1
|
Feng Y, Zhu Z, Zhao S, Jiang X, Zhang W, Xu Z. Bioorthogonally Activatable Photosensitizer for NIR Fluorescence Imaging-Guided Highly Selective Elimination of Senescent Tumor Cells and Chemotherapy Enhancement. Bioconjug Chem 2025. [PMID: 40329576 DOI: 10.1021/acs.bioconjchem.5c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Chemotherapy is a primary modality in cancer treatment, but it may induce cellular senescence, which in turn triggers the release of senescence-associated secretory phenotypes (SASPs) that promote tumor growth and metastasis. To selectively identify senescent cells and mitigate their negative impact on cancer therapy, herein, we have developed a β-galactosidase (β-Gal)-activated and self-immobilizing photosensitizer CyGF-DBCO-T. This photosensitizer can be selectively activated and fluorescently label proteins in situ within senescent cells, enabling near-infrared (NIR) fluorescence imaging-guided photodynamic therapy (PDT) for the precise ablation of these cells. First, we developed an activatable NIR fluorescent probe CyGF-N3 that can specifically in situ label senescent cells. Subsequently, DBCO-T with free radicals underwent a bioorthogonal click reaction with activated CyGF-N3 in senescent cells to generate the photosensitizer CyO-DBCO-T. Under light irradiation, CyO-DBCO-T generated singlet oxygen (1O2) in situ, thereby enabling precise PDT with fluorescence guidance and photoactivation. Both CyGF-N3 and DBCO-T were encapsulated in biotinylated liposomes (CyGF-N3@LIP-B and DBCO-T@LIP-B), which enhanced their water solubility, tumor targeting, and in vivo circulation time. This promoted the accumulation of the probes in senescent tumor cells, thus enabling intense fluorescence imaging of tumor senescence regions in mice and enhancing the efficacy of PDT. This dual-module strategy, guided by fluorescence imaging for PDT, has achieved selective identification and precise ablation of senescent tumor cells in a chemotherapy-induced senescence model, effectively alleviating chemotherapy resistance and suppressing tumor growth.
Collapse
Affiliation(s)
- Yun Feng
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Zifan Zhu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Shirui Zhao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Xingyu Jiang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Wen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai 200062, China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| |
Collapse
|
2
|
Wu Y, Chen L, Pi D, Cui J, Liang Y, Wu P, Ouyang M, Zuo Q. Saikosaponin A induces cellular senescence in triple-negative breast cancer by inhibiting the PI3K/Akt signalling pathway. Front Pharmacol 2025; 16:1532579. [PMID: 40351423 PMCID: PMC12062077 DOI: 10.3389/fphar.2025.1532579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Background Breast cancer has now become the most prevalent cancer worldwide. Existing therapeutic agents are generally accompanied by significant side effects. Here, we highlight Saikosaponin A (SSA), a promising natural metabolite characterized by low toxicity, demonstrating significant efficacy against breast cancer through the induction of cellular senescence. Methods The antitumor property of SSA was determined via MTT colorimetric assay, 5-ethynyl-2'-deoxyuridine (EdU) incorporation assay, colony formation, and propidium iodide (PI) staining in vitro, as well as xenograft in vivo model. A network approach was used to predict potential targets of SSA reevant for a potential anti-tumor effect and verified through senescence-associated β-galactosidase (SA-β-gal), flow-cytometry analysis, RT-PCR, Western blotting, and immuno-histochemistry assay. Results SSA significantly suppressed proliferation and triggered cell cycle arrest of SUM159PT and MDA-MB-231 cells. Revealed by network analysis, cellular senescence, and phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway were implemented in the anti-tumor effects of SSA. SSA-stimulated senescence was associated with increased ROS production, distinct senescence-associated secretory phenotype (SASP), and restricted PI3K/Akt signaling, as well as p21 and p53 accumulation. Furthermore, SSA displayed inhibitory effects on tumor growth with minimal toxicity in animal studies, accompanied by activated biomarkers of cellular senescence and decreased expression of p-Akt and p-PI3K. Conclusion Taken together, based on the preliminary results of network analysis and further experimental validation, this study revealed that SSA significantly induced cell cycle arrest and senescence, and the inhibition of ROS-mediated PI3K/Akt pathway may be the potential mechanism in this process.
