1
|
Chavan PR, Pandey R, Patil BM, Murti K, Kumar N. Unravelling key signaling pathways for the therapeutic targeting of non-small cell lung cancer. Eur J Pharmacol 2025; 998:177494. [PMID: 40090536 DOI: 10.1016/j.ejphar.2025.177494] [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: 12/11/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
Lung cancer (LC) remains the foremost cause of cancer-related mortality across the globe. Non-small cell lung cancer (NSCLC) is a type of LC that exhibits significant heterogeneity at histological and molecular levels. Genetic alterations in upstream signaling molecules activate cascades affecting apoptosis, proliferation, and differentiation. Disruption of these signaling pathways leads to the proliferation of cancer-promoting cells, progression of cancer, and resistance to its treatment. Recent insights into the function of signaling pathways and their fundamental mechanisms in the onset of various diseases could pave the way for new therapeutic approaches. Recently, numerous drug molecules have been created that target these cell signaling pathways and could be used alongside other standard therapies to achieve synergistic effects in mitigating the pathophysiology of NSCLC. Additionally, many researchers have identified several predictive biomarkers, and alterations in transcription factors and related pathways are employed to create new therapeutic strategies for NSCLC. Findings suggest using specific inhibitors to target cellular signaling pathways in tumor progression to treat NSCLC. This review investigates the role of signaling pathways in NSCLC development and explores novel therapeutic strategies to enhance clinical treatment options for NSCLC.
Collapse
Affiliation(s)
- Pavan Ramrao Chavan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Ruchi Pandey
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Baswant Malesh Patil
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Nitesh Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India.
| |
Collapse
|
2
|
Qi X, Zhou J, Wang P, Li Y, Li H, Miao Y, Ma X, Luo X, Zhang Z, He Y, Shen W, Zhao W, Cui R, Li C, Zhu H, Lyu J. KLF7-regulated ITGA2 as a therapeutic target for inhibiting oral cancer stem cells. Cell Death Dis 2025; 16:354. [PMID: 40316546 PMCID: PMC12048542 DOI: 10.1038/s41419-025-07689-8] [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: 10/30/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer stem cells (CSCs) play crucial roles in tumor metastasis, therapy resistance, and immune evasion. Identifying and understanding the factors that regulate the stemness of tumor cells presents promising opportunities for developing effective therapeutic strategies. In this study on oral squamous cell carcinoma (OSCC), we confirmed the key role of KLF7 in maintaining the stemness of OSCC. Using chromatin immunoprecipitation sequencing and dual-luciferase assays, we identified ITGA2, a membrane receptor, as a key downstream gene regulated by KLF7 in the maintenance of stemness. Tumor sphere formation assays, flow cytometry analyses, and in vivo limiting dilution tumorigenicity evaluations demonstrated that knocking down ITGA2 significantly impaired stemness. Upon binding to its extracellular matrix (ECM) ligand, type I collagen, ITGA2 activates stemness-associated signaling pathways, including PI3K-AKT, MAPK, and Hippo. TC-I 15, a small-molecule inhibitor of the ITGA2-collagen interaction, significantly sensitizes oral squamous cell carcinoma (OSCC) to cisplatin in xenograft models. In summary, we reveal that the KLF7/ITGA2 axis is a crucial modulator of stemness in OSCC. Our findings suggest that ITGA2 is a promising therapeutic target, offering a novel anti-CSC strategy.
Collapse
Affiliation(s)
- Xin Qi
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Jiang Zhou
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Pan Wang
- Department of Stomatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yunyan Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Haoran Li
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Yuwen Miao
- Zhejiang University, School of Medicine, Affiliated Stomatology Hospital, Hangzhou, Zhejiang, P. R. China
| | - XiaoQing Ma
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiayan Luo
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Zhiling Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Yanling He
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Wenyi Shen
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Wenquan Zhao
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Rutao Cui
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Cang Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China.
| | - Huiyong Zhu
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China.
| | - Jiong Lyu
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China.
| |
Collapse
|
3
|
Liu X, Cui Y, Gong J, Yu X, Cui Y, Xuan Y. SETD5 facilitates stemness and represses ferroptosis via m6A-mediating PKM2 stabilization in non-small cell lung cancer. Oncogene 2025:10.1038/s41388-025-03426-9. [PMID: 40307507 DOI: 10.1038/s41388-025-03426-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
SETD5, an atypical member of the histone lysine methyltransferase family known for its association with cancer stemness, is a significant predictor of unfavorable survival outcomes in non-small cell lung cancer (NSCLC). However, the function of SETD5 in NSCLC stemness remains unclear, and whether it is an active H3K36me3 is controversial. Consequently, further investigation is required to clarify the pivotal role of SETD5 in NSCLC stemness and its related mechanism. Thus, this study employed the NSCLC tissue microarray and bioinformatics tools to analyze SETD5 expression and determine its effect on stemness and investigated the role of SETD5 in the metastasis of NSCLC using in vitro and in vivo analyses. The findings indicated high SETD5 expression in embryonic and NSCLC tissues, which was related to the pathological tumor stage, lymph node metastasis, and clinical stage, indicating that SETD5 could be used as a biomarker and prognostic factor in NSCLC. In addition, we found that SETD5 can promote glycolysis, thereby inhibiting ferroptosis and promoting the stemness of NSCLC, causing tumor metastasis and adverse prognosis in patients. In terms of mechanism, SETD5 as H3K36me3 facilitates the m6A modification of METTL14 and the recruitment of YTHDF1 and mediates PKM2 nuclear translocation and phosphorylation of p-PKM2 Tyr105, regulating GPX4 mediated ferroptosis resistance and SOX9 mediated stemness in NSCLC. The findings emphasize that SETD5 may serve as a promising indicator of stemness in NSCLC, which can help develop therapeutic targets for NSCLC and prognostic evaluation. This study provides evidence that SETD5 as H3K36me3 facilitates the m6A modification of METTL14 and the recruitment of YTHDF1 and mediates the nuclear translocation of PKM2, regulating GPX4 mediated ferroptosis resistance and SOX9 mediated stemness, causing tumor metastasis and adverse prognosis in patients.
Collapse
Affiliation(s)
- Xingzhe Liu
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | - Yuzhen Cui
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Jie Gong
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | - Xinhui Yu
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yan Cui
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yanhua Xuan
- Department of Pathology, Yanbian University College of Medicine, Yanji, China.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China.
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China.
| |
Collapse
|
4
|
Dai W, Zhu M, Sun Y, Xu B, Ma G, Shi H, Li P. NELFCD Promotes Colon Cancer Progression by Regulating the DUSP2-p38 Axis. FRONT BIOSCI-LANDMRK 2025; 30:25221. [PMID: 40302325 DOI: 10.31083/fbl25221] [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: 06/12/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 05/02/2025]
Abstract
BACKGROUND To investigate the significance of the negative elongation factor complex member C/D (NELFCD) in colon cancer progression. METHODS Immunohistochemistry staining, Western blot analysis, and real-time quantitative polymerase chain reaction (RT-qPCR) were used to quantify the protein/gene levels. NELFCD-protein arginine methyltransferase 5 (PRMT5) interaction was determined by co-immunoprecipitation assay. A chromatin immunoprecipitation (ChIP) assay was performed to determine the interaction between the promoter region of dual specificity phosphatase 2 (DUSP2), NELFCD, and PRMT5. Cell growth and cell cycle progression were assessed using the cell counting kit-8 proliferation assay, colony formation assay, and/or flow cytometry. RESULTS NELFCD was upregulated in colon cancer and promoted cancer cell growth. In colon cancer cells, the expression of NELFCD was negatively correlated with DUSP2 expression. The RNA sequencing results indicated that genes in the mitogen-activated protein kinase (MAPK) signaling pathway as well as DUSP2 were affected by NELFCD. The ChIP sequencing results revealed that DUSP2 and genes in the MAPK signaling pathway are direct targets of NELFCD. ChIP assay verified that PRMT5 is enriched at the promoter region of DUSP2 and that NELFCD overexpression promoted this enrichment. A co-immunoprecipitation assay demonstrated that NELFCD was bound to PRMT5, functioning as a macromolecular complex. CONCLUSIONS This study suggests that NELFCD promotes the progression of colon cancer by recruiting PRMT5 to inhibit DUSP2 expression, which subsequently activates the p38 signaling pathway. Targeting the NELFCD-DUSP2-p38 signaling axis may be a promising therapeutic intervention for patients suffering from NELFCD-amplified tumors.
Collapse
Affiliation(s)
- Weiwei Dai
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, 100000, Beijing, China
| | - Min Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, 100000, Beijing, China
| | - Yujing Sun
- Department of Laboratory, Peking University International Hospital, 100000, Beijing, China
| | - Baohong Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, 100000, Beijing, China
| | - Guorong Ma
- Department of Pathology, Beijing Luhe Hospital Affiliated to Capital Medical University, 100000, Beijing, China
| | - Haiyun Shi
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, 100000, Beijing, China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, 100000, Beijing, China
| |
Collapse
|
5
|
Boyacıoğlu Ö, Kalali BD, Tongün E, Korkusuz P. A Niche-Based Perspective to Stem and Cancer Stem Cells of the Lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40178798 DOI: 10.1007/5584_2025_858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Lungs carry the principle function for the conduction and exchange of air through the primary, secondary, tertiary bronchi, bronchioles, and alveoli, resulting in the exchange of oxygen to carbon dioxide within the human tissues. Lung stem and progenitor cells enable differentiation of parenchymal and stromal elements and provide homeostasis and regeneration in the microenvironment against pulmonary diseases. Tumor-initiating cancer cells (TICs) refer to a subpopulation named as cancer stem cells (CSCs) of lung cancer exhibiting high self-renewal and proliferation capacity by Notch, Hippo, Hedgehog, and Wnt signaling pathways that leads to tumor development or recurrence. Lung cancer stem cells (LCSCs) are characterized by distinct genotypic or phenotypic alterations compared to healthy lung stem cells (LSCs) that provide a potential target to treat lung cancer. Therefore, understanding the cascades responsible for the transformation of healthy to CSCs is essential to develop new targeted therapy approaches. In this chapter, we precisely highlight the latest researches on LSCs and CSCs, key signaling mechanisms within the perspective of novel targeted therapy strategies.
