognostic model was constructed and validated based on five NRGs (BCL2, BIRC3, AIFM1, IFNG and VDAC1), which could effectively predict the prognosis of patients with BC. NRGs were found to be highly active in CD4+ T cells and differentially expressed in their developmental trajectories. Finally, the RT-qPCR results showed that most of the model genes were significantly overexpressed in MDA-MB-231 and MCF-7 cells (P<0.05). In conclusion, an NRG signature with excellent predictive properties in prognosis and TIME was successfully established. Moreover, NRGs were involved in the differentiation and development of CD4+ T cells in TIME. These findings provide potential therapeutic strategies for BC.
Collapse
Affiliation(s)
- Li Guo
- Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750003, P.R. China
| | - Xiuzhen Ma
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Hong Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Shuxun Yan
- Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750003, P.R. China
| | - Kai Zhang
- Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750003, P.R. China
| | - Jinping Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
18
|
Zhang Y, Zhou X. Targeting regulated cell death (RCD) in hematological malignancies: Recent advances and therapeutic potential. Biomed Pharmacother 2024; 175:116667. [PMID: 38703504 DOI: 10.1016/j.biopha.2024.116667] [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: 01/15/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024] Open
Abstract
Regulated cell death (RCD) is a form of cell death that can be regulated by numerous biomacromolecules. Accumulating evidence suggests that dysregulated expression and altered localization of related proteins in RCD promote the development of cancer. Targeting subroutines of RCD with pharmacological small-molecule compounds is becoming a promising therapeutic avenue for anti-tumor treatment, especially in hematological malignancies. Herein, we summarize the aberrant mechanisms of apoptosis, necroptosis, pyroptosis, PANoptosis, and ferroptosis in hematological malignancies. In particular, we focus on the relationship between cell death and tumorigenesis, anti-tumor immunotherapy, and drug resistance in hematological malignancies. Furthermore, we discuss the emerging therapeutic strategies targeting different RCD subroutines. This review aims to summarize the significance and potential mechanisms of RCD in hematological malignancies, along with the development and utilization of pertinent therapeutic strategies.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| |
Collapse
|
19
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [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: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
20
|
Arimoto KI, Miyauchi S, Liu M, Zhang DE. Emerging role of immunogenic cell death in cancer immunotherapy. Front Immunol 2024; 15:1390263. [PMID: 38799433 PMCID: PMC11116615 DOI: 10.3389/fimmu.2024.1390263] [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: 02/23/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer immunotherapy, such as immune checkpoint blockade (ICB), has emerged as a groundbreaking approach for effective cancer treatment. Despite its considerable potential, clinical studies have indicated that the current response rate to cancer immunotherapy is suboptimal, primarily attributed to low immunogenicity in certain types of malignant tumors. Immunogenic cell death (ICD) represents a form of regulated cell death (RCD) capable of enhancing tumor immunogenicity and activating tumor-specific innate and adaptive immune responses in immunocompetent hosts. Therefore, gaining a deeper understanding of ICD and its evolution is crucial for developing more effective cancer therapeutic strategies. This review focuses exclusively on both historical and recent discoveries related to ICD modes and their mechanistic insights, particularly within the context of cancer immunotherapy. Our recent findings are also highlighted, revealing a mode of ICD induction facilitated by atypical interferon (IFN)-stimulated genes (ISGs), including polo-like kinase 2 (PLK2), during hyperactive type I IFN signaling. The review concludes by discussing the therapeutic potential of ICD, with special attention to its relevance in both preclinical and clinical settings within the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Kei-ichiro Arimoto
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Sayuri Miyauchi
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Mengdan Liu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- School of Biological Sciences, University of California San Diego, La Jolla, CA, United States
| | - Dong-Er Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- School of Biological Sciences, University of California San Diego, La Jolla, CA, United States
- Department of Pathology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
21
|
Meier P, Legrand AJ, Adam D, Silke J. Immunogenic cell death in cancer: targeting necroptosis to induce antitumour immunity. Nat Rev Cancer 2024; 24:299-315. [PMID: 38454135 DOI: 10.1038/s41568-024-00674-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 03/09/2024]
Abstract
Most metastatic cancers remain incurable due to the emergence of apoptosis-resistant clones, fuelled by intratumour heterogeneity and tumour evolution. To improve treatment, therapies should not only kill cancer cells but also activate the immune system against the tumour to eliminate any residual cancer cells that survive treatment. While current cancer therapies rely heavily on apoptosis - a largely immunologically silent form of cell death - there is growing interest in harnessing immunogenic forms of cell death such as necroptosis. Unlike apoptosis, necroptosis generates second messengers that act on immune cells in the tumour microenvironment, alerting them of danger. This lytic form of cell death optimizes the provision of antigens and adjuvanticity for immune cells, potentially boosting anticancer treatment approaches by combining cellular suicide and immune response approaches. In this Review, we discuss the mechanisms of necroptosis and how it activates antigen-presenting cells, drives cross-priming of CD8+ T cells and induces antitumour immune responses. We also examine the opportunities and potential drawbacks of such strategies for exposing cancer cells to immunological attacks.
Collapse
Affiliation(s)
- Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK.
| | - Arnaud J Legrand
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| | - John Silke
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
| |
Collapse
|
22
|
Favale G, Donnarumma F, Capone V, Della Torre L, Beato A, Carannante D, Verrilli G, Nawaz A, Grimaldi F, De Simone MC, Del Gaudio N, Megchelenbrink WL, Caraglia M, Benedetti R, Altucci L, Carafa V. Deregulation of New Cell Death Mechanisms in Leukemia. Cancers (Basel) 2024; 16:1657. [PMID: 38730609 PMCID: PMC11083363 DOI: 10.3390/cancers16091657] [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: 04/04/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Hematological malignancies are among the top five most frequent forms of cancer in developed countries worldwide. Although the new therapeutic approaches have improved the quality and the life expectancy of patients, the high rate of recurrence and drug resistance are the main issues for counteracting blood disorders. Chemotherapy-resistant leukemic clones activate molecular processes for biological survival, preventing the activation of regulated cell death pathways, leading to cancer progression. In the past decade, leukemia research has predominantly centered around modulating the well-established processes of apoptosis (type I cell death) and autophagy (type II cell death). However, the development of therapy resistance and the adaptive nature of leukemic clones have rendered targeting these cell death pathways ineffective. The identification of novel cell death mechanisms, as categorized by the Nomenclature Committee on Cell Death (NCCD), has provided researchers with new tools to overcome survival mechanisms and activate alternative molecular pathways. This review aims to synthesize information on these recently discovered RCD mechanisms in the major types of leukemia, providing researchers with a comprehensive overview of cell death and its modulation.
Collapse
Affiliation(s)
- Gregorio Favale
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Federica Donnarumma
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Vincenza Capone
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Laura Della Torre
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Antonio Beato
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Daniela Carannante
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Giulia Verrilli
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Asmat Nawaz
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
- Biogem, Molecular Biology and Genetics Research Institute, 83031 Ariano Irpino, Italy
| | - Francesco Grimaldi
- Dipartimento di Medicina Clinica e Chirurgia, Divisione di Ematologia, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | | | - Nunzio Del Gaudio
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Wouter Leonard Megchelenbrink
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Michele Caraglia
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
- Biogem, Molecular Biology and Genetics Research Institute, 83031 Ariano Irpino, Italy
| | - Rosaria Benedetti
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
- Biogem, Molecular Biology and Genetics Research Institute, 83031 Ariano Irpino, Italy
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS)-National Research Council (CNR), 80131 Napoli, Italy
- Programma di Epigenetica Medica, A.O.U. “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Vincenzo Carafa
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.F.); (F.D.); (V.C.); (L.D.T.); (A.B.); (D.C.); (G.V.); (A.N.); (N.D.G.); (W.L.M.); (M.C.); (R.B.); (L.A.)
- Biogem, Molecular Biology and Genetics Research Institute, 83031 Ariano Irpino, Italy
| |
Collapse
|
23
|
Lawlor KE, Murphy JM, Vince JE. Gasdermin and MLKL necrotic cell death effectors: Signaling and diseases. Immunity 2024; 57:429-445. [PMID: 38479360 DOI: 10.1016/j.immuni.2024.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/29/2023] [Accepted: 02/14/2024] [Indexed: 01/22/2025]
Abstract
Diverse inflammatory conditions, from infections to autoimmune disease, are often associated with cellular damage and death. Apoptotic cell death has evolved to minimize its inflammatory potential. By contrast, necrotic cell death via necroptosis and pyroptosis-driven by membrane-damaging MLKL and gasdermins, respectively-can both initiate and propagate inflammatory responses. In this review, we provide insights into the function and regulation of MLKL and gasdermin necrotic effector proteins and drivers of plasma membrane rupture. We evaluate genetic evidence that MLKL- and gasdermin-driven necrosis may either provide protection against, or contribute to, disease states in a context-dependent manner. These cumulative insights using gene-targeted mice underscore the necessity for future research examining pyroptotic and necroptotic cell death in human tissue, as a basis for developing specific necrotic inhibitors with the potential to benefit a spectrum of pathological conditions.