Collapse
Affiliation(s)
- Yingchao Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Liushan Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| | - Dajin Pi
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jiaqi Cui
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yuqi Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| | - Peng Wu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mingzi Ouyang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Qian Zuo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Nehme J, Maassen S, Bravaccini S, Zanoni M, Gianni C, De Giorgi U, Soto-Gamez A, Altulea A, Gheorghe T, Wang B, Demaria M. Pharmacological CDK4/6 inhibition promotes vulnerability to lysosomotropic agents in breast cancer. EMBO J 2025; 44:1921-1942. [PMID: 39930269 PMCID: PMC11961731 DOI: 10.1038/s44318-025-00371-x] [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: 07/04/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/26/2025] Open
Abstract
Breast cancer is a leading cause of mortality worldwide. Pharmacological inhibitors of cyclin-dependent kinases (CDK) 4 and 6 (CDK4/6i) inhibit breast cancer growth by inducing a senescent-like state. However, the long-term treatment efficacy remains limited by the development of drug resistance, so clearance of senescent-like cancer cells may extend the durability of treatment. However, we show here that while CDK4/6i-treated breast cancer cells exhibit various senescence-associated phenotypes, they remain insensitive to common senolytic compounds. By searching for novel vulnerabilities, we identify a significantly increased lysosomal mass and altered lysosomal structure across various breast cancer cell types upon exposure to CDK4/6i in preclinical systems and clinical specimens. We demonstrate that these CDK4/6i-induced lysosomal alterations render breast cancer cells sensitive to lysosomotropic agents, such as L-leucyl-L-leucine methyl ester (LLOMe) and salinomycin. Importantly, sequential treatment with CDK4/6i and lysosomotropic agents effectively reduces the growth of both hormone receptor-positive (HR+) and subsets of triple-negative breast cancer (TNBC) cells in vivo. This sequential therapeutic strategy offers a promising approach to eliminate CDK4/6i-induced senescent(-like) cells, potentially reducing tumor recurrence and enhancing the overall efficacy of breast cancer therapy.
Collapse
Affiliation(s)
- Jamil Nehme
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Sjors Maassen
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Zanoni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Caterina Gianni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ugo De Giorgi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Abel Soto-Gamez
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Abdullah Altulea
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Teodora Gheorghe
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Boshi Wang
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands.
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands.
| |
Collapse
|
4
|
Czajkowski K, Herbet M, Murias M, Piątkowska-Chmiel I. Senolytics: charting a new course or enhancing existing anti-tumor therapies? Cell Oncol (Dordr) 2025; 48:351-371. [PMID: 39633108 PMCID: PMC11996976 DOI: 10.1007/s13402-024-01018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Cell senescence is a natural response within our organisms. Initially, it was considered an effective anti-tumor mechanism. However, it is now believed that while cell senescence initially acts as a robust barrier against tumor initiation, the subsequent accumulation of senescent cells can paradoxically promote cancer recurrence and cause damage to neighboring tissues. This intricate balance between cell proliferation and senescence plays a pivotal role in maintaining tissue homeostasis. Moreover, senescence cells secrete many bioactive molecules collectively termed the senescence-associated secretory phenotype (SASP), which can induce chronic inflammation, alter tissue architecture, and promote tumorigenesis through paracrine signaling. Among the myriads of compounds, senotherapeutic drugs have emerged as exceptionally promising candidates in anticancer treatment. Their ability to selectively target senescent cells while sparing healthy tissues represents a paradigm shift in therapeutic intervention, offering new avenues for personalized oncology medicine. Senolytics have introduced new therapeutic possibilities by enabling the targeted removal of senescent cells. As standalone agents, they can clear tumor cells in a senescent state and, when combined with chemo- or radiotherapy, eliminate residual senescent cancer cells after treatment. This dual approach allows for the intentional use of lower-dose therapies or the removal of unintended senescent cells post-treatment. Additionally, by targeting non-cancerous senescent cells, senolytics may help reduce tumor formation risk, limit recurrence, and slow disease progression. This article examines the mechanisms of cellular senescence, its role in cancer treatment, and the importance of senotherapy, with particular attention to the therapeutic potential of senolytic drugs.
Collapse
Affiliation(s)
- Konrad Czajkowski
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Mariola Herbet
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, Poznań, Poland
| | - Iwona Piątkowska-Chmiel
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
5
|
Zhang J, Zhang S, Cheng C, Zhu C, Wang T, Tang L, Lou J, Li X, Wang H, Hu F, Sun M, Zhang K, Yu F. Targeting senescence with radioactive 223Ra/Ba SAzymes enables senolytics-unlocked One-Two punch strategy to boost anti-tumor immunotherapy. Biomaterials 2025; 315:122915. [PMID: 39461062 DOI: 10.1016/j.biomaterials.2024.122915] [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: 08/15/2024] [Revised: 10/13/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Senescent cells are characterized by a persistent cessation of their cell cycle, rendering them valuable targets for anti-tumor strategies in cancer treatment. Numerous studies have explored induced senescence as a promising approach in tumor therapy. Nevertheless, these treatments often come with drawbacks, including adverse side effects and weaker senescence-inducing effects. To address these challenges, we synthesized 223Ra/Ba single-atom nanozyme (SAzyme), wherein Ba SAzyme acts concurrently as a carrier for 223RaCl2, facilitating targeted delivery and minimizing side effects. The 223Ra/Ba SAzyme complex enhances various enzyme-mimicking functions, including catalase (CAT) and peroxidase (POD) activities. Importantly, 223Ra/Ba SAzyme induces cellular senescence and boost anti-tumor immunity. The persistent presence of a senescence-associated secretory phenotype (SASP) in the tumor microenvironment presents risks of immune suppression and tumor recurrence, which can be effectively mitigated by senolytics. As a result, 223Ra/Ba SAzyme were combined with anti-PD-L1 checkpoint blockade to achieve a one-two punch therapy, wherein 223Ra/Ba SAzyme exploits senescence followed by anti-PD-L1 therapy to eradicate senescent cells. This one-two punch strategy approach presents a straightforward and potent intervention for both primary tumors and distant tumor.