Collapse
Affiliation(s)
- Özge Boyacıoğlu
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, Turkey
- Department of Medical Biochemistry, Faculty of Medicine, Atılım University, Gölbaşı, Ankara, Turkey
| | - Berfin Deniz Kalali
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, Turkey
| | - Ege Tongün
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Sıhhiye, Ankara, Turkey.
- METU MEMS Center, Ankara, Turkey.
| |
Collapse
|
6
|
Xu J, Zhang H, Nie Z, He W, Zhao Y, Huang Z, Jia L, Du Z, Zhang B, Xia S. Cancer stem-like cells stay in a plastic state ready for tumor evolution. Neoplasia 2025; 61:101134. [PMID: 39919692 PMCID: PMC11851212 DOI: 10.1016/j.neo.2025.101134] [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: 08/01/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
Cell plasticity emerges as a novel cancer hallmark and is pivotal in driving tumor heterogeneity and adaptive resistance to different therapies. Cancer stem-like cells (CSCs) are considered the root of cancer. While first defined as tumor-initiating cells with the potential to develop a heterogeneous tumor, CSCs further demonstrate their roles in cancer metastasis and adaptive therapeutic resistance. Generally, CSCs come from the malignant transformation of somatic stem cells or the de-differentiation of other cancer cells. The resultant cells gain more plasticity and are ready to differentiate into different cell states, enabling them to adapt to therapies and metastatic ecosystems. Therefore, CSCs are likely the nature of tumor cells that gain cell plasticity. However, the phenotypic plasticity of CSCs has never been systematically discussed. Here, we review the distinct intrinsic signaling pathways and unique microenvironmental niches that endow CSC plasticity in solid tumors to adapt to stressful conditions, as well as emerging opportunities for CSC-targeted therapy.
Collapse
Affiliation(s)
- Jiali Xu
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Houde Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhihao Nie
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wenyou He
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yichao Zhao
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zhenhui Huang
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
| | - Lin Jia
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China.
| | - Zhiye Du
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.
| | - Baotong Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Siyuan Xia
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
7
|
Wu M, Li W, Leung H, Wang Y, Wan Q, Chen P, Chen C, Li Y, Yao X, He M. Targeting WDPF domain of Hsp27 achieves a broad spectrum of antiviral. MedComm (Beijing) 2025; 6:e70032. [PMID: 40013315 PMCID: PMC11862887 DOI: 10.1002/mco2.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 02/28/2025] Open
Abstract
Enterovirus A71 (EV-A71) is a positive-sense single-stranded RNA virus, which hijacks host proteins to benefit viral internal ribosome entry site (IRES)-dependent protein translation and further propagation. We demonstrated that serine 78 (S78) phosphorylation of Hsp27 is critical for Hsp27/hnRNP A1 relocalization upon EV-A71 infection. Here, we report that the deletion of WDPF and ACD domains disturbs subcellular localization of Hsp27, resulting in partial nuclear translocation. The domain deletion-induced Hsp27 nuclear translocation fails to direct hnRNP A1 translocation. The 2Apro-induced IRES activity and viral replication are suppressed by the deletion of WDPF or ACD domain. Surprisingly, a peptide (WDPF) dramatically inhibits S78 phosphorylation. Therefore, hnRNP A1 translocation, viral IRES activity, and viral protein translation and propagation are all strongly suppressed by the WDPF peptide, but not by peptide without WDPFR sequence (ΔWDPF). Moreover, the WDPF peptide also has potent antiviral activity on other RNA virus (e.g., coronavirus HCoV-OC43) and DNA virus (e.g., HSV-1 and HBV). Peptide treatment with kinase inhibitor Sorafenib brings an additional inhibitory effect on HCoV-OC43 and HSV-1. Taken together, we uncover a crucial role of WDPF domain in S78 phosphorylation for EV-A71-induced hnRNP A1 nuclear translocation, IRES-dependent viral protein translation, and EV-A71 propagation. Our results explore a new path for target-based pan-antiviral strategy.
Collapse
Affiliation(s)
- Mandi Wu
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
| | - Wei Li
- Weihai Municipal HospitalCheeloo College of MedicineShandong UniversityWeihaiShandongChina
| | - Houying Leung
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
| | - Yiran Wang
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
| | - Qianya Wan
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
| | - Peiran Chen
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
| | - Cien Chen
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
| | - Yichen Li
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
| | - Xi Yao
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
| | - Ming‐Liang He
- Department of Biomedical SciencesCity University of Hong KongHong Kong Special Administrative RegionHong KongChina
- CityU Shenzhen Research InstituteNanshanShenzhenChina
| |
Collapse
|
8
|
Kucheryavenko AS, Muzyko EA, Perfilova VN, Kaplanov KD, Frolov MY. The role of the PPM1D gene in tumor pathogenesis. BIOMEDITSINSKAIA KHIMIIA 2025; 71:19-28. [PMID: 40045720 DOI: 10.18097/pbmcr1495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The PPM1D gene and its protein product (serine-threonine protein phosphatase, PPM1D or Wip1) are involved in regulation of cell's DNA damage response, cell cycle control, and repair. Amplification, overexpression, or mutations of the PPM1D gene have a significant impact on cell responses to stress factors and genetic instability as well as impairments of processes of double-strand break repair, nucleotide excision repair, base excision repair, cell cycle, and apoptosis. PPM1D dephosphorylates and thus inactivates p53, proteins that respond to DNA strand integrity damage, cell cycle checkpoint proteins, and apoptotic proteins. This contributes to tumor development, growth, and maintenance of the tumor phenotype. In this review we consider data on the role of the PPM1D gene in the formation and maintenance of various oncological processes, including tumors of the mammary glands, ovaries, prostate gland, esophagus, stomach, intestines, liver and pancreas, hemoblastoses, and others.
Collapse
Affiliation(s)
| | - E A Muzyko
- Volgograd State Medical University, Volgograd, Russia
| | - V N Perfilova
- Volgograd State Medical University, Volgograd, Russia; Volgograd Medical Research Center, Volgograd, Russia
| | | | - M Yu Frolov
- Volgograd State Medical University, Volgograd, Russia
| |
Collapse
|
9
|
Joseph S, Zhang X, Droby GN, Wu D, Bae-Jump V, Lyons S, Mordant A, Mills A, Herring L, Rushing B, Bowser JL, Vaziri C. MAPK14/p38α shapes the molecular landscape of endometrial cancer and promotes tumorigenic characteristics. Cell Rep 2025; 44:115104. [PMID: 39708320 DOI: 10.1016/j.celrep.2024.115104] [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: 06/18/2024] [Revised: 10/25/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
The molecular underpinnings of high-grade endometrial carcinoma (HGEC) metastatic growth and survival are poorly understood. Here, we show that ascites-derived and primary tumor HGEC cell lines in 3D spheroid culture faithfully recapitulate key features of malignant peritoneal effusion and exhibit fundamentally distinct transcriptomic, proteomic, and metabolomic landscapes compared with conventional 2D monolayers. Using a genetic screening platform, we identify MAPK14 (which encodes the protein kinase p38α) as a specific requirement for HGEC in spheroid culture. MAPK14/p38α has broad roles in programming the phosphoproteome, transcriptome, and metabolome of HGEC spheroids, yet has negligible impact on monolayer cultures. MAPK14 promotes tumorigenicity in vivo and is specifically required to sustain a sub-population of spheroid cells that is enriched in cancer stemness markers. Therefore, spheroid growth of HGEC activates unique biological programs, including p38α signaling, that cannot be captured using 2D culture models and are highly relevant to malignant disease pathology.
Collapse
Affiliation(s)
- Sayali Joseph
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xingyuan Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gaith N Droby
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Di Wu
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Victoria Bae-Jump
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Scott Lyons
- Department of Pharmacology, UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Angie Mordant
- Department of Pharmacology, UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Allie Mills
- Department of Pharmacology, UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Laura Herring
- Department of Pharmacology, UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Blake Rushing
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica L Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
10
|
Gupta S, Silveira DA, Mombach JCM, Hashimoto RF. DNA Damage-Induced Ferroptosis: A Boolean Model Regulating p53 and Non-Coding RNAs in Drug Resistance. Proteomes 2025; 13:6. [PMID: 39846637 PMCID: PMC11755436 DOI: 10.3390/proteomes13010006] [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: 11/14/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
The tumor suppressor p53, in its wild-type form, plays a central role in cellular homeostasis by regulating senescence, apoptosis, and autophagy within the DNA damage response (DDR). Recent findings suggest that wild-type p53 also governs ferroptosis, an iron-dependent cell death process driven by lipid peroxidation. Post-translational modifications of p53 generate proteoforms that significantly enhance its functional diversity in regulating these mechanisms. A key target in this process is the cystine/glutamate transporter (xCT), which is essential for redox balance and ferroptosis resistance. Additionally, p53-induced miR-34c-5p suppresses cancer cell proliferation and drug resistance by modulating Myc, an oncogene further influenced by non-coding RNAs like circular RNA NOTCH1 (CricNOTCH1) and long non-coding RNA MALAT1. However, the exact role of these molecules in ferroptosis remains unclear. To address this, we introduce the first dynamic Boolean model that delineates the influence of these ncRNAs and p53 on ferroptosis, apoptosis, and senescence within the DDR context. Validated through gain- and loss-of-function perturbations, our model closely aligns with experimental observations in cancers such as oral squamous cell carcinoma, nasopharyngeal carcinoma, and osteosarcoma. The model identifies crucial positive feedback loops (CricNOTCH1/miR-34c/Myc, MALAT1/miR-34c/Myc, and Myc/xCT) and highlights the therapeutic potential of using p53 proteoforms and ncRNAs to combat drug resistance and induce cancer cell death.