Collapse
Affiliation(s)
- Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| | - James M Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
24
|
Luo L, Feng F, Zhong A, Guo N, He J, Li C. The advancement of polysaccharides in disease modulation: Multifaceted regulation of programmed cell death. Int J Biol Macromol 2024; 261:129669. [PMID: 38272424 DOI: 10.1016/j.ijbiomac.2024.129669] [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: 10/26/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024]
Abstract
Programmed cell death (PCD), also known as regulatory cell death (RCD), is a process that occurs in all organisms and is closely linked to both normal physiological processes and disease states. Various signaling pathways, such as TP53, KRAS, NOTCH, hypoxia, and metabolic reprogramming, have been found to regulate RCD. Polysaccharides, which are essential natural products, have been the subject of extensive research in the fields of food, nutrition, and medicine due to their wide range of pharmacological effects. Studies have shown that polysaccharides have biological activities and the potential to target signal transduction pathways for the treatment of diseases. This paper provides a review of the mechanisms through which polysaccharides exert their therapeutic effects at different levels and explores the relationship between different types of RCD and human diseases. The aim of this review is to provide a theoretical basis for the further clinical use and application of polysaccharide bioactivities.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| | - Fuhai Feng
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Ai Zhong
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Nuoqing Guo
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Jiake He
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Chenying Li
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| |
Collapse
|
25
|
Arroyo Villora S, Castellanos Silva P, Zenz T, Kwon JS, Schlaudraff N, Nitaj D, Meckbach C, Dammann R, Richter AM. Biomarker RIPK3 Is Silenced by Hypermethylation in Melanoma and Epigenetic Editing Reestablishes Its Tumor Suppressor Function. Genes (Basel) 2024; 15:175. [PMID: 38397165 PMCID: PMC10888250 DOI: 10.3390/genes15020175] [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: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
For several decades, cancers have demonstrably been one of the most frequent causes of death worldwide. In addition to genetic causes, cancer can also be caused by epigenetic gene modifications. Frequently, tumor suppressor genes are epigenetically inactivated due to hypermethylation of their CpG islands, actively contributing to tumorigenesis. Since CpG islands are usually localized near promoters, hypermethylation of the promoter can have a major impact on gene expression. In this study, the potential tumor suppressor gene Receptor Interacting Serine/Threonine Protein Kinase 3 (RIPK3) was examined for an epigenetic regulation and its gene inactivation in melanomas. A hypermethylation of the RIPK3 CpG island was detected by bisulfite pyrosequencing and was accompanied by a correlated loss of its expression. In addition, an increasing RIPK3 methylation rate was observed with increasing tumor stage of melanomas. For further epigenetic characterization of RIPK3, epigenetic modulation was performed using a modified CRISPR/dCas9 (CRISPRa activation) system targeting its DNA hypermethylation. We observed a reduced fitness of melanoma cells by (re-)expression and demethylation of the RIPK3 gene using the epigenetic editing-based method. The tumor suppressive function of RIPK3 was evident by phenotypic determination using fluorescence microscopy, flow cytometry and wound healing assay. Our data highlight the function of RIPK3 as an epigenetically regulated tumor suppressor in melanoma, allowing it to be classified as a biomarker.
Collapse
Affiliation(s)
- Sarah Arroyo Villora
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | | | - Tamara Zenz
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Ji Sun Kwon
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
- Department of Mathematics, Natural Sciences and Computer Science, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany
| | - Nico Schlaudraff
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Dafina Nitaj
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Cornelia Meckbach
- Department of Mathematics, Natural Sciences and Computer Science, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany
| | - Reinhard Dammann
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Antje M. Richter
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| |
Collapse
|
26
|
Li X, Sun Z, Ma J, Yang M, Cao H, Jiao G. Identification of TNFRSF21 as an inhibitory factor of osteosarcoma based on a necroptosis-related prognostic gene signature and molecular experiments. Cancer Cell Int 2024; 24:14. [PMID: 38184626 PMCID: PMC10770912 DOI: 10.1186/s12935-023-03198-w] [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: 10/23/2023] [Accepted: 12/26/2023] [Indexed: 01/08/2024] Open
Abstract
BACKGROUND Osteosarcoma is one of the most common malignant bone tumors with bad prognosis. Necroptosis is a form of programmed cell death. Recent studies showed that targeting necroptosis was a new promising approach for tumor therapy. This study aimed to establish a necroptosis-related gene signature to evaluated prognosis and explore the relationship between necroptosis and osteosarcoma. METHODS Data from The Cancer Genome Atlas was used for developing the signature and the derived necroptosis score (NS). Data from Gene Expression Omnibus served as validation. Principal component analysis (PCA), Cox regression, receiver operating characteristic (ROC) curves and Kaplan-Meier survival analysis were used to assess the performance of signature. The association between the NS and osteosarcoma was analyzed via gene set enrichment analysis, gene set variation analysis and Pearson test. Single-cell data was used for further exploration. Among the genes that constituted the signature, the role of TNFRSF21 in osteosarcoma was unclear. Molecular experiments were used to explore TNFRSF21 function. RESULTS Our data revealed that lower NS indicated more active necroptosis in osteosarcoma. Patients with lower NS had a better prognosis. PCA and ROC curves demonstrated NS was effective to predict prognosis. NS was negatively associated with immune infiltration levels and tumor microenvironment scores and positively associated with tumor purity and stemness index. Single-cell data showed necroptosis heterogeneity in osteosarcoma. The cell communication pattern of malignant cells with high NS was positively correlated with tumor progression. The expression of TNFRSF21 was down-regulated in osteosarcoma cell lines. Overexpression of TNFRSF21 inhibited proliferation and motility of osteosarcoma cells. Mechanically, TNFRSF21 upregulated the phosphorylation levels of RIPK1, RIPK3 and MLKL to promote necroptosis in osteosarcoma. CONCLUSIONS The necroptosis prognostic signature and NS established in this study could be used as an independent prognostic factor, TNFRSF21 may be a necroptosis target in osteosarcoma therapy.
Collapse
Affiliation(s)
- Xiang Li
- Department of Orthopedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, 250000, Shandong Province, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Zhenqian Sun
- Department of Orthopedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, 250000, Shandong Province, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Jinlong Ma
- Department of Orthopedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, 250000, Shandong Province, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Miaomiao Yang
- Department of Oncology, Yantai Yuhuangding Hospital, Yantai, Shandong Province, China
| | - Hongxin Cao
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Guangjun Jiao
- Department of Orthopedics, Qilu Hospital of Shandong University, No.107, Wenhuaxi Road, Lixia District, Jinan, 250000, Shandong Province, China.
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
27
|
Workenhe ST, Inkol JM, Westerveld MJ, Verburg SG, Worfolk SM, Walsh SR, Kallio KL. Determinants for Antitumor and Protumor Effects of Programmed Cell Death. Cancer Immunol Res 2024; 12:7-16. [PMID: 37902605 PMCID: PMC10762341 DOI: 10.1158/2326-6066.cir-23-0321] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/30/2023] [Accepted: 09/14/2023] [Indexed: 10/31/2023]
Abstract
Cytotoxic anticancer therapies activate programmed cell death in the context of underlying stress and inflammatory signaling to elicit the emission of danger signals, cytokines, and chemokines. In a concerted manner, these immunomodulatory secretomes stimulate antigen presentation and T cell-mediated anticancer immune responses. In some instances, cell death-associated secretomes attract immunosuppressive cells to promote tumor progression. As it stands, cancer cell death-induced changes in the tumor microenvironment that contribute to antitumor or protumor effects remain largely unknown. This is complicated to examine because cell death is often subverted by tumors to circumvent natural, and therapy-induced, immunosurveillance. Here, we provide insights into important but understudied aspects of assessing the contribution of cell death to tumor elimination or cancer progression, including the role of tumor-associated genetics, epigenetics, and oncogenic factors in subverting immunogenic cell death. This perspective will also provide insights on how future studies may address the complex antitumor and protumor immunologic effects of cell death, while accounting for variations in tumor genetics and underlying microenvironment.