Collapse
Affiliation(s)
- Jiajia Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China; Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China; Central Laboratory and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, China
| | - Shenghong Zhang
- Department of Nuclear Medicine the First Affiliated Hospital of Navy Medical University (Changhai Hospital), No. 168 Changhai Road, Shanghai, 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine the First Affiliated Hospital of Navy Medical University (Changhai Hospital), No. 168 Changhai Road, Shanghai, 200433, China
| | - Chunyan Zhu
- Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China; Central Laboratory and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, China
| | - Taixia Wang
- Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China; Central Laboratory and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, China
| | - Linglin Tang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai, 200127, China
| | - Jingjing Lou
- Department of Nuclear Medicine, Pudong Medical Center, Fudan University, No. 2800 Gongwei Road, Shanghai, 201399, China
| | - Xian Li
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China; Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China
| | - Hai Wang
- Central Laboratory and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, China
| | - Fan Hu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China; Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China
| | - Ming Sun
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China; Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China
| | - Kun Zhang
- Central Laboratory and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, China.
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China; Institute of Nuclear Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai, 200072, China.
| |
Collapse
|
6
|
Xue Y, Chen T, Ma Z, Pu X, Xu J, Zhai S, Du X, Ji Y, Simon MC, Zhai W, Xue W. Osalmid sensitizes clear cell renal cell carcinoma to navitoclax through a STAT3/BCL-XL pathway. Cancer Lett 2025; 613:217514. [PMID: 39894195 DOI: 10.1016/j.canlet.2025.217514] [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: 08/22/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common and lethal urinary malignancy characterized by its resistance to apoptosis. Despite the emerging treatment options available for ccRCC, only a small proportion of patients achieve long-term survival benefits. Previous studies have demonstrated that inducing tumor cell senescence, followed by treatment using senolytics, represents a potential strategy for triggering tumor cell apoptosis. However, it remains unclear whether this strategy is suitable for the treatment of ccRCC. Using the whole-genome CRISPR screening database Dependency Map portal (DepMap), we identified ribonucleotide reductase family member 2 (RRM2), which catalyzes the conversion of ribonucleotides to deoxyribonucleotides (dNTPs), as an essential targetable gene for ccRCC. Herein, we report that the combination of the choleretic drug osalmid targeting RRM2 and the senolytic compound navitoclax targeting BCL-XL represents a novel therapeutic approach for ccRCC. Furthermore, we have validated this approach across a panel of human ccRCC cells with different genetic backgrounds and multiple preclinical models, including cell line-derived xenografts (CDX), patient-derived xenografts (PDX), and patient-derived organoids (PDO). Mechanistically, osalmid-mediated inhibition of dNTPs generation induces cellular senescence in ccRCC, concomitant with STAT3 activation and upregulation of BCL-XL, thus rendering these cells vulnerable to navitoclax, which targets the BCL-2 protein family.
Collapse
Affiliation(s)
- Yizheng Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tianyi Chen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zehua Ma
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550001, China
| | - Xinyuan Pu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Junyao Xu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Shuanfeng Zhai
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yiyi Ji
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, Chicago, IL, 60637, USA, Howard Hughes Medical Institute
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wei Zhai
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China; Shanghai Immune Therapy Institute State, Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
7
|
Zhang X, Gao Y, Zhang S, Wang Y, Du Y, Hao S, Ni T. The Regulation of Cellular Senescence in Cancer. Biomolecules 2025; 15:448. [PMID: 40149983 PMCID: PMC11940315 DOI: 10.3390/biom15030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Cellular senescence is a stable state of cell cycle arrest caused by telomere shortening or various stresses. After senescence, cells cease dividing and exhibit many age-related characteristics. Unlike the halted proliferation of senescence cells, cancer cells are considered to have unlimited growth potential. When cells display senescence-related features, such as telomere loss or stem cell failure, they can inhibit tumor development. Therefore, inducing cells to enter a senescence state can serve as a barrier to tumor cell development. However, many recent studies have found that sustained senescence of tumor cells or normal cells under certain circumstances can exert environment-dependent effects of tumor promotion and inhibition by producing various cytokines. In this review, we first introduce the causes and characteristics of induced cellular senescence, analyze the senescence process of immune cells and cancer cells, and then discuss the dual regulatory role of cell senescence on tumor growth and senescence-induced therapies targeting cancer cells. Finally, we discuss the role of senescence in tumor progression and treatment opportunities, and propose further studies on cellular senescence and cancer therapy.