Collapse
Affiliation(s)
- Shantanu Gupta
- Instituto de Matemática e Estatística, Departamento de Ciência da Computação, Universidade de São Paulo, Rua do Matão 1010, São Paulo 05508-090, SP, Brazil;
| | | | - José Carlos M. Mombach
- Departamento de Física, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil;
| | - Ronaldo F. Hashimoto
- Instituto de Matemática e Estatística, Departamento de Ciência da Computação, Universidade de São Paulo, Rua do Matão 1010, São Paulo 05508-090, SP, Brazil;
| |
Collapse
|
11
|
Xie X, Shin YR, Kim TS, Seong YS, Yi YW, Kim DJ. Identification of sinensetin as a selective inhibitor for mitogen-activated protein kinase kinase 6 and an anticancer agent for non-small cell lung cancer. Am J Cancer Res 2025; 15:113-126. [PMID: 39949939 PMCID: PMC11815360 DOI: 10.62347/rgzg2120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/03/2025] [Indexed: 02/16/2025] Open
Abstract
Natural compounds are an invaluable source for bioactive small molecules. Cellular activities modulated by them are generally achieved by binding specific cellular targets. However, identification of target(s) for a natural compound is challenging and a hurdle for further development of them as drugs. Sinensetin is derived from Schisandra sphenanthera and the major component of a traditional medicine. Although Sinensetin possesses pharmacological activities, including antioxidants, anti-inflammatory, and anticancer, the molecular mechanisms for its activities remain unclear due to lack of information for its target. In addition, the anticancer effects of sinensetin against non-small cell lung cancer (NSCLC) have not been studied. Here, we described sinensetin as a specific inhibitor of MKK6 with a KD value of 66.27 μM. Sinensetin inhibited the proliferation of NSCLC cells and lung patient-derived xenograft-derived organoids (LPDXO), and induced G1 phase cell-cycle arrest. Sinensetin attenuated the MAPK signaling pathway by directly inhibiting MKK6, but not MKK3. In silico molecular docking analysis indicated that sinensetin was specifically bound near the αG-helix of MKK6, but not MKK3. High MKK6 expression levels were observed in NSCLC patients. MKK6 knockout abolished the sinensetin-mediated inhibition of NSCLC cell proliferation. Taken together, sinensetin is a novel MKK6 inhibitor with therapeutic potential for NSCLC.
Collapse
Affiliation(s)
- Xiaomeng Xie
- Chest Hospital of Zhengzhou UniversityZhengzhou 450000, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou UniversityZhengzhou 450008, Henan, China
- China-US (Henan) Hormel Cancer InstituteZhengzhou 450008, Henan, China
| | - Young Ran Shin
- Department of Microbiology, College of Medicine, Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
| | - Tae-Sung Kim
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Biochemistry, College of Medicine, Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
| | - Yeon-Sun Seong
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Biochemistry, College of Medicine, Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
| | - Yong Weon Yi
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Department of Biochemistry, College of Medicine, Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
| | - Dong Joon Kim
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou UniversityZhengzhou 450008, Henan, China
- China-US (Henan) Hormel Cancer InstituteZhengzhou 450008, Henan, China
- Department of Microbiology, College of Medicine, Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
- Multidrug-Resistant Refractory Cancer Convergence Research Center (MRCRC), Dankook UniversityCheonan-si 31116, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
12
|
Dai Q, Yao X, Zhang Y, Chai Q, Feng X, Zhu H, Zhao L. CTSG is a prognostic marker involved in immune infiltration and inhibits tumor progression though the MAPK signaling pathway in non-small cell lung cancer. J Cancer Res Clin Oncol 2024; 151:21. [PMID: 39724501 PMCID: PMC11671429 DOI: 10.1007/s00432-024-06051-3] [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: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE This study aims to investigate the biological roles and molecular mechanisms of Cathepsin G (CTSG) in the progression of non-small cell lung cancer (NSCLC). METHODS Western blotting and immunohistochemistry analyses of clinical samples were performed to determine the expression levels of CTSG in patients with NSCLC. Bioinformatic analysis of clinical datasets was conducted to evaluate the correlation between CTSG and lymph node metastasis, tumor stage, and immune cell infiltration. Gain-of-function assays and tumor implantation experiments were employed to determine the effects of CTSG on malignant behaviors of NSCLC cells. Transcriptome sequencing and subsequent bioinformatic analysis were performed to explore the signaling pathways regulated by CTSG. Western blotting and qPCR were utilized to assess the influence of CTSG on the MAPK and EMT signaling pathways. RESULTS CTSG is expressed at low levels and serves as a prognostic marker in NSCLC. The downregulation of CTSG expression was associated with lymph node metastasis, tumor stage, and immune cell infiltration. CTSG inhibits NSCLC cell proliferation, migration, and invasion as well as tumor growth in nude mice. There exists a significant correlation between CTSG expression and endoplasmic reticulum function, cell cycling, and the IL-17 signaling pathway. CTSG suppresses the MAPK and EMT signaling pathways in NSCLC cells. Moreover, DNA methylation and histone deacetylation have been identified as crucial mechanisms contributing to the decreased expression of CTSG. CONCLUSION CTSG inhibits NSCLC development by suppressing the MAPK signaling pathway and is also associated with tumor immunity.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/immunology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Animals
- Prognosis
- Mice
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- MAP Kinase Signaling System/physiology
- Disease Progression
- Female
- Male
- Mice, Nude
- Cell Proliferation
- Middle Aged
- Gene Expression Regulation, Neoplastic
- Mice, Inbred BALB C
- Cell Line, Tumor
Collapse
Affiliation(s)
- Qian Dai
- School of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xufeng Yao
- School of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yanke Zhang
- Department of Respiratory Medicine, The Fuyang Affiliated Hospital of Anhui Medical University, Fuyang, 236000, Anhui, China
| | - Qian Chai
- Department of Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230012, Anhui, China
- Department of Respiratory Medicine, Anhui Public Health Clinical Center, Hefei, 230012, Anhui, China
| | - Xueyi Feng
- School of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Hongbin Zhu
- Department of Respiratory Medicine, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China.
| | - Lei Zhao
- Department of Respiratory Medicine, The Fuyang Affiliated Hospital of Anhui Medical University, Fuyang, 236000, Anhui, China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230012, Anhui, China.
- Department of Respiratory Medicine, Anhui Public Health Clinical Center, Hefei, 230012, Anhui, China.
| |
Collapse
|
13
|
Lagorgette L, Bogdanova DA, Belotserkovskaya EV, Garrido C, Demidov ON. PP2C phosphatases-terminators of suicidal thoughts. Cell Death Dis 2024; 15:919. [PMID: 39702569 DOI: 10.1038/s41419-024-07269-2] [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: 06/14/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024]
Abstract
Cell death and related signaling pathways are essential during development and in various physiological and pathological conditions. Post-translational modifications such as ubiquitination and phosphorylation play an important role in these signaling pathways. The involvement of kinases - enzymes that catalyze protein phosphorylation - in cell death signaling has been extensively studied. On the other hand, not many studies have been devoted to analyzing the role in cell death of phosphatases, enzymes involved in the removal of phosphorylated residues added to proteins by kinases. Obviously, the two opposite reactions, phosphorylation and dephosphorylation, are equally important in the regulation of protein functions and subsequently in the execution of the cell death program. Here, we have summarized recent work on the involvement of serine-threonine PP2C phosphatases in cell death pathways, senescence and autophagy, focusing in particular on the most studied phosphatase PPM1D (PP2Cδ) as an example of the regulatory role of PP2Cs in cell death. The review should help to draw attention to the importance of PP2C family phosphatases in cell death checkpoints and to discover new targets for drug development.
Collapse
Affiliation(s)
- Lisa Lagorgette
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
| | - Daria A Bogdanova
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia
- Institute of Cytology RAS, St. Petersburg, Russia
| | | | - Carmen Garrido
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
- Center for Cancer Georges-François Leclerc, Dijon, France
| | - Oleg N Demidov
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France.
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia.
- Institute of Cytology RAS, St. Petersburg, Russia.
| |
Collapse
|
14
|
Yan W, Wang X, Wang W, Guo Q, Huang N, Chen H, Liang XJ, Han Y, Liu D, Zhang J. The p38/MAPK pathway as a therapeutic target to prevent therapeutic escape of breast cancer stem cells. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1867-1880. [PMID: 38951428 DOI: 10.1007/s11427-023-2585-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 03/15/2024] [Indexed: 07/03/2024]
Abstract
Cancer stem cells (CSCs) play an important role in metastasis development, tumor recurrence, and treatment resistance, and are essential for the eradication of cancer. Currently, therapies fail to eradicate CSCs due to their therapeutic stress-induced cellular escape, which leads to enhanced aggressive behaviors compared with CSCs that have never been treated. However, the underlying mechanisms regulating the therapeutic escape remain unknown. To this end, we established a model to isolate the therapeutic escaped CSCs (TSCSCs) from breast CSCs and performed the transcription profile to reveal the mechanism. Mechanistically, we demonstrated that the behavior of therapeutic escape was regulated through the p38/MAPK signaling pathway, resulting in TSCSCs exhibiting enhanced motility and metastasis. Notably, blocking the p38/MAPK signaling pathway effectively reduced motility and metastasis ability both in vitro and in vivo, which were further supported by downregulated motility-related genes and epithelial-mesenchymal transition (EMT)-related proteins vimentin and N-cadherin. The obtained findings reveal the p38/MAPK pathway as a potential therapeutic target for TSCSCs and would provide profound implications for cancer therapy.