Collapse
Affiliation(s)
- Samuel T. Workenhe
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jordon M. Inkol
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Michael J. Westerveld
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Shayla G. Verburg
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Sarah M. Worfolk
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Scott R. Walsh
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Kaslyn L.F. Kallio
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
28
|
Xu T, Jiang J, Xiang X, Jahanshahi H, Zhang Y, Chen X, Li L. Conduction and validation of a novel prognostic signature in cervical cancer based on the necroptosis characteristic genes via integrating of multiomics data. Comput Biol Med 2024; 168:107656. [PMID: 38029530 DOI: 10.1016/j.compbiomed.2023.107656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/12/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023]
Abstract
The significance of necroptosis in recurrent or metastatic cervical cancer remains unclear. In this study, we utilized various bioinformatics methods to analyze the cancer genome atlas (TCGA) data, gene chip and the single-cell RNA-sequencing (scRNA seq) data. And a necroptosis-related genes signature for prognostic assessment of patients with cervical cancer was constructed successfully. Survival analysis, receiver operating characteristic (ROC) curve, the support vector machine recursive feature elimination (SVM-RFE) algorithm and random forest analysis were performed to validate this signature. Patients in TCGA-CESC cohort were grouped into "high-necroptosis score (H-NCPS)" vs "low-necroptosis score (L-NCPS)" subgroups based on the median of necroptosis score of each patient. Analyses of the tumor microenvironment manifested "H-NCPS" patients associated with lower degree of immune infiltration. Through the utilization of survival analysis, cell communication, and Gene Set Enrichment Analysis (GSEA), PGK1 was determined to be the pivotal gene within the 9-gene signature associated with necroptosis. The high expression of PGK1 in cervical cancer cells was confirmed through the utilization of quantitative real-time polymerase chain reaction (RT-qPCR) and the human protein atlas (HPA). In the interim, PGK1 prompted the transition of M1 macrophages to M2 macrophages and influenced the occurrence and development of necroptosis. In conclusion, the 9-gene signature developed from necroptosis-related genes has shown significant predictive capabilities for the prognosis of cervical cancer, offered valuable guidance for individualized and targeted treatment approaches for patients.
Collapse
Affiliation(s)
- Tu Xu
- Department of Clinical Medicine, Medical College of Hunan Normal University, Changsha, Hunan, China.
| | - Jingwen Jiang
- Department of Clinical Medicine, Medical College of Hunan Normal University, Changsha, Hunan, China.
| | - Xiaoqing Xiang
- Department of Clinical Medicine, Medical College of Hunan Normal University, Changsha, Hunan, China.
| | - Hadi Jahanshahi
- Institute of Electrical and Electronics Engineers, Toronto, ON, Canada.
| | - Yong Zhang
- Department of Clinical Medicine, Medical College of Hunan Normal University, Changsha, Hunan, China.
| | - Xiaoyan Chen
- Department of Pathology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| | - Lesai Li
- Department of Gynecologic Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
29
|
Edlinger L. In Vivo Modeling of Leukemia by Murine Stem Cell Transplantation. Methods Mol Biol 2024; 2773:1-7. [PMID: 38236531 DOI: 10.1007/978-1-0716-3714-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Murine stem cell transplantation is a well-established method for the in vivo study of leukemic pathophysiology. Adoptive transfer of murine leukemic cells into lethally irradiated recipient mice leads to reconstitution of the hematopoietic system with malignant cells and eventually to leukemic progression in the recipient mice. Here, we describe the detailed protocol of the production of retroviral particles carrying the leukemic oncogene of interest as well as the isolation, retroviral transduction, and adoptive transfer of murine bone marrow cells.
Collapse
Affiliation(s)
- Leo Edlinger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| |
Collapse
|
30
|
Ji L, Liang S, Cheng Y, Gao R, Yan W, Pang F, Zhang F. Identification of a novel necroptosis-related LncRNA signature for prognostic prediction and immune response in oral squamous cell carcinoma. Cancer Biomark 2024; 40:319-342. [PMID: 39213052 PMCID: PMC11380221 DOI: 10.3233/cbm-230407] [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] [Indexed: 09/04/2024]
Abstract
BACKGROUND Necroptosis is a caspase-independent regulated necrotic cell death modality that elicits strong adaptive immune responses, and has the potential to activate antitumor immunity. Long non-coding RNAs (lncRNAs) have critical effects on oral squamous cell carcinoma (OSCC), which are closely associated with the prognosis and immune regulation of OSCC patients. OBJECTIVE This study aimed to identify a novel necroptosis-related lncRNAs signature to predict the prognosis and immune response of OSCC patients and provide patients with anti-tumor drug selection through bioinformatics analysis and in vitro experiments. METHODS A series of analyses, including differential lncRNA screening, survival analysis, Cox regression analysis, ROC analysis, nomogram prediction, enrichment analysis, tumor-infiltrating immune cells, drug sensitivity analysis, and consensus cluster analysis, were performed to determine and validate the prognostic value of necroptosis-associated lncRNAs signature in OSCC. And real-time quantitative polymerase chain reaction (RT-qPCR) was used to determine the expression levels of these lncRNAs. RESULTS This signature including 5 lncRNAs (AC099850.3, StarD4-AS1, AC011978.1, LINC01503, CDKN2A-DT) in OSCC associated with necroptosis were established and verified by bioinformatics. Further, ROC, K-M, univariate/multivariate Cox regression, and nomogram analysis were used to evaluate the model's features for OSCC prognosis. Using multiple bioinformatics techniques, the levels of tumor-infiltrating immune cells, immune checkpoints and semi-inhibitory concentrations showed significant differences across risk subtypes. By consensus cluster analysis, there were significant differences between clusters in survival, immune checkpoint expression, clinicopathological correlation, and tumor immunity. RT-qPCR showed that AC099850.3, AC011978.1, LINC01503 were up-regulated, STARD4-AS1 and CDKN2A-DT were down-regulated in OSCC cell lines compared with human normal oral keratinoid cell line. CONCLUSION We established 5-NRLs markers, which is useful for assessing OSCC immune response and prognosis, recommending personalized antitumor drugs. The expression level of 5-NRLs in OSCC was identified in vitro, and the results preliminarily verified this model. And this study would generate new insights for future experimental research.
Collapse
Affiliation(s)
- Lanting Ji
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Shuang Liang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | | | - Ruifang Gao
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Wenpeng Yan
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Fang Pang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Fang Zhang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| |
Collapse
|
31
|
Zhang G, Wang L, Zhao L, Yang F, Lu C, Yan J, Zhang S, Wang H, Li Y. Silibinin Induces Both Apoptosis and Necroptosis with Potential Anti-tumor Efficacy in Lung Cancer. Anticancer Agents Med Chem 2024; 24:1327-1338. [PMID: 39069713 DOI: 10.2174/0118715206295371240724092314] [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: 02/22/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The incidence of lung cancer is steadily on the rise, posing a growing threat to human health. The search for therapeutic drugs from natural active substances and elucidating their mechanism have been the focus of anti-tumor research. OBJECTIVE Silibinin (SiL) has been shown to be a natural product with a wide range of pharmacological activities, including anti-tumour activity. In our work, SiL was chosen as a possible substance that could inhibit lung cancer. Moreover, its effects on inducing tumor cell death were also studied. METHODS CCK-8 analysis and morphological observation were used to assess the cytotoxic impacts of SiL on lung cancer cells in vitro. The alterations in mitochondrial membrane potential (MMP) and apoptosis rate of cells were detected by flow cytometry. The level of lactate dehydrogenase (LDH) release out of cells was measured. The expression changes of apoptosis or necroptosis-related proteins were detected using western blotting. Protein interactions among RIPK1, RIPK3, and MLKL were analyzed using the co-immunoprecipitation (co-IP) technique. Necrosulfonamide (Nec, an MLKL inhibitor) was used to carry out experiments to assess the changes in apoptosis following the blockade of cell necroptosis. in vitro, SiL was evaluated for its antitumor effects using LLC tumor-bearing mice with mouse lung cancer. RESULTS With an increased dose of SiL, the proliferation ability of A549 cells was considerably inhibited, and the accompanying cell morphology changed. The results of flow cytometry showed that after SiL treatment, MMP levels decreased, and the proportion of cells undergoing apoptosis increased. There was an increase in cleaved caspase-9, caspase-3, and PARP, with a down-regulation of Bcl-2 and an up-regulation of Bax. In addition, the amount of LDH released from the cells increased following SiL treatment, accompanied by augmented expression and phosphorylation levels of necroptosis-related proteins (MLKL, RIPK1, and RIPK3), and the co-IP assay further confirmed the interactions among these three proteins, indicating the necrosome formation induced by SiL. Furthermore, Nec increased the apoptotic rate of SiL-treated cells and aggravated the cytotoxic effect of SiL, indicating that necroptosis blockade could switch cell death to apoptosis and increase the inhibitory effect of SiL on A549 cells. In LLC-bearing mice, gastric administration of SiL significantly inhibited tumor growth, and H&E staining showed significant damage to the tumour tissue. The results of the IHC showed that the expression of RIPK1, RIPK3, and MLKL was more pronounced in the tumor tissue. CONCLUSIONS This study confirmed the dual effect of SiL, as it can induce both biological processes, apoptosis and necroptosis, in lung cancer. SiL-induced apoptosis involved the mitochondrial pathway, as indicated by changes in caspase-9, Bcl-2, and Bax. Necroptosis may be activated due to the changes in the expression of associated proteins in tumour cells and tissues. It has been observed that blocking necroptosis by SiL increased cell death efficiency. This study helps clarify the anti-tumor mechanism of SiL against lung cancer, elucidating its role in the dual induction of apoptosis and necroptosis. Our work provides an experimental basis for the research on cell death induced by SiL and reveals its possible applications for improving the management of lung cancer.