Collapse
Affiliation(s)
- Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Siyu Zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China;
| | - Yixiong Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| |
Collapse
|
8
|
Yasuda T, Alan Wang Y. Immune therapeutic strategies for the senescent tumor microenvironment. Br J Cancer 2025; 132:237-244. [PMID: 39468331 PMCID: PMC11790855 DOI: 10.1038/s41416-024-02865-7] [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: 06/26/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/30/2024] Open
Abstract
Senescent cells can either to promote immunosuppressive tumor microenvironment or facilitate immune surveillance. Despite the revolutionary impact of cancer immunotherapy, durable responses in solid tumors, particularly in advanced stages, remain limited. Recent studies have shed light on the influence of senescent status within the tumor microenvironment (TME) on therapy resistance and major efforts are needed to overcome these challenges. This review summarizes recent advancements in targeting cellular senescence, with a particular focus on immunomodulatory approaches on the hallmarks of cellular senescence.
Collapse
Affiliation(s)
- Tadahito Yasuda
- Brown Center for Immunotherapy, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA.
| | - Y Alan Wang
- Brown Center for Immunotherapy, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center Indianapolis, Indianapolis, USA
| |
Collapse
|
9
|
Hwang HJ, Kang D, Shin J, Jung J, Ko S, Jung KH, Hong SS, Park JE, Oh MJ, An HJ, Yang WH, Ko YG, Cha JH, Lee JS. Therapy-induced senescent cancer cells contribute to cancer progression by promoting ribophorin 1-dependent PD-L1 upregulation. Nat Commun 2025; 16:353. [PMID: 39753537 PMCID: PMC11699195 DOI: 10.1038/s41467-024-54132-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/02/2024] [Indexed: 01/06/2025] Open
Abstract
Conventional chemotherapy- and radiotherapy-induced cancer senescence, which is characterized by poor proliferation, drug resistance, and senescence-associated secretory phenotype, has gained attention as contributing to cancer relapse and the development of an immunosuppressive tumor microenvironment. However, the association between cancer senescence and anti-tumor immunity is not fully understood. Here, we demonstrate that senescent cancer cells increase the level of PD-L1 by promoting its transcription and glycosylation. We identify ribophorin 1 as a key regulator of PD-L1 glycosylation during cancer senescence. Ribophorin 1 depletion reduces this elevated level of PD-L1 through the ER-lysosome-associated degradation pathway, thereby increasing the susceptibility of senescent cancer cells to T-cell-mediated killing. Consistently, ribophorin 1 depletion suppresses tumor growth by decreasing PD-L1 levels and boosting cytotoxic T lymphocyte activity in male mice. Moreover, ribophorin 1-targeted or anti-PD-1 therapy reduces the number of senescent cancer cells in irradiated tumors and suppresses cancer recurrence through the activation of cytotoxic T lymphocytes. These results provide crucial insights into how senescent cancer cells can escape T-cell immunity following cancer treatment and thereby contribute to cancer recurrence. Our findings also highlight the therapeutic promise of targeting senescent cancer cells for cancer treatment.
Collapse
Affiliation(s)
- Hyun Jung Hwang
- Department of Molecular Medicine, Inha University, Incheon, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University, Incheon, Republic of Korea
| | - Donghee Kang
- Department of Molecular Medicine, Inha University, Incheon, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University, Incheon, Republic of Korea
- Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon, Republic of Korea
| | - Jisoo Shin
- Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Jonghun Jung
- Department of Molecular Medicine, Inha University, Incheon, Republic of Korea
- Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon, Republic of Korea
| | - Soyeon Ko
- Research Center for Controlling Intercellular Communication, Inha University, Incheon, Republic of Korea
- Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Kyung Hee Jung
- Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Soon-Sun Hong
- Research Center for Controlling Intercellular Communication, Inha University, Incheon, Republic of Korea
- Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Ji Eun Park
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
- Asia-Pacific Glycomics Reference Site, Daejeon, Republic of Korea
| | - Myung Jin Oh
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
- Asia-Pacific Glycomics Reference Site, Daejeon, Republic of Korea
| | - Hyun Joo An
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
- Asia-Pacific Glycomics Reference Site, Daejeon, Republic of Korea
| | - Wen-Hao Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Young-Gyu Ko
- Division of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Jong-Ho Cha
- Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea.
- Biohybrid Systems Research Center, Inha University, Incheon, Republic of Korea.
| | - Jae-Seon Lee
- Department of Molecular Medicine, Inha University, Incheon, Republic of Korea.
- Research Center for Controlling Intercellular Communication, Inha University, Incheon, Republic of Korea.
- Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon, Republic of Korea.
| |
Collapse
|
10
|
Franco-Caspueñas S, García-Montoya C, Contreras J, Lassaletta L, Varela-Nieto I, Jiménez-Lara AM. Uncovering cellular senescence as a therapeutic target in NF2-related vestibular schwannoma. Hear Res 2025; 455:109165. [PMID: 39647233 DOI: 10.1016/j.heares.2024.109165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Vestibular schwannomas (VS) are complex and heterogeneous human tumors arising from the Schwann cell compartment of the vestibulocochlear nerve. VS cause significant neurological deficit such as hearing loss and vestibular impairment, and in some cases death due to brainstem compression. There is an urgent need to find pharmacotherapies for VS since surgical removal and stereotactic radiosurgery are the only effective treatments. Cancer therapy based in the combination of drug-induced senescence and senolytics may provide an innovative pharmacological alternative for VS management. METHODS Senescence-associated β-galactosidase (SA-β-GAL) activity detection assay, real-time polymerase chain reaction (RT-PCR), western blotting and immunofluorescence, together with viability assays were used to analyze the response to different chemotherapy drugs of the human VS HEI-193 cell line. Human VS tumor paraffin sections were also studied for SA-β-GAL-stained cells. RESULTS We found that chemotherapy compounds induced genotoxic stress and cellular senescence in HEI-193 VS cells, as characterized by increased SA-β-GAL activity, growth arrest, increased levels of the cyclin-dependent kinase inhibitor p21 and the accumulation of DNA damage. These cellular senescence markers were also accompanied by an increase of senescence-associated secretory phenotype (SASP): IL6, IL8, IL1B and MMP1. Induction of senescence by chemotherapy rendered HEI-193 VS cells as druggable targets for senolytic compounds, as navitoclax. Thus, treatment with navitoclax selectively eliminated bleomycin-induced senescent HEI-193 VS cells by activating the extrinsic and intrinsic apoptosis pathways. Our data also show the presence of senescent cells, SA-β-GAL-positive stain, in human VS tumors, which are not present in healthy great auricular nerve sections. CONCLUSIONS These findings suggest that a one-two punch strategy of pro-senescence therapy induced by chemotherapy treatment followed by senolytic therapy represents a new paradigm for the pharmacological treatment of VS.
Collapse
Affiliation(s)
- Sandra Franco-Caspueñas
- Neuropathology of Hearing and Myelinopathies Group. Institute for Biomedical Research Sols-Morreale, Spanish National Research Council, Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; Rare Disease Network Biomedical Research Centre (CIBERER), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
| | - Carmen García-Montoya
- Neuropathology of Hearing and Myelinopathies Group. Institute for Biomedical Research Sols-Morreale, Spanish National Research Council, Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; Rare Disease Network Biomedical Research Centre (CIBERER), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Julio Contreras
- Neuropathology of Hearing and Myelinopathies Group. Institute for Biomedical Research Sols-Morreale, Spanish National Research Council, Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; Rare Disease Network Biomedical Research Centre (CIBERER), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; Department of Anatomy, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Luis Lassaletta
- Rare Disease Network Biomedical Research Centre (CIBERER), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain; Department of Otorhinolaryngology, La Paz University Hospital, 28046 Madrid, Spain
| | - Isabel Varela-Nieto
- Neuropathology of Hearing and Myelinopathies Group. Institute for Biomedical Research Sols-Morreale, Spanish National Research Council, Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; Rare Disease Network Biomedical Research Centre (CIBERER), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
| | - Ana M Jiménez-Lara
- Neuropathology of Hearing and Myelinopathies Group. Institute for Biomedical Research Sols-Morreale, Spanish National Research Council, Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain.
| |
Collapse
|
11
|
Shimizu K, Inuzuka H, Tokunaga F. The interplay between cell death and senescence in cancer. Semin Cancer Biol 2025; 108:1-16. [PMID: 39557316 DOI: 10.1016/j.semcancer.2024.11.001] [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: 08/15/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Cellular senescence is a state of permanent proliferative arrest that occurs in response to DNA damage-inducing endogenous and exogenous stresses, and is often accompanied by dynamic molecular changes such as a senescence-associated secretory phenotype (SASP). Accumulating evidence indicates that age-associated increases in the upstream and downstream signals of regulated cell death, including apoptosis, necroptosis, pyroptosis, and ferroptosis, are closely related to the induction of cellular senescence and its phenotype. Furthermore, elevated levels of pro-inflammatory SASP factors with aging can be both a cause and consequence of several cell death modes, suggesting the reciprocal effects of cellular senescence and cells undergoing regulated cell death. Here, we review the critical molecular pathways of the regulated cell death forms and describe the crosstalk between aging-related signals and cancer. In addition, we discuss how targeting regulated cell death could be harnessed in therapeutic interventions for cancer. ABBREVIATIONS: Abbreviations that are not standard in this field are defined at their first occurrence in the article and are used consistently throughout the article.
Collapse
Affiliation(s)
- Kouhei Shimizu
- Department of Medical Biochemistry, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan.