Collapse
Affiliation(s)
- Weixiao Yan
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Xiaotong Wang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Wenjing Wang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Qi Guo
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Na Huang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Hao Chen
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yu Han
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Dandan Liu
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Jinchao Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education; College of Chemistry and Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China.
| |
Collapse
|
15
|
Wang Y, Cui C, Zhao W, Tian X, Liu P, Wei L, Zhu Z, Liu M, Fu R, Jia L. WIP1-mediated regulation of p38 MAPK signaling attenuates pyroptosis in sepsis-associated acute kidney injury. Immunobiology 2024; 229:152832. [PMID: 38943814 DOI: 10.1016/j.imbio.2024.152832] [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: 01/27/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/01/2024]
Abstract
Wild-Type p53-Induced Phosphatase 1 (WIP1/PPM1D) is a serine/threonine phosphatase that plays a significant role in various physiological processes. However, the involvement of WIP1 in kidney remains unclear. Lipopolysaccharide (LPS) was administered to induce acute injury in mice and human kidney 2 (HK2) cells in the study. The WIP1 inhibitor, CCT007093, was administered both in vitro and in vivo to assess its effect on kidney. The single-cell sequencing (scRNA-seq) data revealed that Ppm1d mRNA reached peak on day 2 following unilateral ischemia-reperfusion injury (uni-IRI) in mice, especially in the proximal renal tubules during repair phase. Compared to the control group, WIP1 protein exhibited a significant increase in renal tubules of patients with acute tubular injury (ATI) and mice with LPS-induced acute kidney injury (AKI), as well as in LPS-injured HK2 cells. In vitro experiments showed that CCT007093 increased the protein levels of NLRP3, cleaved-Caspase1, GSDMD-N and IL-1β in HK2 cells and further reduced the viability of LPS-stimulated HK2 cells. In vivo experiments showed that inhibition of WIP1 activity with CCT007093 further increased cleaved-Caspase1, GSDMD-N protein levels in kidney tissue from mice with LPS-induced AKI. In addition, LPS induces phosphorylation of p38 MAPK, a key regulator of pyroptosis, which is further activated by CCT007093. In conclusion, inhibition of WIP1 activity acts as a positive regulator of renal tubular pyroptosis mainly through the mediation of phospho-p38 MAPK.
Collapse
Affiliation(s)
- Yinhong Wang
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chenkai Cui
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Weihao Zhao
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xuefei Tian
- Section of Nephrology, Department of Internal medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Pengfei Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Linting Wei
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zikun Zhu
- Department of Computer Science, School of Computing & Department of Electrical and Computer Engineering, National University of Singapore, Singapore
| | - Ming Liu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rongguo Fu
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Lining Jia
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
16
|
Joseph S, Zhang X, Droby G, Wu D, Bae-Jump V, Lyons S, Mordant A, Mills A, Herring L, Rushing B, Bowser J, Vaziri C. MAPK14 /p38α Shapes the Molecular Landscape of Endometrial Cancer and promotes Tumorigenic Characteristics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600674. [PMID: 38979238 PMCID: PMC11230443 DOI: 10.1101/2024.06.25.600674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The molecular underpinnings of H igh G rade E ndometrial C arcinoma (HGEC) metastatic growth and survival are poorly understood. Here we show that ascites-derived and primary tumor HGEC cell lines in 3D spheroid culture faithfully recapitulate key features of malignant peritoneal effusion and exhibit fundamentally distinct transcriptomic, proteomic and metabolomic landscapes when compared with conventional 2D monolayers. Using genetic screening platform we identify MAPK14 (which encodes the protein kinase p38α) as a specific requirement for HGEC in spheroid culture. MAPK14 /p38α has broad roles in programing the phosphoproteome, transcriptome and metabolome of HGEC spheroids, yet has negligible impact on monolayer cultures. MAPK14 promotes tumorigenicity in vivo and is specifically required to sustain a sub-population of spheroid cells that is enriched in cancer stemness markers. Therefore, spheroid growth of HGEC activates unique biological programs, including p38α signaling, that cannot be captured using 2D culture models and are highly relevant to malignant disease pathology.
Collapse
|
17
|
Qamar F, Sharif Z, Idrees J, Wasim A, Haider S, Salman S. SARS-CoV-2-induced phosphorylation and its pharmacotherapy backed by artificial intelligence and machine learning. Future Sci OA 2024; 10:FSO917. [PMID: 38827795 PMCID: PMC11140666 DOI: 10.2144/fsoa-2023-0112] [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: 06/16/2023] [Accepted: 10/04/2023] [Indexed: 06/05/2024] Open
Abstract
Aims: To investigate the role of phosphorylation in SARS-CoV-2 infection, potential therapeutic targets and its harmful genetic sequences. Materials & Methods: Data mining techniques were employed to identify upregulated kinases responsible for proteomic changes induced by SARS-CoV-2. Spike and nucleocapsid proteins' sequences were analyzed using predictive tools, including SNAP2, MutPred2, PhD-SNP, SNPs&Go, MetaSNP, Predict-SNP and PolyPhen-2. Missense variants were identified using ensemble-based algorithms and homology/structure-based models like SIFT, PROVEAN, Predict-SNP and MutPred-2. Results: Eight missense variants were identified in viral sequences. Four damaging variants were found, with SNPs&Go and PolyPhen-2. Promising therapeutic candidates, including gilteritinib, pictilisib, sorafenib, RO5126766 and omipalisib, were identified. Conclusion: This research offers insights into SARS-CoV-2 pathogenicity, highlighting potential treatments and harmful variants in viral proteins.
Collapse
Affiliation(s)
- Fouzia Qamar
- Department of Biology, Lahore Garrison University, Lahore-54000, Punjab, Pakistan
| | - Zubair Sharif
- Faculty of Medical Laboratory Sciences, Superior University, Lahore-54000, Punjab, Pakistan
| | - Jawaria Idrees
- Khyber Pakhtunkhwa Education Monitoring Authority, Khyber-Pakhtunkhwa, Peshawar-25000, Pakistan
| | - Asif Wasim
- Department of Pharmacy, CECOS University of IT & Emerging Sciences, Peshawar-25000, Khyber Pakhtunkhwa, Peshawar, Pakistan
| | - Sana Haider
- Department of Pharmacy, CECOS University of IT & Emerging Sciences, Peshawar-25000, Khyber Pakhtunkhwa, Peshawar, Pakistan
| | - Saad Salman
- Department of Pharmacy, CECOS University of IT & Emerging Sciences, Peshawar-25000, Khyber Pakhtunkhwa, Peshawar, Pakistan
| |
Collapse
|
18
|
Ten A, Kumeiko V, Farniev V, Gao H, Shevtsov M. Tumor Microenvironment Modulation by Cancer-Derived Extracellular Vesicles. Cells 2024; 13:682. [PMID: 38667297 PMCID: PMC11049026 DOI: 10.3390/cells13080682] [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: 02/11/2024] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in the process of tumorigenesis, regulating the growth, metabolism, proliferation, and invasion of cancer cells, as well as contributing to tumor resistance to the conventional chemoradiotherapies. Several types of cells with relatively stable phenotypes have been identified within the TME, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), neutrophils, and natural killer (NK) cells, which have been shown to modulate cancer cell proliferation, metastasis, and interaction with the immune system, thus promoting tumor heterogeneity. Growing evidence suggests that tumor-cell-derived extracellular vesicles (EVs), via the transfer of various molecules (e.g., RNA, proteins, peptides, and lipids), play a pivotal role in the transformation of normal cells in the TME into their tumor-associated protumorigenic counterparts. This review article focuses on the functions of EVs in the modulation of the TME with a view to how exosomes contribute to the transformation of normal cells, as well as their importance for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Artem Ten
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Vadim Kumeiko
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Vladislav Farniev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China;
| | - Maxim Shevtsov
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str., 2, 197341 St. Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str., 22, 81675 Munich, Germany
| |
Collapse
|
19
|
Gupta S, Silveira DA, Piedade GP, Ostrowski MP, Mombach JCM, Hashimoto RF. A dynamic Boolean network reveals that the BMI1 and MALAT1 axis is associated with drug resistance by limiting miR-145-5p in non-small cell lung cancer. Noncoding RNA Res 2024; 9:185-193. [PMID: 38125755 PMCID: PMC10730431 DOI: 10.1016/j.ncrna.2023.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 12/23/2023] Open
Abstract
Patients with non-small cell lung cancer (NSCLC) are often treated with chemotherapy. Poor clinical response and the onset of chemoresistance limit the anti-tumor benefits of drugs such as cisplatin. According to recent research, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long non-coding RNA related to cisplatin resistance in NSCLC. Furthermore, MALAT1 targets microRNA-145-5p (miR-145), which activates Krüppel-like factor 4 (KLF4) in associated cell lines. B lymphoma Mo-MLV insertion region 1 homolog (BMI1), on the other hand, inhibits miR-145 expression, which stimulates Specificity protein 1 (Sp1) to trigger the epithelial-mesenchymal transition (EMT) process in pemetrexed-resistant NSCLC cells. The interplay between these molecules in drug resistance is still unclear. Therefore, we propose a dynamic Boolean network that can encapsulate the complexity of these drug-resistant molecules. Using published clinical data for gain or loss-of-function perturbations, our network demonstrates reasonable agreement with experimental observations. We identify four new positive circuits: miR-145/Sp1/MALAT1, BMI1/miR-145/Myc, KLF4/p53/miR-145, and miR-145/Wip1/p38MAPK/p53. Notably, miR-145 emerges as a central player in these regulatory circuits, underscoring its pivotal role in NSCLC drug resistance. Our circuit perturbation analysis further emphasizes the critical involvement of these new circuits in drug resistance for NSCLC. In conclusion, targeting MALAT1 and BMI1 holds promise for overcoming drug resistance, while activating miR-145 represents a potential strategy to significantly reduce drug resistance in NSCLC.
Collapse
Affiliation(s)
- Shantanu Gupta
- Instituto de Matemática e Estatística, Departamento de Ciência da Computação, Universidade de São Paulo, Rua do Matão 1010, 05508-090, São Paulo, SP, Brazil
| | - Daner A. Silveira
- Children's Cancer Institute, Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel P.S. Piedade
- Instituto de Matemática e Estatística, Departamento de Ciência da Computação, Universidade de São Paulo, Rua do Matão 1010, 05508-090, São Paulo, SP, Brazil
| | - Miguel P. Ostrowski
- Instituto de Matemática e Estatística, Departamento de Ciência da Computação, Universidade de São Paulo, Rua do Matão 1010, 05508-090, São Paulo, SP, Brazil
| | - José Carlos M. Mombach
- Departamento de Física, Universidade Federal de Santa Maria, Santa Maria, 97105-900, RS, Brazil
| | - Ronaldo F. Hashimoto
- Instituto de Matemática e Estatística, Departamento de Ciência da Computação, Universidade de São Paulo, Rua do Matão 1010, 05508-090, São Paulo, SP, Brazil
| |
Collapse
|
20
|
Zhang C, Meng Y, Han J. Emerging roles of mitochondrial functions and epigenetic changes in the modulation of stem cell fate. Cell Mol Life Sci 2024; 81:26. [PMID: 38212548 PMCID: PMC11072137 DOI: 10.1007/s00018-023-05070-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024]
Abstract
Mitochondria serve as essential organelles that play a key role in regulating stem cell fate. Mitochondrial dysfunction and stem cell exhaustion are two of the nine distinct hallmarks of aging. Emerging research suggests that epigenetic modification of mitochondria-encoded genes and the regulation of epigenetics by mitochondrial metabolites have an impact on stem cell aging or differentiation. Here, we review how key mitochondrial metabolites and behaviors regulate stem cell fate through an epigenetic approach. Gaining insight into how mitochondria regulate stem cell fate will help us manufacture and preserve clinical-grade stem cells under strict quality control standards, contributing to the development of aging-associated organ dysfunction and disease.