Collapse
Affiliation(s)
- Guoqing Zhang
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, P.R. China
| | - Li Wang
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, P.R. China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, P.R. China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, P.R. China
| | - Limei Zhao
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, P.R. China
| | - Fang Yang
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, P.R. China
| | - Chunhua Lu
- Medical Experimental Center, The First People's Hospital of Nanning, Nanning, Guangxi, 530021, P.R. China
| | - Jianhua Yan
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, P.R. China
| | - Song Zhang
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei, 430070, P.R. China
| | - Haiping Wang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, P.R. China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, Hubei, 430056, P.R. China
| | - Yixiang Li
- Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, P.R. China
| |
Collapse
|
32
|
Huang J, Xu Z, Chen D, Zhou C, Shen Y. Pancancer analysis reveals the role of disulfidptosis in predicting prognosis, immune infiltration and immunotherapy response in tumors. Medicine (Baltimore) 2023; 102:e36830. [PMID: 38206694 PMCID: PMC10754585 DOI: 10.1097/md.0000000000036830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024] Open
Abstract
Disulfidptosis has been reported as a novel cell death process, suggesting a therapeutic strategy for cancer treatment. Herein, we constructed a multiomics data analysis to reveal the effects of disulfidptosis in tumors. Data for 33 kinds of tumors were downloaded from UCSC Xene, and disulfidptosis-related genes (DRGs) were selected from a previous study. After finishing processing data by the R packages, the expression and coexpression of DRGs in different tumors were assessed as well as copy number variations. The interaction network was drawn by STRING, and the activity of disulfidptosis was compared to the single-sample gene set enrichment analysis algorithm. Subsequently, the differences in DRGs for prognosis and clinicopathological features were evaluated, and the tumor immune microenvironment was assessed by the TIMER and TISCH databases. Tumor mutation burden, stem cell features and microsatellite instability were applied to predict drug resistance, and the expression of checkpoints was identified for the prediction of immunotherapy. Moreover, the TCIA, CellMiner and Enrichr databases were also utilized for selecting potential agents. Ten DRGs were differentially expressed in tumors, and the plots of coexpression and interaction revealed their correlation. Survival analysis suggested SLC7A11 as the most prognosis-related DRG with the most significant results. Additionally, the comparison also reflected the differences in DRGs in the status of pathologic lymph node metastasis for 5 types of tumors. The tumor immune microenvironment showed commonality among tumors based on immune infiltration and single-cell sequencing, and the analysis of tumor mutation burden, stemness and microsatellite instability showed a mostly positive correlation with DRGs. Moreover, referring to the prediction about clinical treatment, most DRGs can enhance sensitivity to chemotherapeutic agents but decrease the response to immune inhibitors with increasing expression. In this study, a primarily synthetic landscape of disulfidptosis in tumors was established and provided guidance for further exploration and investigation.
Collapse
Affiliation(s)
- Juntao Huang
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Ziqian Xu
- Department of Dermatology, Ningbo First Hospital, Zhejiang University, Zhejiang, China
| | - Dahua Chen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Chongchang Zhou
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yi Shen
- Centre for Medical Research, Ningbo No.2 Hospital, Ningbo, China
- School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
33
|
Wei K, Zhang X, Yang D. Identification and validation of prognostic and tumor microenvironment characteristics of necroptosis index and BIRC3 in clear cell renal cell carcinoma. PeerJ 2023; 11:e16643. [PMID: 38130918 PMCID: PMC10734432 DOI: 10.7717/peerj.16643] [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: 07/03/2023] [Accepted: 11/19/2023] [Indexed: 12/23/2023] Open
Abstract
Background Necroptosis is a form of programmed cell death; it has an important role in tumorigenesis and metastasis. However, details of the regulation and function of necroptosis in clear cell renal cell carcinoma (ccRCC) remain unclear. It is necessary to explore the significance of necroptosis in ccRCC. Methods Necroptosis-related clusters were discerned through the application of Consensus Clustering. Based on the TCGA and GEO databases, we identified prognostic necroptosis-related genes (NRGs) with univariate COX regression analysis. The necroptosis-related model was constructed through the utilization of LASSO regression analysis, and the immune properties, tumor mutation burden, and immunotherapy characteristics of the model were assessed using multiple algorithms and datasets. Furthermore, we conducted comprehensive GO, KEGG, and GSVA analyses to probe into the functional aspects of biological pathways. To explore the expression and of hub gene (BIRC3) in different ccRCC cell types and cell lines, single-cell sequencing data was analysed and we performed Quantitative Real-time PCR to detect the expression of BIRC3 in ccRCC cell lines. Function of BIRC3 in ccRCC was assessed through Cell Counting Kit-8 (CCK8) assay (for proliferation), transwell and wound healing assays (for migration and invasion). Results Distinct necroptosis-related clusters exhibiting varying prognostic implications, and enrichment pathways were identified in ccRCC. A robust necroptosis-related model formulated based on the expression of six prognostic NRGs, presented substantial predictive capabilities of overall survival and was shown to be related with patients' immune profiles, tumor mutation burden, and response to immunotherapy. Notably, the hub gene BIRC3 was markedly upregulated in both ccRCC tissues and cell lines, and showed significant correlations with immunosuppressive cells, immune checkpoints, and oncogenic pathways. Downregulation of BIRC3 demonstrated a negative regulatory effect on ccRCC cell proliferation migration and invasion. Conclusion The necroptosis-related model assumed a pivotal role in determining the prognosis, tumor mutation burden, immunotherapy response, and immune cell infiltration characteristics among ccRCC patients. BIRC3 exhibited significant correlations with the immunosuppressive microenvironment, which highlighted its potential for informing the design of innovative immunotherapies for ccRCC patients.
Collapse
Affiliation(s)
- Kai Wei
- Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xi Zhang
- Urology, The State Key Lab of Reproductive; The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongrong Yang
- Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
34
|
Krishnan RP, Pandiar D, Ramani P, Jayaraman S. Necroptosis in human cancers with special emphasis on oral squamous cell carcinoma. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2023; 124:101565. [PMID: 37459966 DOI: 10.1016/j.jormas.2023.101565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/12/2023] [Indexed: 11/06/2023]
Abstract
Necroptosis is a type of caspase independent 'programmed or regulated' necrotic cell death that has a morphological resemblance to necrosis and mechanistic analogy to apoptosis. This type of cell death requires RIPK1, RIPK3, MLKL, death receptors, toll like receptors, interferons, and various other proteins. Necroptosis is implicated in plethora of diseases like rheumatoid arthritis, Alzheimer's disease, Crohn's disease, and head and neck cancers including oral squamous cell carcinoma. Oral carcinomas show dysregulation or mutation of necroptotic proteins, mediate antitumoral immunity, activate immune response and control tumor progression. Necroptosis is known to play a dual role (pro tumorigenic and anti-tumorigenic) in cancer progression and targeting this pathway could be an effective approach in cancer therapy. Necroptosis based chemotherapy has been proposed in malignancies, highlighting the importance of necroptotic pathway to overcome apoptosis resistance and serve as a "fail-safe" pathway to modulate cancer initiation, progression, and metastasis. However, there is dearth of information regarding the use of necroptotic cell death mechanism in the treatment of oral squamous cell carcinoma. In this review, we summarise molecular mechanism of necroptosis, and its protumorigenic and antitumorigenic role in cancers to shed light on the possible therapeutic significance of necroptosis in oral squamous cell carcinoma.