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA02215, USA
| | - Fuminori Tokunaga
- Department of Medical Biochemistry, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| |
Collapse
|
12
|
Balamurli G, Liew AQX, Tee WW, Pervaiz S. Interplay between epigenetics, senescence and cellular redox metabolism in cancer and its therapeutic implications. Redox Biol 2024; 78:103441. [PMID: 39612910 PMCID: PMC11629570 DOI: 10.1016/j.redox.2024.103441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
There is accumulating evidence indicating a close crosstalk between key molecular events regulating cell growth and proliferation, which could profoundly impact carcinogenesis and its progression. Here we focus on reviewing observations highlighting the interplay between epigenetic modifications, irreversible cell cycle arrest or senescence, and cellular redox metabolism. Epigenetic alterations, such as DNA methylation and histone modifications, dynamically influence tumour transcriptome, thereby impacting tumour phenotype, survival, growth and spread. Interestingly, the acquisition of senescent phenotype can be triggered by epigenetic changes, acting as a double-edged sword via its ability to suppress tumorigenesis or by facilitating an inflammatory milieu conducive for cancer progression. Concurrently, an aberrant redox metabolism, which is a function of the balance between reactive oxygen species (ROS) generation and intracellular anti-oxidant defences, influences signalling cascades and genomic stability in cancer cells by serving as a critical link between epigenetics and senescence. Recognizing this intricate interconnection offers a nuanced perspective for therapeutic intervention by simultaneously targeting specific epigenetic modifications, modulating senescence dynamics, and restoring redox homeostasis.
Collapse
Affiliation(s)
- Geoffrey Balamurli
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Chromatin Dynamics and Disease Epigenetics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Angeline Qiu Xia Liew
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), NUS, Singapore
| | - Wee Wei Tee
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Chromatin Dynamics and Disease Epigenetics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, NUS, Singapore; Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), NUS, Singapore; NUS Medicine Healthy Longevity Program, NUS, Singapore; National University Cancer Institute, National University Health System, Singapore.
| |
Collapse
|
13
|
Nicolas E, Kosmider B, Cukierman E, Borghaei H, Golemis EA, Borriello L. Cancer treatments as paradoxical catalysts of tumor awakening in the lung. Cancer Metastasis Rev 2024; 43:1165-1183. [PMID: 38963567 PMCID: PMC11554904 DOI: 10.1007/s10555-024-10196-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Much of the fatality of tumors is linked to the growth of metastases, which can emerge months to years after apparently successful treatment of primary tumors. Metastases arise from disseminated tumor cells (DTCs), which disperse through the body in a dormant state to seed distant sites. While some DTCs lodge in pre-metastatic niches (PMNs) and rapidly develop into metastases, other DTCs settle in distinct microenvironments that maintain them in a dormant state. Subsequent awakening, induced by changes in the microenvironment of the DTC, causes outgrowth of metastases. Hence, there has been extensive investigation of the factors causing survival and subsequent awakening of DTCs, with the goal of disrupting these processes to decrease cancer lethality. We here provide a detailed overview of recent developments in understanding of the factors controlling dormancy and awakening in the lung, a common site of metastasis for many solid tumors. These factors include dynamic interactions between DTCs and diverse epithelial, mesenchymal, and immune cell populations resident in the lung. Paradoxically, among key triggers for metastatic outgrowth, lung tissue remodeling arising from damage induced by the treatment of primary tumors play a significant role. In addition, growing evidence emphasizes roles for inflammation and aging in opposing the factors that maintain dormancy. Finally, we discuss strategies being developed or employed to reduce the risk of metastatic recurrence.
Collapse
Affiliation(s)
- Emmanuelle Nicolas
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Beata Kosmider
- Center for Inflammation and Lung Research, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
- Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Hossein Borghaei
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Erica A Golemis
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
| | - Lucia Borriello
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA.
| |
Collapse
|
14
|
Fu M, Zhang Y, Peng B, Luo N, Zhang Y, Zhu W, Yang F, Chen Z, Zhang Q, Li Q, Chen X, Liu Y, Long G, Hu G, Peng X. All-trans retinoic acid inhibits glioblastoma progression and attenuates radiation-induced brain injury. JCI Insight 2024; 9:e179530. [PMID: 39513361 PMCID: PMC11601587 DOI: 10.1172/jci.insight.179530] [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: 01/19/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Radiotherapy (RT) remains a primary treatment modality for glioblastoma (GBM), but it induces cellular senescence and is strongly implicated in GBM progression and RT-related injury. Recently, eliminating senescent cells has emerged as a promising strategy for treating cancer and for mitigating radiation-induced brain injury (RBI). Here, we investigated the impact of all-trans retinoic acid (RA) on radiation-induced senescence. The findings of this study revealed that RA effectively eliminated astrocytes, which are particularly prone to senescence after radiation, and that the removal of senescence-associated secretory phenotype factor-producing astrocytes inhibited GBM cell proliferation in vitro. Moreover, RA-mediated clearance of senescent cells improved survival in GBM-bearing mice and alleviated radiation-induced cognitive impairment. Through RNA sequencing, we found that the AKT/mTOR/PPARγ/Plin4 signaling pathway is involved in RA-mediated clearance of senescent cells. In summary, these results suggest that RA could be a potential senolytic drug for preventing GBM progression and improving RBI.