Collapse
Affiliation(s)
- Chensong Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yang Meng
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
21
|
Wang Y, Hu S, Zhang W, Zhang B, Yang Z. Emerging role and therapeutic implications of p53 in intervertebral disc degeneration. Cell Death Discov 2023; 9:433. [PMID: 38040675 PMCID: PMC10692240 DOI: 10.1038/s41420-023-01730-5] [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: 08/27/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023] Open
Abstract
Lower back pain (LBP) is a common degenerative musculoskeletal disease that imposes a huge economic burden on both individuals and society. With the aggravation of social aging, the incidence of LBP has increased globally. Intervertebral disc degeneration (IDD) is the primary cause of LBP. Currently, IDD treatment strategies include physiotherapy, medication, and surgery; however, none can address the root cause by ending the degeneration of intervertebral discs (IVDs). However, in recent years, targeted therapy based on specific molecules has brought hope for treating IDD. The tumor suppressor gene p53 produces a transcription factor that regulates cell metabolism and survival. Recently, p53 was shown to play an important role in maintaining IVD microenvironment homeostasis by regulating IVD cell senescence, apoptosis, and metabolism by activating downstream target genes. This study reviews research progress regarding the potential role of p53 in IDD and discusses the challenges of targeting p53 in the treatment of IDD. This review will help to elucidate the pathogenesis of IDD and provide insights for the future development of precision treatments.
Collapse
Affiliation(s)
- Yidian Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Shouye Hu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Weisong Zhang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Binfei Zhang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhi Yang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
22
|
Yu N, Li T, Qiu Z, Xu J, Li Y, Huang J, Yang Y, Li Z, Long X, Zhang H. Wip1 regulates wound healing by affecting activities of keratinocytes and endothelial cells through ATM-p53 and mTOR signaling. Burns 2023; 49:1969-1982. [PMID: 37357059 DOI: 10.1016/j.burns.2023.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND As a p53-regulated gene, Wip1 regulates proliferation, migration, apoptosis, and senescence of several type cells, but its biological functions in keratinocytes and endothelial cells which are involved wound healing are not fully understood. This study aims to reveal the function and underlying mechanism of Wip1 in wound healing using models of transgenic animal, keratinocytes, and endothelial cells. METHODS Using Wip1 knockout C57 BL/6 mice, we investigated effect of Wip1 deficiency on wound healing and angiogenesis; And using HaCaT and HUVEC as keratinocytes and endothelial cells, combined using primary keratinocytes from Wip1 knockout mice, we studied the effects of Wip1 knockdown/knockout or overexpression on proliferation, migration, and protein expressions of signaling components in ATM-p53 and mTOR pathway. RESULTS Wip1 deficiency in mice impaired the wound repair and endothelial angiogenesis, reduced the thickness of granulation tissue, and decreased the number of Ki67-positive cells and CD31 positive vessels in granulation tissue. Knockdown of Wip1 by shRNAs suppressed the proliferation and migration of HaCaT and HUVEC cells and induced notably apoptosis in the two cells. In western blot, Wip1 knockdown enriched p53 and ATM proteins, while decreased activated AKT, mTOR and activated S6 ribosomal protein (pS6) levels in HaCaT and HUVEC cells. Ectopic expression of Wip1 decreased the p53 and ATM proteins, while increased activated AKT, mTOR and pS6 levels in HaCaT and HUVEC cells. And in primary keratinocytes from mice tail skin, Wip1 knockout increased p53 and ATM, while decreased activated AKT, mTOR and pS6 protein levels. CONCLUSION Our study directly supports that Wip1 regulated skin wound healing possibly by affecting bioactivities including proliferation, migration and apoptosis of keratinocytes and endothelial cells at least through by modulating ATM-p53 and mTOR signaling.
Collapse
Affiliation(s)
- Nanze Yu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianhao Li
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zikai Qiu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Xu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunzhu Li
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiuzuo Huang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yilan Yang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhujun Li
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Long
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hongbing Zhang
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
23
|
Chen Z, Qi Y, Shen J, Chen Z. Histone demethylase KDM6A coordinating with KMT2B regulates self-renewal and chemoresistance of non-small cell lung cancer stem cells. Transl Oncol 2023; 37:101778. [PMID: 37683307 PMCID: PMC10493599 DOI: 10.1016/j.tranon.2023.101778] [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: 05/14/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND AND AIMS Wnt signaling is essential for the maintenance of cancer stem cells (CSCs), but mutations in the β-catenin and APC genes are less common in non-small cell lung carcinoma (NSCLC). Thus, the mechanism underlying the constitutive activation of Wnt signaling in lung CSCs is still unknown. MATERIALS AND METHODS Gene set enrichment analysis and immunohistochemistry were performed to establish the correlation between KDM6A/KM2B and CSC stemness. Human NSCLC cell lines were genetically manipulated for functional studies. Sphere formation assay and stemness gene expression profiling were examined to investigate the role of KDM6A/KMT2B in lung CSCs. Tumor xenograft assay were used to identify the function of KDM6A/KMT2B on tumorigenicity and tumor recurrence in vivo. Western blot analysis, coimmunoprecipitation and chromatin immunoprecipitation were performed to understand KDM6A/KMT2B mediated epigenetic regulation of Histone 3 lysine 4 methylation (H3K4me) on Wnt signaling pathway. RESULTS We discovered that the expression of Histone demethylase KDM6A and methyltransferase KMT2B correlate with the stemness of CSCs in NSCLC. KDM6A coordinates with KMT2B to activate the Wnt/β-catenin signaling pathway by regulating the H3K4me3 level and promotes the tumorigenicity and maintenance of CSC stemness. Furthermore, KDM6A/ KMT2B overexpression promotes the CSC chemoresistance and tumor recurrence both in vitro and in vivo. Inhibition of KDM6A and KMT2B potently suppress tumor initiation and recurrence in xenografted animal models. CONCLUSION Our findings suggest that KDM6A and KMT2B mediate the constitutive activation of Wnt/β-catenin signaling in lung CSCs, potentially providing a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Zhiwei Chen
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuwen Qi
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Shen
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhen Chen
- Department of Pathology, Shidong hospital, Yangpu District, Shidong hospital affiliated to University of Shanghai for Science and Technology, China.
| |
Collapse
|
24
|
Wu R, Ma R, Duan X, Zhang J, Li K, Yu L, Zhang M, Liu P, Wang C. Identification of specific prognostic markers for lung squamous cell carcinoma based on tumor progression, immune infiltration, and stem index. Front Immunol 2023; 14:1236444. [PMID: 37841237 PMCID: PMC10570622 DOI: 10.3389/fimmu.2023.1236444] [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: 06/07/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction Lung squamous cell carcinoma (LUSC) is a unique subform of nonsmall cell lung cancer (NSCLC). The lack of specific driver genes as therapeutic targets leads to worse prognoses in patients with LUSC, even with chemotherapy, radiotherapy, or immune checkpoint inhibitors. Furthermore, research on the LUSC-specific prognosis genes is lacking. This study aimed to develop a comprehensive LUSC-specific differentially expressed genes (DEGs) signature for prognosis correlated with tumor progression, immune infiltration,and stem index. Methods RNA sequencing data for LUSC and lung adenocarcinoma (LUAD) were extracted from The Cancer Genome Atlas (TCGA) data portal, and DEGs analyses were conducted in TCGA-LUSC and TCGA-LUAD cohorts to identify specific DEGs associated with LUSC. Functional analysis and protein-protein interaction network were performed to annotate the roles of LUSC-specific DEGs and select the top 100 LUSC-specific DEGs. Univariate Cox regression and least absolute shrinkage and selection operator regression analyses were performed to select prognosis-related DEGs. Results Overall, 1,604 LUSC-specific DEGs were obtained, and a validated seven-gene signature was constructed comprising FGG, C3, FGA, JUN, CST3, CPSF4, and HIST1H2BH. FGG, C3, FGA, JUN, and CST3 were correlated with poor LUSC prognosis, whereas CPSF4 and HIST1H2BH were potential positive prognosis markers in patients with LUSC. Receiver operating characteristic analysis further confirmed that the genetic profile could accurately estimate the overall survival of LUSC patients. Analysis of immune infiltration demonstrated that the high risk (HR) LUSC patients exhibited accelerated tumor infiltration, relative to low risk (LR) LUSC patients. Molecular expressions of immune checkpoint genes differed significantly between the HR and LR cohorts. A ceRNA network containing 19 lncRNAs, 50 miRNAs, and 7 prognostic DEGs was constructed to demonstrate the prognostic value of novel biomarkers of LUSC-specific DEGs based on tumor progression, stemindex, and immune infiltration. In vitro experimental models confirmed that LUSC-specific DEG FGG expression was significantly higher in tumor cells and correlated with immune tumor progression, immune infiltration, and stem index. In vitro experimental models confirmed that LUSC-specific DEG FGG expression was significantly higher in tumor cells and correlated with immune tumor progression, immune infiltration, and stem index. Conclusion Our study demonstrated the potential clinical implication of the 7- DEGs signature for prognosis prediction of LUSC patients based on tumor progression, immune infiltration, and stem index. And the FGG could be an independent prognostic biomarker of LUSC promoting cell proliferation, migration, invasion, THP-1 cell infiltration, and stem cell maintenance.