Collapse
Affiliation(s)
| | - Deepak Pandiar
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Chennai, Tamil Nadu.
| | - Pratibha Ramani
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Chennai, Tamil Nadu.
| | - Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Chennai, Tamil Nadu.
| |
Collapse
|
35
|
Zhang F, Qi C, Yao Z, Xu H, Zhou G, Li C, Xia H. Identification and validation of a novel necroptosis-related molecular signature to evaluate prognosis and immune features in breast cancer. Apoptosis 2023; 28:1628-1645. [PMID: 37787960 DOI: 10.1007/s10495-023-01887-5] [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] [Accepted: 08/22/2023] [Indexed: 10/04/2023]
Abstract
Necroptosis has been shown to play an important role in the development of tumors. However, the characteristics of the necroptosis-related subtypes and the associated immune cell infiltration in the tumor microenvironment (TME) of breast cancer (BRCA) remain unclear. In this study, we identified three clusters related to necroptosis using the expression patterns of necroptosis-relevant genes (NRGs), and found that these three clusters had different clinicopathological features, prognosis and immune cell infiltration in the TME. Cluster 2 was characterized by less infiltration of immune cells in the TME and was associated with a worse prognosis. Then, a necroptosis risk score (NRS) composed of 14 NRGs was constructed using the least absolute shrinkage and selection operator regression (LASSO) Cox regression method. Based on NRS, all BRCA patients in the TCGA datasets were classified into a low-risk group and a high-risk group. Patients in the low-risk group were characterized by longer overall survival (OS), lower mutation burden, and higher infiltration level of immune cells in the TME. Moreover, the NRS was significantly associated with chemotherapeutic drug sensitivity. Finally, the knockdown of VDAC1 reduced the proliferation and migration of BRCA cells, and promoted cell death induced by necroptosis inducer. This study identified a novel necroptosis-related subtype of BRCA, and a comprehensive analysis of NRGs in BRCA revealed its potential roles in prognosis, clinicopathological features, TME, chemotherapy, tumor proliferation, and tumor necroptosis. These results may improve our understanding of NRGs in BRCA and provide a reference for developing individualized therapeutic strategies.
Collapse
Affiliation(s)
- Fan Zhang
- School of Chemistry and Chemical Engineering & Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, 210009, China
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing, 211166, China
| | - Chenxue Qi
- Department of Gynecologic Oncology, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zhipeng Yao
- School of Chemistry and Chemical Engineering & Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, 210009, China
| | - Haojun Xu
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing, 211166, China
| | - Guoren Zhou
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China.
| | - Congzhu Li
- Department of Gynecologic Oncology, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China.
| | - Hongping Xia
- School of Chemistry and Chemical Engineering & Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, 210009, China.
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing, 211166, China.
- Department of Gynecologic Oncology, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
36
|
Fang W, Lin H, Chen J, Guo W. A novel necroptosis-related gene signature in acute myeloid leukemia. Hematology 2023; 28:2227491. [PMID: 37367830 DOI: 10.1080/16078454.2023.2227491] [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: 04/08/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
OBJECTIVE Necroptosis has been reported to play an important role in different cancers, including leukemia. However, biomarkers of necroptosis-related genes (NRGs) that help predict the prognosis of AML are still lacking. Our research aims to build a novel signature of NRGs that could enhance our understanding of the molecular heterogeneity in leukemia. METHOD Gene expression profiles as well as clinical features were downloaded from TCGA and GEO databases. Data analysis were executed using R software version 4.2.1 and GraphPad Prism version 9.0.0. RESULT Univariate cox regression and lasso regression were applied to identify survival-specific genes. Four genes including FADD, PLA2G4A, PYCARD and ZBP1 were considered as independent risk factors that affect the prognosis of patients. Risk scores were calculated according to the coefficient of four genes. Then clinical characteristics and risk scores were enrolled to construct a nomogram. CellMiner was also used to screen potential drugs and analyze the correlations between genes and drug sensitivity. CONCLUSION In general, we established a signature of four genes related to necroptosis that could be helpful for future risk stratification in patients with AML.
Collapse
Affiliation(s)
- Weiyue Fang
- Department of Hematology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Hongdou Lin
- Department of Hematology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Junyi Chen
- Department of Hematology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wenjian Guo
- Medical and Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
37
|
Wen XM, Xu ZJ, Ma JC, Xia PH, Jin Y, Chen XY, Qian W, Lin J, Qian J. Identification and validation of necroptosis-related gene signatures to predict clinical outcomes and therapeutic responses in acute myeloid leukemia. Aging (Albany NY) 2023; 15:14677-14702. [PMID: 37993258 PMCID: PMC10781507 DOI: 10.18632/aging.205231] [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: 05/15/2023] [Accepted: 10/02/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Necroptosis is a tightly regulated form of necrotic cell death that promotes inflammation and contributes to disease development. However, the potential roles of necroptosis-related genes (NRGs) in acute myeloid leukemia (AML) have not been elucidated fully. METHODS We conducted a study to identify a robust biomarker signature for predicting the prognosis and immunotherapy efficacy based on NRGs in AML. We analyzed the genetic and transcriptional alterations of NRGs in 151 patients with AML. Then, we identified three necroptosis clusters. Moreover, a necroptosis score was constructed and assessed based on the differentially expressed genes (DEGs) between the three necroptosis clusters. RESULTS Three necroptosis clusters were correlated with clinical characteristics, prognosis, the tumor microenvironment, and infiltration of immune cells. A high necroptosis score was positively associated with a poor prognosis, immune-cell infiltration, expression of programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1), immune score, stromal score, interferon-gamma (IFNG), merck18, T-cell dysfunction-score signatures, and cluster of differentiation-86, but negatively correlated with tumor immune dysfunction and exclusion score, myeloid-derived suppressor cells, and M2-type tumor-associated macrophages. Our observations indicated that a high necroptosis score might contribute to immune evasion. More interestingly, AML patients with a high necroptosis score may benefit from treatment based on immune checkpoint blockade. CONCLUSIONS Consequently, our findings may contribute to deeper understanding of NRGs in AML, and facilitate assessment of the prognosis and treatment strategies.
Collapse
Affiliation(s)
- Xiang-Mei Wen
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- Zhenjiang Clinical Research Center of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- Zhenjiang Clinical Research Center of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- Zhenjiang Clinical Research Center of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Pei-Hui Xia
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- Zhenjiang Clinical Research Center of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Ye Jin
- Zhenjiang Clinical Research Center of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- Department of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Xin-Yi Chen
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- Zhenjiang Clinical Research Center of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Wei Qian
- Department of Otolaryngology-Head and Neck Surgery, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Jiang Lin
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- Zhenjiang Clinical Research Center of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| | - Jun Qian
- Zhenjiang Clinical Research Center of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
- Department of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, P.R. China
| |
Collapse
|
38
|
Wang Y, Pan KH, Chen M. Necroptosis-related genes are associated with prognostic features of kidney renal clear cell carcinoma. Discov Oncol 2023; 14:192. [PMID: 37878133 PMCID: PMC10600093 DOI: 10.1007/s12672-023-00794-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
INTRODUCTION Renal clear cell carcinoma is a common type of cancer in the adult urological system. It has a high mortality rate, with 30% of patients developing metastasis and 60% dying within 1-2 years of diagnosis. Recent advancements in tumor immunology and necroptosis have provided new insights into kidney cancer therapy. Therefore, it is crucial to identify potential targets for combining immunotherapy with necroptosis. MATERIALS AND METHODS Using the GSE168845 dataset and necroptosis-related genes, we identified genes that are differentially expressed in relation to necroptosis. We analyzed the prognostic value of these genes through differential expression analysis, prognostic analysis, and Cox regression analysis. The expression levels of the MYCN and CDKN2A genes were verified using the GSE53757 dataset. We also examined the association between the differentially expressed genes and clinicopathological features, as well as overall survival in our cohorts. In addition, we constructed a lasso Cox regression model to assess the correlation between these genes and immune score, ICP, and OCLR score. We conducted qRT-PCR to detect the expression of MYCN, CDKN2A, and ZBP1 in different samples of kidney renal clear cell carcinoma (KIRC). The expression levels of these genes were verified in a normal kidney cell line (HK-2 cells) and two KIRC cell lines (786-O, ACHN). The protein levels of MYCN and CDKN2A were detected using immunohistochemistry (IHC). SiRNA was used to silence the expression of MYCN and CDKN2A in the ACHN cell line, and wound healing assays were performed to measure cell migration. RESULTS MYCN, CDKN2A, and ZBP1 were identified as necroptosis-related genes with independent prognostic value, leading to the development of a risk prognostic model. The expression of the CDKN2A gene was significantly higher in KIRC tissues compared to normal tissues, while the expression of the MYCN gene was significantly lower in KIRC tissues. The expression of MYCN and CDKN2A was associated with tumor stage, metastasis, and overall survival in our cohort. Furthermore, MYCN, CDKN2A, and ZBP1 were significantly correlated with immune score, ICP, and OCLR score. The expression levels of CDKN2A and ZBP1 were higher in KIRC cells compared to normal kidney cells, while the expression of MYCN was lower in KIRC cells. The protein expression of MYCN and CDKN2A was also higher in KIRC tissues, as confirmed by IHC. The results of the wound healing assay indicated that silencing CDKN2A inhibited cell migration, while silencing MYCN enhanced cell migration. CONCLUSIONS MYCN and CDKN2A are potential targets and valuable prognostic biomarkers for combining immunotherapy with necroptosis in kidney renal clear cell carcinoma. CDKN2A promotes the migration of renal cancer cells, while MYCN inhibits their migration.
Collapse
Affiliation(s)
- Yiduo Wang
- Affiliated Zhongda Hospital of Southeast University, Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China
| | - Ke-Hao Pan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Ming Chen
- Affiliated Zhongda Hospital of Southeast University, Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China.