Collapse
Affiliation(s)
- Min Fu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiling Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bi Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Zhang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjun Zhu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziqi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanhui Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoxian Long
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohong Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Zheng H, Li T, Hu Z, Zheng Q, Wang J. The potential of flavonoids to mitigate cellular senescence in cardiovascular disease. Biogerontology 2024; 25:985-1010. [PMID: 39325277 DOI: 10.1007/s10522-024-10141-7] [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: 06/28/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Aging is one of the most significant factors affecting cardiovascular health, with cellular senescence being a central hallmark. Senescent cells (SCs) secrete a specific set of signaling molecules known as the senescence-associated secretory phenotype (SASP). The SASP has a remarkable impact on age-associated diseases, particularly cardiovascular diseases (CVD). Targeting SCs through anti-aging therapies represents a novel strategy to effectively retard senescence and attenuate disease progression. Accumulating evidence demonstrates that the flavonoids, widely presented in fruits and vegetables worldwide, can delay or treat CVD via selectively eliminating SCs (senolytics) and modulating SASPs (senomorphics). Nevertheless, only sporadic research has illustrated the application of flavonoids in targeting SCs for CVD, which requires further exploration. This review recapitulates the hallmarks and key molecular mechanisms involved in cellular senescence, then summarizes senescence of different types of cardiac cells and describes the mechanisms by which cellular senescence affects CVD development. The discussion culminates with the potential use of flavonoids via exerting their biological effects on cellular senescence to reduce CVD incidence. This summary will provide valuable insights for cardiovascular drug design, development and clinical applications leveraging flavonoids.
Collapse
Affiliation(s)
- Huimin Zheng
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China
| | - Tiantian Li
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China
| | - Ziyun Hu
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China
| | - Qi Zheng
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China
| | - Junsong Wang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China.
| |
Collapse
|
16
|
Ordónez-Rubiano EG, Cómbita A, Baldoncini M, Payán-Gómez C, Gómez-Amarillo DF, Hakim F, Camargo J, Zorro-Sepúlveda V, Luzzi S, Zorro O, Parra-Medina R. Cellular Senescence in Diffuse Gliomas: From Physiopathology to Possible Treatments. World Neurosurg 2024; 191:138-148. [PMID: 39233309 DOI: 10.1016/j.wneu.2024.08.060] [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: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
Cellular senescence in gliomas is a complex process that is induced by aging and replication, ionizing radiation, oncogenic stress, and the use of temozolomide. However, the escape routes that gliomas must evade senescence and achieve cellular immortality are much more complex, in which the expression of telomerase and the alternative lengthening of telomeres, as well as the mutation of some proto-oncogenes or tumor suppressor genes, are involved. In gliomas, these molecular mechanisms related to cellular senescence can have a tumor-suppressing or promoting effect and are directly involved in tumor recurrence and progression. From these cellular mechanisms related to cellular senescence, it is possible to generate targeted senostatic and senolytic therapies that improve the response to currently available treatments and improve survival rates. This review aims to summarize the mechanisms of induction and evasion of cellular senescence in gliomas, as well as review possible treatments with therapies targeting pathways related to cellular senescence.
Collapse
Affiliation(s)
- Edgar G Ordónez-Rubiano
- Department of Neurological Surgery, Fundación Universitaria de Ciencias de la Salud (FUCS), Hospital de San José - Sociedad de Cirugía de Bogotá, Bogotá, Colombia; School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia; Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia.