Collapse
Affiliation(s)
- Rihan Wu
- School of Life Science, Inner Mongolia University, Hohhot, China
- The Department of Oncology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Ru Ma
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Xiaojun Duan
- School of Life Science, Inner Mongolia University, Hohhot, China
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Jiandong Zhang
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Kexin Li
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Lei Yu
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Mingyang Zhang
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Pengxia Liu
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Changshan Wang
- School of Life Science, Inner Mongolia University, Hohhot, China
| |
Collapse
|
25
|
Dai Y, Wu Z, Chen Y, Ye X, Wang C, Zhu H. OCT4's role and mechanism underlying oral squamous cell carcinoma. J Zhejiang Univ Sci B 2023; 24:796-806. [PMID: 37701956 PMCID: PMC10500100 DOI: 10.1631/jzus.b2200602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/06/2023] [Indexed: 09/14/2023]
Abstract
Oral squamous cell carcinoma (OSCC), a common malignancy of the head and neck, ranks sixth worldwide in terms of cancers with the most negative impact, owing to tumor relapse rates, cervical lymphnode metastasis, and the lack of an efficacious systemic therapy. Its prognosis is poor, and its mortality rate is high. Octamer-binding transcription factor 4 (OCT4) is a member of the Pit-Oct-Unc (POU) family and is a key reprogramming factor that produces a marked effect in preserving the pluripotency and self-renewal state of embryonic stem cells (ESCs). According to recent studies, OCT4 participates in retaining the survival of OSCC cancer stem cells (CSCs), which has far-reaching implications for the occurrence, recurrence, metastasis, and prognosis of oral carcinogenesis. Therefore, we summarize the structure, subtypes, and function of OCT4 as well as its role in the occurrence, progression, and prognosis of OSCC.
Collapse
Affiliation(s)
- Yuwei Dai
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ziqiong Wu
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yitong Chen
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xinjian Ye
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Chaowei Wang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huiyong Zhu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
26
|
Zhang J, Yang T, Han M, Wang X, Yang W, Guo N, Ren Y, Cui W, Li S, Zhao Y, Zhai X, Jia L, Yang J, Wu C, Wang L. Gain-of-function mutations in the catalytic domain of DOT1L promote lung cancer malignant phenotypes via the MAPK/ERK signaling pathway. SCIENCE ADVANCES 2023; 9:eadc9273. [PMID: 37256945 PMCID: PMC10413674 DOI: 10.1126/sciadv.adc9273] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 04/25/2023] [Indexed: 06/02/2023]
Abstract
Lung cancer is a lethal malignancy lacking effective therapies. Emerging evidence suggests that epigenetic enzyme mutations are closely related to the malignant phenotype of lung cancer. Here, we identified a series of gain-of-function mutations in the histone methyltransferase DOT1L. The strongest of them is R231Q, located in the catalytic DOT domain. R231Q can enhance the substrate binding ability of DOT1L. Moreover, R231Q promotes cell growth and drug resistance of lung cancer cells in vitro and in vivo. Mechanistic studies also revealed that the R231Q mutant specifically activates the MAPK/ERK signaling pathway by enriching H3K79me2 on the RAF1 promoter and epigenetically regulating the expression of downstream targets. The combination of a DOT1L inhibitor (SGC0946) and a MAPK/ERK axis inhibitor (binimetinib) can effectively reverse the R231Q-induced phenomena. Our results reveal gain-of-function mutations in an epigenetic enzyme and provide promising insights for the precise treatment of lung cancer patients.
Collapse
Affiliation(s)
- Jiayu Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Ting Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Mei Han
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Xiaoxuan Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Weiming Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Ning Guo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Yong Ren
- Department of Pathology, General Hospital of Central Theater Command of People's Liberation Army, Wuhan 430070, China
| | - Wei Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shangxiao Li
- Department of Biochemistry and Molecular Biology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yongshan Zhao
- Department of Biochemistry and Molecular Biology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lina Jia
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| |
Collapse
|
27
|
Chang H, Sun W, Zhao Y, Sun T, Zhao Z. Targeting Pokemon is a novel strategy to suppress cancer aggressiveness of non-small cell lung cancer: Identification of Pokemon as ideal target for developing anti-NSCLC drugs. Arch Biochem Biophys 2023; 742:109637. [PMID: 37182800 DOI: 10.1016/j.abb.2023.109637] [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: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/16/2023]
Abstract
Although it is widely reported that Pokemon acts as an oncogene in the pathogenesis of multiple cancers, but its role and detailed molecular mechanisms in regulating non-small cell lung cancer (NSCLC) progression have not been fully delineated. Here, by performing Real-Time qPCR analysis, we verified that Pokemon was high-expressed in NSCLC tissues and cells, compared to the corresponding normal lung tissues and epithelial cells. Then, the small interfering RNA (siRNA) for Pokemon was transfected into the NSCLC cells to verify its biological functions, and our results suggested that silencing of Pokemon suppressed the malignant phenotypes, including cell viability, mitosis, colony formation, epithelial-mesenchymal transition (EMT), mobility and cancer stem cell (CSC) properties in NSCLC cells. Mechanistically, we confirmed that knockdown of Pokemon decreased the expression levels of phosphorylated Akt (p-Akt), phosphorylated GSK-3β (p-GSK-3β) and Snail to inactivate the oncogenic Akt/GSK-3β/Snail signal pathway, and deletion of Snail also had similar effects to hamper the development of NSCLC. Next, our rescuing experiments validated that Pokemon ablation-induced suppressing effects on NSCLC cell malignancy were all abrogated by overexpressing Snail. Finally, the in vivo experiments confirmed that silencing of Pokemon downregulated Snail to hamper tumorigenesis of NSCLC cells in xenograft tumor-bearing mice models. Taken together, we firstly uncovered the underlying mechanisms by which the Pokemon/Akt/GSK-3β/Snail signal pathway contributed to the development of NSCLC, and this signal pathway could be potentially used as therapeutic targets for the development of personalized anti-NSCLC drugs.
Collapse
Affiliation(s)
- Hao Chang
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Wenxue Sun
- Department of Respiratory Medicine, The Third People's Hospital of Shenzhen City, Shenzhen, 518000, Guangdong, China.
| | - Yiming Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Tianhao Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Zhihong Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street No. 23, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
28
|
Tan B, Zhang J, Wang W, Ma H, Yang Y. Tumor-suppressive E3 ubiquitin ligase CHIP inhibits the PBK/ERK axis to repress stem cell properties and radioresistance in non-small cell lung cancer. Apoptosis 2022; 28:397-413. [PMID: 36436119 DOI: 10.1007/s10495-022-01789-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 11/28/2022]
Abstract
Recently, radioresistant cancer cells surviving radiotherapy have been suggested to show more aggressive phenotypes than parental cells, and the underlying mechanisms may be associated with cancer stem cells. This study provided novel mechanistic insights for E3 ubiquitin ligase CHIP in stem cell properties and radioresistance of non-small cell lung cancer (NSCLC). After bioinformatic prediction for key genes involved, NSCLC tissues and cells were collected to measure the expression of CHIP and PBK. E3 ubiquitin ligase CHIP was poorly expressed, while PBK was highly expressed in NSCLC tissues and cells. CHIP reduced the protein stability of PBK through the ubiquitin-protease pathway to repress the activation of ERK pathway. Based on the gain- or loss-of-function experiments, it was noted that restoration of CHIP curtailed stem cell properties and radioresistance in NSCLC, as manifested by inhibited sphere formation and cell proliferation, decreased number of CD133+CD44+ cells and expression of OCT4, SOX2, and NANOG, as well as facilitated apoptosis of NSCLC cells. Besides, in vivo animal experiments further confirmed that CHIP restrained tumorigenic ability and improved radiosensitivity of NSCLC cells by inhibiting PBK/ERK axis. Collectively, CHIP suppressed stem cell properties and radioresistance of NSCLC cells by inhibiting PBK/ERK axis, therefore offering a potential therapeutic target for enhancing efficacy of radiotherapy.
Collapse
Affiliation(s)
- Bo Tan
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui, Zhengzhou, 450008, Henan, China.
| | - Jingwei Zhang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui, Zhengzhou, 450008, Henan, China
| | - Wen Wang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui, Zhengzhou, 450008, Henan, China
| | - Haibo Ma
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yuanyuan Yang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui, Zhengzhou, 450008, Henan, China
| |
Collapse
|
29
|
Chen W, Yang W, Zhang C, Liu T, Zhu J, Wang H, Li T, Jin A, Ding L, Xian J, Tian T, Pan B, Guo W, Wang B. Modulation of the p38 MAPK Pathway by Anisomycin Promotes Ferroptosis of Hepatocellular Carcinoma through Phosphorylation of H3S10. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6986445. [PMID: 36466092 PMCID: PMC9715334 DOI: 10.1155/2022/6986445] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/27/2022] [Accepted: 10/08/2022] [Indexed: 07/25/2023]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignant tumor worldwide. Ferroptosis is emerging as an effective target for tumor treatment as it has been shown to potentiate cell death in some malignancies. However, it remains unclear whether histone phosphorylation events, an epigenetic mechanism that regulates transcriptional expression, are involved in ferroptosis. Our study found that supplementation with anisomycin, an agonist of p38 mitogen-activated protein kinase (MAPK), induced ferroptosis in HCC cells, and the phosphorylation of histone H3 on serine 10 (p-H3S10) was participated in anisomycin-induced ferroptosis. To investigate the anticancer effects of anisomycin-activated p38 MAPK in HCC, we analyzed cell viability, colony formation, cell death, and cell migration in Hep3B and HCCLM3 cells. The results showed that anisomycin could significantly suppress HCC cell colony formation and migration and induce HCC cell death. The hallmarks of ferroptosis, such as abnormal accumulation of iron and elevated levels of lipid peroxidation and malondialdehyde, were detected to confirm the ability of anisomycin to promote ferroptosis. Furthermore, coincubation with SB203580, an inhibitor of activated p38 MAPK, partially rescued anisomycin-induced ferroptosis. And the levels of p-p38 MAPK and p-H3S10 were successively increased by anisomycin treatment. The relationship between p-H3S10 and ferroptosis was revealed by ChIP sequencing. The reverse transcription PCR and immunofluorescence results showed that NCOA4 was upregulated both in mRNA and protein levels after anisomycin treatment. And by C11-BODIPY staining, we found that anisomycin-induced lipid reactive oxygen species was reduced after NCOA4 knockdown. In conclusion, the anisomycin-activated p38 MAPK promoted ferroptosis of HCC cells through H3S10 phosphorylation.