- Department of Urology, Lishui District People's Hospital, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China.
| |
Collapse
|
39
|
Alves-Hanna FS, Crespo-Neto JA, Nogueira GM, Pereira DS, Lima AB, Ribeiro TLP, Santos VGR, Fonseca JRF, Magalhães-Gama F, Sadahiro A, Costa AG. Insights Regarding the Role of Inflammasomes in Leukemia: What Do We Know? J Immunol Res 2023; 2023:5584492. [PMID: 37577033 PMCID: PMC10421713 DOI: 10.1155/2023/5584492] [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: 04/28/2023] [Revised: 07/02/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Inflammation is a physiological mechanism of the immune response and has an important role in maintaining the hematopoietic cell niche in the bone marrow. During this process, the participation of molecules produced by innate immunity cells in response to a variety of pathogen-associated molecular patterns and damage-associated molecular patterns is observed. However, chronic inflammation is intrinsically associated with leukemogenesis, as it induces DNA damage in hematopoietic stem cells and contributes to the creation of the preleukemic clone. Several factors influence the malignant transformation within the hematopoietic microenvironment, with inflammasomes having a crucial role in this process, in addition to acting in the regulation of hematopoiesis and its homeostasis. Inflammasomes are intracellular multimeric complexes responsible for the maturation and secretion of the proinflammatory cytokines interleukin-1β and interleukin-18 and the cell death process via pyroptosis. Therefore, dysregulation of the activation of these complexes may be a factor in triggering several diseases, including leukemias, and this has been the subject of several studies in the area. In this review, we summarized the current knowledge on the relationship between inflammation and leukemogenesis, in particular, the role of inflammasomes in different types of leukemias, and we describe the potential therapeutic targets directed at inflammasomes in the leukemic context.
Collapse
Affiliation(s)
- Fabíola Silva Alves-Hanna
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Juniel Assis Crespo-Neto
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Glenda Menezes Nogueira
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil
| | - Daniele Sá Pereira
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil
| | - Amanda Barros Lima
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Thaís Lohana Pereira Ribeiro
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil
| | | | - Joey Ramone Ferreira Fonseca
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Fábio Magalhães-Gama
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
| | - Aya Sadahiro
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
- Escola de Enfermagem de Manaus, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| |
Collapse
|
40
|
Li S, Wang X, Liu Y, Xiao J, Yi J. The implication of necroptosis-related lncRNAs in orchestrating immune infiltration and predicting therapeutic efficacy in colon adenocarcinoma: an integrated bioinformatic analysis with preliminarily experimental validation. Front Genet 2023; 14:1170640. [PMID: 37600653 PMCID: PMC10433646 DOI: 10.3389/fgene.2023.1170640] [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: 02/21/2023] [Accepted: 04/17/2023] [Indexed: 08/22/2023] Open
Abstract
Background: Necroptosis contributes significantly to colon adenocarcinoma (COAD). We aim to assess the relationship between immunoinfiltration and stemness in COAD patients through the development of a risk score profile using necroptosis-related long noncoding RNAs (NRLs). Methods: Our study was based on gene expression data and relevant clinical information from The Cancer Genome Atlas (TCGA). Necroptosis-related genes (NRGs) were obtained from the Kyoto Encyclopedia of Genes and Genome (KEGG) database. Pearson correlation analysis, Cox regression, and least absolute shrinkage and selection operator (LASSO) regression were used to determine the NRL prognositic signature (NRLPS). NRLs expression was examined using qRT-PCR method. Several algorithms were used to identify relationships between immune cell infiltration and NRLPS risk scores. Further analysis of somatic mutations, tumor stemness index (TSI), and drug sensitivity were also explored. Results: To construct NRLPS, 15 lncRNAs were investigated. Furthermore, NRLPS patients with high-risk subgroups had lower survival rates than that of patients with low-risk subgroups. Using GSEA analysis, NRL was found to be enriched in Notch, Hedgehog and Smoothened pathways. Immune infiltration analysis showed significant differences in CD8+ T cells, dendritic cell DCs, and CD4+ T cells between the two risk groups. In addition, our NRLPS showed a relevance with the regulation of tumor microenvironment, tumor mutation burden (TMB) and stemness. Finally, NRLPS demonstrated potential applications in predicting the efficacy of immunotherapy and chemotherapy in patients with COAD. Conclusion: Based on NRLs, a prognostic model was developed for COAD patients that allows a personalized tailoring immunotherapy and chemotherapy to be tailored.
Collapse
Affiliation(s)
- Shizhe Li
- Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial People’s Hospital, Changsha, Hunan, China
| | - Xiaotong Wang
- Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial People’s Hospital, Changsha, Hunan, China
| | - Yajun Liu
- Hunan Provincial People’s Hospital, Changsha, Hunan, China
| | - Junbo Xiao
- Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial People’s Hospital, Changsha, Hunan, China
| | - Jun Yi
- Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial People’s Hospital, Changsha, Hunan, China
| |
Collapse
|
41
|
Duan X, Du H, Yuan M, Liu L, Liu R, Shi J. Bioinformatics analysis of necroptosis‑related lncRNAs and immune infiltration, and prediction of the prognosis of patients with esophageal carcinoma. Exp Ther Med 2023; 26:331. [PMID: 37346407 PMCID: PMC10280318 DOI: 10.3892/etm.2023.12030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/21/2023] [Indexed: 06/23/2023] Open
Abstract
Esophageal carcinoma (ESCA) is one of the most common malignancies in the world, and has high morbidity and mortality rates. Necrosis and long noncoding RNAs (lncRNAs) are involved in the progression of ESCA; however, the specific mechanism has not been clarified. The aim of the present study was to investigate the role of necrosis-related lncRNAs (nrlncRNAs) in patients with ESCA by bioinformatics analysis, and to establish a nrlncRNA model to predict ESCA immune infiltration and prognosis. To form synthetic matrices, ESCA transcriptome data and related information were obtained from The Cancer Genome Atlas. A nrlncRNA model was established by coexpression, univariate Cox (Uni-Cox), and least absolute shrinkage and selection operator analyses. The predictive ability of this model was evaluated by Kaplan-Meier, receiver operating characteristic (ROC) curve, Uni-Cox, multivariate Cox regression, nomogram and calibration curve analyses. A model containing eight nrlncRNAs was generated. The areas under the ROC curves for 1-, 3- and 5-year overall survival were 0.746, 0.671 and 0.812, respectively. A high-risk score according to this model could be used as an indicator for systemic therapy use, since the half-maximum inhibitory concentration values varied significantly between the high-risk and low-risk groups. Based on the expression of eight prognosis-related nrlncRNAs, the patients with ESCA were regrouped using the 'ConsensusClusterPlus' package to explore potential molecular subgroups responding to immunotherapy. The patients with ESCA were divided into three clusters based on the eight nrlncRNAs that constituted the risk model: The most low-risk group patients were classified into cluster 1, and the high-risk group patients were mainly concentrated in clusters 2 and 3. Survival analysis showed that Cluster 1 had a better survival than the other groups (P=0.016). This classification system could contribute to precision treatment. Furthermore, two nrlncRNAs (LINC02811 and LINC00299) were assessed in the esophageal epithelial cell line HET-1A, and in the human esophageal cancer cell lines KYSE150 and TE1. There were significant differences in the expression levels of these lncRNAs between tumor and normal cells. In conclusion, the present study suggested that nrlncRNA models may predict the prognosis of patients with ESCA, and provide guidance for immunotherapy and chemotherapy decision making. Furthermore, the present study provided strategies to promote the development of individualized and precise treatment for patients with ESCA.
Collapse
Affiliation(s)
- Xiaoyang Duan
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Huazhen Du
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Meng Yuan
- Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 804-8550, Japan
| | - Lie Liu
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Rongfeng Liu
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jian Shi
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
42
|
Yu X, Liu X, Liu X, Jin S, Zhong M, Nie D, Zeng X, Wang X, Tan J, Li Y, Zeng C. Overexpression of CASP1 triggers acute promyelocytic leukemia cell pyroptosis and differentiation. Eur J Pharmacol 2023; 945:175614. [PMID: 36822457 DOI: 10.1016/j.ejphar.2023.175614] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023]
Abstract
Caspase-1 (CASP1)-mediated classical pyroptosis plays a key role in cancer development and management, however, the role of CASP1 and its regulation has not yet been documented for acute promyelocytic leukemia (APL). Here, we found that CASP1/GSDMD had lower expression in patients with APL and most other subtypes of primary de novo acute myeloid leukemia (AML) and was increased in all-trans-retinoic acid (ATRA)-treated APL cells. We showed that ATRA increases and activates CASP1 to trigger the pyroptosis and differentiation of APL cells. Mechanistically, ATRA could induce CASP1 expression via the IFNγ/STAT1 pathway in APL cells. In conclusion, ATRA-induced activation of CASP1 may serve as a suppressor in APL progression, as it triggers pyroptotic cell death and differentiation.
Collapse
Affiliation(s)
- Xibao Yu
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - Xin Liu
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China; Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, China
| | - Xuan Liu
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - Shuang Jin
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - Mengjun Zhong
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - Dingrui Nie
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiangbo Zeng
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xianfeng Wang
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China; Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, China
| | - Jiaxiong Tan
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yangqiu Li
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China.
| | - Chengwu Zeng
- The First Affiliated Hospital and Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
43
|
Nandi S, Mondal A, Ghosh A, Mukherjee S, Das C. Lnc-ing epigenetic mechanisms with autophagy and cancer drug resistance. Adv Cancer Res 2023; 160:133-203. [PMID: 37704287 DOI: 10.1016/bs.acr.2023.03.002] [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] [Indexed: 09/15/2023]
Abstract
Long noncoding RNAs (lncRNAs) comprise a diverse class of RNA molecules that regulate various physiological processes and have been reported to be involved in several human pathologies ranging from neurodegenerative disease to cancer. Therapeutic resistance is a major hurdle for cancer treatment. Over the past decade, several studies has emerged on the role of lncRNAs in cancer drug resistance and many trials have been conducted employing them. LncRNAs also regulate different cell death pathways thereby maintaining a fine balance of cell survival and death. Autophagy is a complex cell-killing mechanism that has both cytoprotective and cytotoxic roles. Similarly, autophagy can lead to the induction of both chemosensitization and chemoresistance in cancer cells upon therapeutic intervention. Recently the role of lncRNAs in the regulation of autophagy has also surfaced. Thus, lncRNAs can be used in cancer therapeutics to alleviate the challenges of chemoresistance by targeting the autophagosomal axis. In this chapter, we discuss about the role of lncRNAs in autophagy-mediated cancer drug resistance and its implication in targeted cancer therapy.
Collapse
Affiliation(s)
- Sandhik Nandi
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India
| | - Aritra Ghosh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Indian Institute of Science Education and Research, Kolkata, India
| | - Shravanti Mukherjee
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
44
|
Wang Y, Su X, Yin Y, Wang Q. Identification and Analysis of Necroptosis-Related Genes in COPD by Bioinformatics and Experimental Verification. Biomolecules 2023; 13:biom13030482. [PMID: 36979417 PMCID: PMC10046193 DOI: 10.3390/biom13030482] [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: 12/13/2022] [Revised: 02/16/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous and complex progressive inflammatory disease. Necroptosis is a newly identified type of programmed cell death. However, the role of necroptosis in COPD is unclear. This study aimed to identify necroptosis-related genes in COPD and explore the roles of necroptosis and immune infiltration through bioinformatics. The analysis identified 49 differentially expressed necroptosis-related genes that were primarily engaged in inflammatory immune response pathways. The infiltration of CD8+ T cells and M2 macrophages in COPD lung tissue was relatively reduced, whereas that of M0 macrophages was increased. We identified 10 necroptosis-related hub genes significantly associated with infiltrated immune cells. Furthermore, 7 hub genes, CASP8, IL1B, RIPK1, MLKL, XIAP, TNFRSF1A, and CFLAR, were validated using an external dataset and experimental mice. CFLAR was considered to have the best COPD-diagnosing capability. TF and miRNA interactions with common hub genes were identified. Several related potentially therapeutic molecules for COPD were also identified. The present findings suggest that necroptosis occurs in COPD pathogenesis and is correlated with immune cell infiltration, which indicates that necroptosis may participate in the development of COPD by interacting with the immune response.
Collapse
Affiliation(s)
- Yingxi Wang
- Institute of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xin Su
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yan Yin
- Institute of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Hospital of China Medical University, Shenyang 110001, China
- Correspondence: (Y.Y.); (Q.W.)
| | - Qiuyue Wang
- Institute of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Hospital of China Medical University, Shenyang 110001, China
- Correspondence: (Y.Y.); (Q.W.)
| |
Collapse
|
45
|
Peng YL, Wang LX, Li MY, Liu LP, Li RS. Construction and validation of a prognostic signature based on necroptosis-related genes in hepatocellular carcinoma. PLoS One 2023; 18:e0279744. [PMID: 36795724 PMCID: PMC9934426 DOI: 10.1371/journal.pone.0279744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/04/2022] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Necroptosis is a necrotic programmed cell death with potent immunogenicity. Due to the dual effects of necroptosis on tumor growth, metastasis and immunosuppression, we evaluated the prognostic value of necroptosis-related genes (NRGs) in hepatocellular carcinoma (HCC). METHODS We first analyzed RNA sequencing and clinical HCC patient data obtained to develop an NRG prognostic signature based on the TCGA dataset. Differentially expressed NRGs were further evaluated by GO and KEGG pathway analyses. Next, we conducted univariate and multivariate Cox regression analyses to build a prognostic model. We also used the dataset obtained from the International Cancer Genome Consortium (ICGC) database to verify the signature. The Tumor Immune Dysfunction and Exclusion (TIDE) algorithm was used to investigate the immunotherapy response. Furthermore, we investigated the relationship between the prediction signature and chemotherapy treatment response in HCC. RESULTS We first identified 36 differentially expressed genes out of 159 NRGs in hepatocellular carcinoma. Enrichment analysis showed that they were mainly enriched in the necroptosis pathway. Four NRGs were screened by Cox regression analysis to establish a prognostic model. The survival analysis revealed that the overall survival of patients with high-risk scores was significantly shorter than that of patients with low-risk scores. The nomogram demonstrated satisfactory discrimination and calibration. The calibration curves validated a fine concordance between the nomogram prediction and actual observation. The efficacy of the necroptosis-related signature was also validated by an independent dataset and immunohistochemistry experiments. TIDE analysis revealed that patients in the high-risk group were possibly more susceptible to immunotherapy. Furthermore, high-risk patients were found to be more sensitive to conventional chemotherapeutic medicines such as bleomycin, bortezomib, and imatinib. CONCLUSION We identified 4 necroptosis-related genes and established a prognostic risk model that could potentially predict prognosis and response to chemotherapy and immunotherapy in HCC patients in the future.
Collapse
Affiliation(s)
- Yue-ling Peng
- Department of Nephrology, Shanxi Provincial People’s Hospital (Fifth Hospital of Shanxi Medical University), Taiyuan, China
| | - Ling-xiao Wang
- Department of Colorectal and Anal Surgery, Shanxi Provincial People’s Hospital (Fifth Hospital of Shanxi Medical University), Taiyuan, China
| | - Mu-ye Li
- Department of Ocular Fundus Diseases, Shanxi Eye Hospital, Shanxi Medical University, Taiyuan, China
| | - Li-ping Liu
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Rong-shan Li
- Department of Nephrology, Shanxi Provincial People’s Hospital (Fifth Hospital of Shanxi Medical University), Taiyuan, China
- * E-mail:
| |
Collapse
|
46
|
Lin P, Lin C, He R, Chen H, Teng Z, Yao H, Liu S, Hoffman RM, Ye J, Zhu G. TRAF6 regulates the abundance of RIPK1 and inhibits the RIPK1/RIPK3/MLKL necroptosis signaling pathway and affects the progression of colorectal cancer. Cell Death Dis 2023; 14:6. [PMID: 36604411 PMCID: PMC9816173 DOI: 10.1038/s41419-022-05524-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023]
Abstract
Colorectal cancer cannot be completely cured at present, and it is still an important clinical medical problem. TRAF6 is highly expressed in many malignant tumors. However, the role of TRAF6 in colorectal cancer is still controversial, mainly because the specific regulatory mechanism of colorectal cancer is still unclear, and the death mode of colorectal cancer cells has not been elucidated. The recent study found that TRAF6 inhibits necroptosis in colorectal cancer cells via the RIPK1/RIPK3/MLKL signaling pathway. The RIPK1 inhibitor Necrostain-1 inhibits colorectal cancer cell necroptosis via the RIPK1/RIPK3/MLKL signaling pathway. TRAF6 directly interacts with RIPK1 through the polyubiquitination of Lys48-linked RIPK1 and reduces the levels of RIPK1 protein in colorectal cancer cells, leading to necroptosis, thus promoting the proliferation of colorectal cancer cells. The recent study demonstrated that TRAF6 promotes colorectal cell progression by inhibiting the RIPK1/RIPK3/MLKL necroptosis signaling pathway, which may provide a new therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Penghang Lin
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Chunlin Lin
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Ruofan He
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Hui Chen
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Zuhong Teng
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Hengxin Yao
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Songyi Liu
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Robert M Hoffman
- AntiCancer, Inc., San Diego, CA, USA
- Department of Surgery, University of California, San Diego, CA, USA
| | - Jianxin Ye
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
| | - Guangwei Zhu
- Department of Gastrointestinal Surgery 2 Section, Institute of Abdominal Surgery, Key Laboratory of Accurate Diagnosis and Treatment of Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
47
|
Wu YY, Li CC, Lin X, Xu F, Shan SK, Guo B, Li FXZ, Zheng MH, Xu QS, Lei LM, Duan JY, Tang KX, Cao YC, Yuan LQ. Global publication trends and research trends of necroptosis application in tumor: A bibliometric analysis. Front Pharmacol 2023; 14:1112484. [PMID: 37169000 PMCID: PMC10164947 DOI: 10.3389/fphar.2023.1112484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/03/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction: Necroptosis is an alternative, caspase-independent programmed cell death that appears when apoptosis is inhibited. A gowing number of studies have reflected the link between necroptosis and tumors. However, only some systematical bibliometric analyses were focused on this field. In this study, we aimed to identify and visualize the cooperation between countries, institutions, authors, and journals through a bibliometric analysis to help understand the hotspot trends and emerging topics regarding necroptosis and cancer research. Methods: The articles and reviews on necroptosis and cancer were obtained from the Web of Science Core Collection on 16 September 2022. Countries, institutions, authors, references, and keywords in this field were visually analyzed by CtieSpace 5.8.R3, VOSviewer 1.6.18, and R package "bibliometrix." Results: From 2006 to 2022, 2,216 qualified original articles and reviews on necroptosis in tumors were published in 685 academic journals by 13,009 authors in 789 institutions from 75 countries/regions. Publications focusing on necroptosis and cancer have increased violently in the past 16 years, while the citation number peaked around 2008-2011. Most publications were from China, while the United States maintained the dominant position as a "knowledge bridge" in necroptosis and cancer research; meanwhile, Ghent University and the Chinese Academy of Sciences were the most productive institutions. Moreover, only a tiny portion of the articles were multiple-country publications. Peter Vandenabeele had the most significant publications, while Alexei Degterev was most often co-cited. Peter Vandenabeele also gets the highest h-index and g-index in this research field. Cell Death and Disease was the journal with the most publications on necroptosis and cancer, which was confirmed to be the top core source by Bradford's Law. At the same time, Cell was the leading co-cited journal, and the focus area of these papers was molecular, biology, and immunology. High-frequency keywords mainly contained those that are molecularly related (MLKL, NF-kB, TNF, RIPK3, RIPK1), pathological process related (necroptosis, apoptosis, cell-death, necrosis, autophagy), and mechanism related (activation, expression, mechanisms, and inhibition). Conclusion: This study comprehensively overviews necroptosis and cancer research using bibliometric and visual methods. Research related to necroptosis and cancer is flourishing. Cooperation and communication between countries and institutions must be further strengthened. The information in our paper would provide valuable references for scholars focusing on necroptosis and cancer.