Collapse
Affiliation(s)
- Wei Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Te Liu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Zhu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tong Li
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Anli Jin
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Ding
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingrong Xian
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tongtong Tian
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Baishen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Shanghai Zhongshan Hospital, Fudan University, Shanghai, China
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Shanghai Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Liu Z, Li X, Gao J, Yin P, Teng Y, Yu P. The therapeutic inhibition of topoisomerase inhibitor and crizotinib combination in EGFR wild and mutant lung cancer cells. Biochem Pharmacol 2022; 205:115294. [DOI: 10.1016/j.bcp.2022.115294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/25/2022] [Accepted: 10/03/2022] [Indexed: 11/02/2022]
|
31
|
Kudaravalli S, den Hollander P, Mani SA. Role of p38 MAP kinase in cancer stem cells and metastasis. Oncogene 2022; 41:3177-3185. [PMID: 35501462 PMCID: PMC9166676 DOI: 10.1038/s41388-022-02329-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 01/01/2023]
Abstract
Therapeutic resistance and metastatic progression are responsible for the majority of cancer mortalities. In particular, the development of resistance is a significant barrier to the efficacy of cancer treatments such as chemotherapy, radiotherapy, targeted therapies, and immunotherapies. Cancer stem cells (CSCs) underlie treatment resistance and metastasis. p38 mitogen-activated protein kinase (p38 MAPK) is downstream of several CSC-specific signaling pathways, and it plays an important role in CSC development and maintenance and contributes to metastasis and chemoresistance. Therefore, the development of therapeutic approaches targeting p38 can sensitize tumors to chemotherapy and prevent metastatic progression.
Collapse
Affiliation(s)
- Sriya Kudaravalli
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Rice University, Houston, TX, 77030, USA
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
32
|
Chen W, Tang D, Lin J, Huang X, Lin S, Shen G, Dai Y. Exosomal circSHKBP1 participates in non-small cell lung cancer progression through PKM2-mediated glycolysis. Mol Ther Oncolytics 2022; 24:470-485. [PMID: 35229026 PMCID: PMC8844869 DOI: 10.1016/j.omto.2022.01.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/24/2022] [Indexed: 11/04/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has a high morbidity and mortality, and it is imperative to explore the latent pathogenesis mechanism of NSCLC progression to find potential prognostic biomarkers and therapeutic targets. The present study aimed to explore the biological function of circSHKBP1 in NSCLC. circSHKBP1 was found to be upregulated in NSCLC tissues and cell lines and was enriched in exosomes derived from NSCLC cells. Exosomal circSHKBP1 enhanced the proliferation, migration, invasion, and stemness of NSCLC cells. miRNA-1294 was identified as a target for circSHKBP1, and circSHKBP1 upregulated PKM2 expression by sponging miR-1294. Exosomal circSHKBP1 regulated glycolysis through PKM2 in a HIF-1α-dependent manner in NSCLC cells and promoted M2 polarization and macrophage recruitment. Moreover, exosomal circSHKBP1 promoted NSCLC cell growth, metastasis, and M2 infiltration in vivo. Thus, exosomal circSHKBP1 participated in the progression of NSCLC via the miR-1294/PKM2 axis. circSHKBP1 may be potential biomarker for the diagnosis and treatment of NSCLC.
Collapse
|
33
|
Elhassan RM, Hou X, Fang H. Recent advances in the development of allosteric protein tyrosine phosphatase inhibitors for drug discovery. Med Res Rev 2021; 42:1064-1110. [PMID: 34791703 DOI: 10.1002/med.21871] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 09/26/2021] [Accepted: 10/24/2021] [Indexed: 01/07/2023]
Abstract
Protein tyrosine phosphatases (PTPs) superfamily catalyzes tyrosine de-phosphorylation which affects a myriad of cellular processes. Imbalance in signal pathways mediated by PTPs has been associated with development of many human diseases including cancer, metabolic, and immunological diseases. Several compelling evidence suggest that many members of PTP family are novel therapeutic targets. However, the clinical development of conventional PTP-based active-site inhibitors originally was hampered by the poor selectivity and pharmacokinetic properties. In this regard, PTPs has been widely dismissed as "undruggable." Nonetheless, allosteric modulation has become increasingly an influential and alternative approach that can be exploited for drug development against PTPs. Unlike active-site inhibitors, allosteric inhibitors exhibit a remarkable target-selectivity, drug-likeness, potency, and in vivo activity. Intriguingly, there has been a high interest in novel allosteric PTPs inhibitors within the last years. In this review, we focus on the recent advances of allosteric inhibitors that have been explored in drug discovery and have shown an excellent result in the development of PTPs-based therapeutics. A special emphasis is placed on the structure-activity relationship and molecular mechanistic studies illustrating applications in chemical biology and medicinal chemistry.
Collapse
Affiliation(s)
- Reham M Elhassan
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Xuben Hou
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Hao Fang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| |
Collapse
|
34
|
Turdo A, D'Accardo C, Glaviano A, Porcelli G, Colarossi C, Colarossi L, Mare M, Faldetta N, Modica C, Pistone G, Bongiorno MR, Todaro M, Stassi G. Targeting Phosphatases and Kinases: How to Checkmate Cancer. Front Cell Dev Biol 2021; 9:690306. [PMID: 34778245 PMCID: PMC8581442 DOI: 10.3389/fcell.2021.690306] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/04/2021] [Indexed: 12/21/2022] Open
Abstract
Metastatic disease represents the major cause of death in oncologic patients worldwide. Accumulating evidence have highlighted the relevance of a small population of cancer cells, named cancer stem cells (CSCs), in the resistance to therapies, as well as cancer recurrence and metastasis. Standard anti-cancer treatments are not always conclusively curative, posing an urgent need to discover new targets for an effective therapy. Kinases and phosphatases are implicated in many cellular processes, such as proliferation, differentiation and oncogenic transformation. These proteins are crucial regulators of intracellular signaling pathways mediating multiple cellular activities. Therefore, alterations in kinases and phosphatases functionality is a hallmark of cancer. Notwithstanding the role of kinases and phosphatases in cancer has been widely investigated, their aberrant activation in the compartment of CSCs is nowadays being explored as new potential Achille's heel to strike. Here, we provide a comprehensive overview of the major protein kinases and phosphatases pathways by which CSCs can evade normal physiological constraints on survival, growth, and invasion. Moreover, we discuss the potential of inhibitors of these proteins in counteracting CSCs expansion during cancer development and progression.
Collapse
Affiliation(s)
- Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Caterina D'Accardo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Antonino Glaviano
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Gaetana Porcelli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Cristina Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), Catania, Italy
| | - Lorenzo Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), Catania, Italy
| | - Marzia Mare
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), Catania, Italy
| | | | - Chiara Modica
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Giuseppe Pistone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Maria Rita Bongiorno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy.,Azienda Ospedaliera Universitaria Policlinico (AOUP), Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| |
Collapse
|
35
|
Xia M, Cao H, Zheng J, Yao Y, Xu F, Lu G, Ma Y, Zhou J. A novel stilbene derivative (GMQ3) suppressed proliferation and induced apoptosis in lung cancer via the p38-MAPK/SIRT1 pathway. Biochem Pharmacol 2021; 193:114808. [PMID: 34678220 DOI: 10.1016/j.bcp.2021.114808] [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: 08/03/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Lung cancer is the primary cause of cancer-related mortality worldwide. The anticancer effect of stilbene has been noted in various tumor types. GMQ3, which has a stilbene-mimicking skeleton, is a novel small-molecule compound with promising antitumor activity. Our results revealed that GMQ3 not only suppressed cell proliferation and cell migration of lung cancer cells but also led to G1 phase cell cycle arrest and triggered caspase-dependent apoptosis. Furthermore, investigation of the molecular mechanism showed that GMQ3 could inhibited proliferation and induced apoptosis via the p38-MAPK/SIRT1 pathway both in vitro and in vivo. Xenograft tumor mouse models showed that GMQ3 significantly inhibited tumor growth in vivo without affecting body weight. Our findings indicated that GMQ3 exerts a strong anticancer action by suppressing cell proliferation, inhibiting cell migration and inducing cell apoptosis. Moreover, the efficacy of GMQ3 was enhanced in the presence of CDK4/6 inhibitor Abemaciclib. We conclude that GMQ3 is a promising agent with potential for lung cancer.
Collapse
Affiliation(s)
- Mengling Xia
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - He Cao
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yinan Yao
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Fei Xu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Guohua Lu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yongmin Ma
- Institute of Advanced Studies and School of Pharmaceutical and Chemical Engineering, Taizhou University, Taizhou, China.
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
36
|
Ju H, Hu Z, Wei D, Huang J, Zhang X, Rui M, Li Z, Zhang X, Hu J, Guo W, Ren G. A novel intronic circular RNA, circGNG7, inhibits head and neck squamous cell carcinoma progression by blocking the phosphorylation of heat shock protein 27 at Ser78 and Ser82. Cancer Commun (Lond) 2021; 41:1152-1172. [PMID: 34498800 PMCID: PMC8626595 DOI: 10.1002/cac2.12213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/02/2021] [Accepted: 08/31/2021] [Indexed: 01/11/2023] Open
Abstract
Background There is increasing evidence that circular RNAs (circRNAs) play a significant role in pathological processes including tumorigenesis. In contrast to exonic circRNAs, which are the most frequently reported circRNAs in cancer so far, the studies of intronic circRNAs have been greatly lagged behind. Here, we aimed to investigate the regulatory role of intronic circRNAs in head and neck squamous cell carcinoma (HNSCC). Methods We conducted whole‐transcriptome sequencing with four pairs of primary tumor tissues and adjacent normal tissues from HNSCC patients. Then, we characterized circGNG7 expression in HNSCC tissues and cell lines and explored its association with the prognosis of HNSCC patients. We also identified interactions between circGNG7 and functional proteins, which alter downstream signaling that regulate HNSCC progression. Results In this study, we identified a new intronic circRNA, circGNG7, and validated its functional roles in HNSCC progression. CircGNG7 was predominately localized to the cytoplasm, and its expression was downregulated in both HNSCC tissues andCAL27, CAL33, SCC4, SCC9, HN6, and HN30 cells. Low expression of circGNG7 was significantly correlated with poor prognosis in HNSCC patients. Consistent with this finding, overexpression of circGNG7 strongly inhibited tumor cell proliferation, colony formation, in vitro migration, and in vivo tumor growth. Mechanistically, the expression of circGNG7 in HNSCC cells was regulated by the transcription factor SMAD family member 4 (SMAD4). Importantly, we discovered that circGNG7 could bind to serine residues 78 and 82 of the functional heat shock protein 27 (HSP27), occupying its phosphorylation sites and hindering its phosphorylation, which reduced HSP27‐JNK/P38 mitogen‐activated protein kinase (MAPK) oncogenic signaling. Downregulation of circGNG7 expression in HNSCC increased HSP27‐JNK/P38 MAPK signaling and promoted tumor progression. Conclusions Our results revealed that a new intronic circRNA, circGNG7, functions as a strong tumor suppressor and that circGNG7/HSP27‐JNK/P38 MAPK signaling is a novel mechanism by which HNSCC progression can be controlled.
Collapse
Affiliation(s)
- Houyu Ju
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China.,National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China
| | - Zhenrong Hu
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,School of Stomatology, Weifang Medical University, Weifang, Shandong, 261053, P. R. China
| | - Dongliang Wei
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China.,National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China
| | - Jinyun Huang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,School of Stomatology, Weifang Medical University, Weifang, Shandong, 261053, P. R. China
| | - Xinyi Zhang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,School of Stomatology, Weifang Medical University, Weifang, Shandong, 261053, P. R. China
| | - Mengyu Rui
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China.,National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China
| | - Zhi Li
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,School of Stomatology, Weifang Medical University, Weifang, Shandong, 261053, P. R. China
| | - Xiaomeng Zhang
- National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillo-facial Implantology, School of Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200011, P. R. China
| | - Jingzhou Hu
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China.,National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China
| | - Wei Guo
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China.,National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China
| | - Guoxin Ren
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China.,National Clinical Research Center of Stomatology, Shanghai, 200011, P. R. China
| |
Collapse
|
37
|
Xu G, Yang Z, Ding Y, Liu Y, Zhang L, Wang B, Tang M, Jing T, Jiao K, Xu X, Chen Z, Xiang L, Xu C, Fu Y, Zhao X, Jin W, Liu Y. The deubiquitinase USP16 functions as an oncogenic factor in K-RAS-driven lung tumorigenesis. Oncogene 2021; 40:5482-5494. [PMID: 34294846 DOI: 10.1038/s41388-021-01964-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
K-RAS mutation and molecular alterations of its surrogates function essentially in lung tumorigenesis and malignant progression. However, it remains elusive how tumor-promoting and deleterious events downstream of K-RAS signaling are coordinated in lung tumorigenesis. Here, we show that USP16, a deubiquitinase involved in various biological processes, functions as a promoter for the development of K-RAS-driven lung tumor. Usp16 deletion significantly attenuates K-rasG12D-mutation-induced lung tumorigenesis in mice. USP16 upregulation upon RAS activation averts reactive oxygen species (ROS)-induced p38 activation that would otherwise detrimentally influence the survival and proliferation of tumor cells. In addition, USP16 interacts with and deubiquitinates JAK1, and thereby promoting lung tumor growth by augmenting JAK1 signaling. Therefore, our results reveal that USP16 functions critically in the K-RAS-driven lung tumorigenesis through modulating the strength of p38 and JAK1 signaling.
Collapse
Affiliation(s)
- Guiqin Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaojuan Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yizong Ding
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Boshi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Tang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiantian Jing
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Jiao
- Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Xiaoli Xu
- Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Zehong Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lvzhu Xiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujie Fu
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Zhao
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weilin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, People's Republic of China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
38
|
Cancer stem cell phosphatases. Biochem J 2021; 478:2899-2920. [PMID: 34319405 DOI: 10.1042/bcj20210254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022]
Abstract
Cancer stem cells (CSCs) are involved in the initiation and progression of human malignancies by enabling cancer tissue self-renewal capacity and constituting the therapy-resistant population of tumor cells. However, despite the exhausting characterization of CSC genetics, epigenetics, and kinase signaling, eradication of CSCs remains an unattainable goal in most human malignancies. While phosphatases contribute equally with kinases to cellular phosphoregulation, our understanding of phosphatases in CSCs lags severely behind our knowledge about other CSC signaling mechanisms. Many cancer-relevant phosphatases have recently become druggable, indicating that further understanding of the CSC phosphatases might provide novel therapeutic opportunities. This review summarizes the current knowledge about fundamental, but yet poorly understood involvement of phosphatases in the regulation of major CSC signaling pathways. We also review the functional roles of phosphatases in CSC self-renewal, cancer progression, and therapy resistance; focusing particularly on hematological cancers and glioblastoma. We further discuss the small molecule targeting of CSC phosphatases and their therapeutic potential in cancer combination therapies.
Collapse
|
39
|
Storchova R, Burdova K, Palek M, Medema RH, Macurek L. A novel assay for screening WIP1 phosphatase substrates in nuclear extracts. FEBS J 2021; 288:6035-6051. [PMID: 33982878 DOI: 10.1111/febs.15965] [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: 02/26/2021] [Revised: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 11/30/2022]
Abstract
Upon exposure to genotoxic stress, cells activate DNA damage response (DDR) that coordinates DNA repair with a temporal arrest in the cell cycle progression. DDR is triggered by activation of ataxia telangiectasia mutated/ataxia telangiectasia and Rad3-related protein kinases that phosphorylate multiple targets including tumor suppressor protein tumor suppressor p53 (p53). In addition, DNA damage can activate parallel stress response pathways [such as mitogen-activated protein kinase p38 alpha (p38)/MAPK-activated protein kinase 2 (MK2) kinases] contributing to establishing the cell cycle arrest. Wild-type p53-induced phosphatase 1 (WIP1) controls timely inactivation of DDR and is needed for recovery from the G2 checkpoint by counteracting the function of p53. Here, we developed a simple in vitro assay for testing WIP1 substrates in nuclear extracts. Whereas we did not detect any activity of WIP1 toward p38/MK2, we confirmed p53 as a substrate of WIP1. Inhibition or inactivation of WIP1 in U2OS cells increased phosphorylation of p53 at S15 and potentiated its acetylation at K382. Further, we identified Deleted in breast cancer gene 1 (DBC1) as a new substrate of WIP1 but surprisingly, depletion of DBC1 did not interfere with the ability of WIP1 to regulate p53 acetylation. Instead, we have found that WIP1 activity suppresses p53-K382 acetylation by inhibiting the interaction between p53 and the acetyltransferase p300. Newly established phosphatase assay allows an easy comparison of WIP1 ability to dephosphorylate various proteins and thus contributes to identification of its physiological substrates.
Collapse
Affiliation(s)
- Radka Storchova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Kamila Burdova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Matous Palek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - René H Medema
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
40
|
Synergy between vinorelbine and afatinib in the inhibition of non-small cell lung cancer progression by EGFR and p53 signaling pathways. Biomed Pharmacother 2020; 134:111144. [PMID: 33360044 DOI: 10.1016/j.biopha.2020.111144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 11/23/2022] Open
Abstract
Currently, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) were approved for the treatment of non-small cell lung cancer (NSCLC) patients harboring EGFR mutation. However, some lung cancer patients fail to respond and eventually develop drug resistance. Therefore, new therapeutic strategies are needed to improve the outcomes for substantial clinical benefit. Here we aimed to explore the combination of vinorelbine with the second EGFR-TKI afatinib in NSCLC cells with or without EGFR mutation. The three cells of H1975, HCC827, and H460 were assessed for the combination of vinorelbine and afatinib. Vinorelbine combined with afatinib synergistically inhibited the three lung cancer cells growth without aggravating adverse effect on the normal lung cells. The combination of low doses of vinorelbine and afatinib suppressed the cancer cell proliferation by cell colony formation assay and significantly induced cell apoptosis. The anti-apoptotic proteins Bcl-xL and Bcl-2 showed significant reduction after the drug combination treatment, while the pro-apoptotic protein Bax as well as apoptosis indicators cytochrome C and cleaved PARP were observed a notable increasing. EGFR downstream pathways including AKT, ERK, JNK, and p38 were highly active and p53 was inactive in the three lung cancer cells, favoring tumor growth. The low doses of vinorelbine plus afatinib blocked the phosphorylation of AKT, ERK, JNK, and p38, but restored the expression of p53. Our findings suggested that the combination of vinorelbine and afatinib could be recommended as a therapeutic regimen for treatment of NSCLC with or without EGFR mutation.
Collapse
|
41
|
Small Heat Shock Proteins in Cancers: Functions and Therapeutic Potential for Cancer Therapy. Int J Mol Sci 2020; 21:ijms21186611. [PMID: 32927696 PMCID: PMC7555140 DOI: 10.3390/ijms21186611] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Small heat shock proteins (sHSPs) are ubiquitous ATP-independent chaperones that play essential roles in response to cellular stresses and protein homeostasis. Investigations of sHSPs reveal that sHSPs are ubiquitously expressed in numerous types of tumors, and their expression is closely associated with cancer progression. sHSPs have been suggested to control a diverse range of cancer functions, including tumorigenesis, cell growth, apoptosis, metastasis, and chemoresistance, as well as regulation of cancer stem cell properties. Recent advances in the field indicate that some sHSPs have been validated as a powerful target in cancer therapy. In this review, we present and highlight current understanding, recent progress, and future challenges of sHSPs in cancer development and therapy.
Collapse
|