Collapse
Affiliation(s)
- Yun-Yun Wu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chang-chun Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fu-Xing-Zi Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yue Duan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Xin Tang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ye-Chi Cao
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Ling-Qing Yuan,
| |
Collapse
|
48
|
Identification and Validation a Necroptosis-Related Prognostic Signature in Cervical Cancer. Reprod Sci 2022; 30:2003-2015. [PMID: 36576713 DOI: 10.1007/s43032-022-01155-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022]
Abstract
Necroptosis is a promising novel target for cervical cancer therapy. Nevertheless, differentially expressed necroptosis-related genes (NRGs) in cervical cancer and their associations with prognosis are far from fully clarified. In this study, differentially expressed NRGs (DE-NRGs) were screened out and their bio-function was elucidated. Subsequently, a prognostic scoring model based on the regression coefficients of the screened out NRGs and their corresponding mRNA expressions were constructed and validated. Finally, the survival probability of cervical cancer patients based on the constructed prognostic scoring model in 3 and 5 years was predicted and assessed. We found 17 DE-NRGs in cervical cancer tissues which were closely related to cancer progression, and most of them were significantly highly expressed. Furthermore, 3 NRG were confirmed as the prognostic signature genes from 17 DE-NRGs by regression analysis. Overall survival predicted through our prognostic scoring model was lower in the high-risk group than in the low-risk group (p < 0.05) in both the TCGA cohort and the external GEO44001 validation cohort. What's more, the prediction performance of our prognostic scoring models well verified by the ROC curve, and the risk score calculated could act as an independent prognostic factor for cervical cancer patients. The calibration curve and C-index (0.776) of the nomogram analysis suggested that the predictive performance of the nomogram was satisfactory. Our study identified and validated a necroptosis-related prognostic signature in cervical cancer, which could well predict the prognosis for cervical cancer patients.
Collapse
|
49
|
Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, Liu J, Yu X, Shi S. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol 2022; 15:174. [PMID: 36482419 PMCID: PMC9733270 DOI: 10.1186/s13045-022-01392-3] [Citation(s) in RCA: 434] [Impact Index Per Article: 144.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Many types of human cells self-destruct to maintain biological homeostasis and defend the body against pathogenic substances. This process, called regulated cell death (RCD), is important for various biological activities, including the clearance of aberrant cells. Thus, RCD pathways represented by apoptosis have increased in importance as a target for the development of cancer medications in recent years. However, because tumor cells show avoidance to apoptosis, which causes treatment resistance and recurrence, numerous studies have been devoted to alternative cancer cell mortality processes, namely necroptosis, pyroptosis, ferroptosis, and cuproptosis; these RCD modalities have been extensively studied and shown to be crucial to cancer therapy effectiveness. Furthermore, evidence suggests that tumor cells undergoing regulated death may alter the immunogenicity of the tumor microenvironment (TME) to some extent, rendering it more suitable for inhibiting cancer progression and metastasis. In addition, other types of cells and components in the TME undergo the abovementioned forms of death and induce immune attacks on tumor cells, resulting in enhanced antitumor responses. Hence, this review discusses the molecular processes and features of necroptosis, pyroptosis, ferroptosis, and cuproptosis and the effects of these novel RCD modalities on tumor cell proliferation and cancer metastasis. Importantly, it introduces the complex effects of novel forms of tumor cell death on the TME and the regulated death of other cells in the TME that affect tumor biology. It also summarizes the potential agents and nanoparticles that induce or inhibit novel RCD pathways and their therapeutic effects on cancer based on evidence from in vivo and in vitro studies and reports clinical trials in which RCD inducers have been evaluated as treatments for cancer patients. Lastly, we also summarized the impact of modulating the RCD processes on cancer drug resistance and the advantages of adding RCD modulators to cancer treatment over conventional treatments.
Collapse
Affiliation(s)
- Xuhui Tong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong Tang
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Mingming Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiang Liu
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
50
|
He WP, Chen YY, Wu LX, Guo YY, You ZS, Yang GF. A novel necroptosis-related lncRNA signature for predicting prognosis and anti-cancer treatment response in endometrial cancer. Front Immunol 2022; 13:1018544. [PMID: 36466815 PMCID: PMC9708746 DOI: 10.3389/fimmu.2022.1018544] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/31/2022] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Necroptosis, a form of programmed cell death, underlies tumorigenesis and the progression of cancers. Anti-cancer strategies targeting necroptosis have increasingly been shown to present a potential cancer therapy. However, the predictive utility and anticancer sensitivity value of necroptosis-related lncRNAs (NRLs) for endometrial cancer (EC) are currently unknown. METHODS EC patient gene expression profiles and the corresponding clinical information collected from The Cancer Genome Atlas were used to identify NRLs that constituted a predictive signature for EC. The functional pathways, immune status, clinicopathological correlation, and anticancer drug sensitivity of the patients relative to the NRLs signatures were analyzed. RESULTS A signature composed of 7 NRLs (AC019080.5, BOLA3-AS1, AC022144.1, AP000345.2, LEF1-AS1, AC010503.4, and RPARP-AS1) was identified. The high-risk patient group with this signature exhibited a poorer prognosis and lower survival rate than low-risk group lacking this signature. This necroptosis-related lncRNA signature had a higher predictive accuracy compared with other clinicopathological variables (area under the receiver operating characteristic curve of the risk score: 0.717). Additionally, when patients were stratified based on other clinicopathological variables, the overall survival was significantly shorter in the high-risk versus low-risk group across all cohorts. Gene set enrichment analysis (GSEA) revealed that immune- and tumor-related signaling pathways and biological processes were enriched in the high-risk group compared to the low-risk group. Single-sample gene set enrichment analysis (ssGSEA) additionally showed that the resulting risk score was strongly correlated with EC patient immune status. Finally, patients with high-risk scores were more sensitive to the anti-cancer drugs such as Docetaxel, Mitomycin.C, Vinblastine, AZD.2281 (olaparib), AZD6244, and PD.0332991 (Palbociclib). CONCLUSION These findings reveal a novel necroptosis-related lncRNA signature for predicting EC patient prognosis and shed new light on anticancer therapy strategies for EC.
Collapse
Affiliation(s)
| | | | | | | | | | - Guo-Fen Yang
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|