| | - Alba Cómbita
- Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia; Department of Microbiology, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Matías Baldoncini
- School of Medicine, Laboratory of Microsurgical Neuroanatomy, Second Chair of Gross Anatomy, University of Buenos Aires, Buenos Aires, Argentina; Department of Neurological Surgery, Hospital San Fernando, Buenos Aires, Argentina
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz, Colombia
| | - Diego F Gómez-Amarillo
- Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Fernando Hakim
- Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Julián Camargo
- Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | | | - Sabino Luzzi
- Neurosurgery Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Oscar Zorro
- Department of Neurological Surgery, Fundación Universitaria de Ciencias de la Salud (FUCS), Hospital de San José - Sociedad de Cirugía de Bogotá, Bogotá, Colombia
| | - Rafael Parra-Medina
- Department of Pathology, Instituto Nacional de Cancerología, Bogotá, Colombia; Research Institute, Fundación Universitaria de Ciencias de la Salud (FUCS), Hospital de San José - Sociedad de Cirugía de Bogotá, Bogotá, Colombia
| |
Collapse
|
17
|
Zhang Y, Xiao B, Yuan S, Ding L, Pan Y, Jiang Y, Sun S, Ke X, Cai L, Jia L. Tryptanthrin targets GSTP1 to induce senescence and increases the susceptibility to apoptosis by senolytics in liver cancer cells. Redox Biol 2024; 76:103323. [PMID: 39180983 PMCID: PMC11388193 DOI: 10.1016/j.redox.2024.103323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/05/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024] Open
Abstract
Targeting senescence has emerged as a promising strategy for liver cancer treatment. However, the lack of a safe agent capable of inducing complete senescence and being combined with senolytics poses a limitation. Here, we screened a natural product library and identified tryptanthrin (TRYP) as a potent inducer of cellular senescence in liver cancer cells both in vitro and in vivo. Mechanistically, Glutathione S-transferase P1 (GSTP1), a key regulator for redox homeostasis, was identified as a target protein for TRYP-induced senescence. TRYP directly bound to GSTP1 and inhibited its enzymatic activity, mediating reactive oxygen species (ROS) accumulation, followed by DNA damage response (DDR), consequently contributing to initiating primary senescence. Furthermore, TRYP triggered DNA damage-dependent activation of NF-κB pathway, which evoked senescence-associated secretory phenotype (SASP), thereby leading to senescence reinforcement. Importantly, TRYP exposed the vulnerability of tumor cells and sensitized senescent cells to apoptosis induced by senolytic agent ABT263, a Bcl2 inhibitor. Taken together, our findings reveal that TRYP induces cellular senescence via GSTP1/ROS/DDR/NF-κB/SASP axis, providing a novel potential application in synergizing with senolytic therapy in liver cancer.
Collapse
Affiliation(s)
- Yuxuan Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Biying Xiao
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Shuying Yuan
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Lele Ding
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yongfu Pan
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yanyu Jiang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Shenghao Sun
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Xisong Ke
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Lili Cai
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| |
Collapse
|
18
|
O'Sullivan EA, Wallis R, Mossa F, Bishop CL. The paradox of senescent-marker positive cancer cells: challenges and opportunities. NPJ AGING 2024; 10:41. [PMID: 39277623 PMCID: PMC11401916 DOI: 10.1038/s41514-024-00168-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/15/2024] [Indexed: 09/17/2024]
Abstract
Senescence is an anti-tumour mechanism and hallmark of cancer. Loss or mutation of key senescence effectors, such as p16INK4A, are frequently observed in cancer. Intriguingly, some human tumours are both proliferative and senescent-marker positive (Sen-Mark+). Here, we explore this paradox, focusing on the prognostic consequences and the current challenges in classifying these cells. We discuss future strategies for Sen-Mark+ cell detection together with emerging opportunities to exploit senescence for cancer.
Collapse
Affiliation(s)
- Emily A O'Sullivan
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ryan Wallis
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Federica Mossa
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Cleo L Bishop
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
19
|
Yu J, Zhang M, Li T, Gao W, Yang Z, Wang K, Liu Z, Zhu S, Wen S, Zhao Y, Cai Q, Shang Z, Wang Y, Niu Y. Monoacylglycerol lipase blockades the senescence-associated secretory phenotype by interfering with NF-κB activation and promotes docetaxel efficacy in prostate cancer. Oncogene 2024; 43:2835-2849. [PMID: 39155296 DOI: 10.1038/s41388-024-03132-y] [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: 12/14/2023] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Metabolic reprogramming and cellular senescence greatly contribute to cancer relapse and recurrence. In aging and treated prostate, persistent accumulating senescence-associated secretory phenotype (SASP) of cancer cells often limits the overall survival of patients. Novel strategic therapy with monoacylglycerol lipase (MGLL) upregulation that counters the cellular and docetaxel induced SASP might overcome this clinical challenge in prostate cancer (PCa). With primary comparative expression and survival analysis screening of fatty acid (FA) metabolism signature genes in the TCGA PCa dataset and our single center cohort, MGLL was detected to be downregulated in malignancy prostate tissues and its low expression predicted worse progression-free and overall survival. Functionally, overexpression of MGLL mainly suppresses NF-κB-driven SASP (N-SASP) which mostly restricts the cancer cell paracrine and autocrine tumorigenic manners and the corresponding cellular senescence. Further investigating metabolites, we determined that MGLL constitutive expression prevents lipid accumulation, decreases metabolites preferably, and consequently downregulates ATP levels. Overexpressed MGLL inhibited IκBα phosphorylation, NF-κB p65 phosphorylation, and NF-κB nuclear translocation to deactivate NF-κB transcriptional activities, and be responsible for the repressed N-SASP, partially through reducing ATP levels. Preclinically, combinational treatment with MGLL overexpression and docetaxel chemotherapy dramatically delays tumor progression in mouse models. Taken together, our findings identify MGLL as a switch for lipase-related N-SASP suppression and provide a potential drug candidate for promoting docetaxel efficacy in PCa.
Collapse
Affiliation(s)
- Jianpeng Yu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Tianjin Institute of Urology, Tianjin, 300211, China.
| | - Minghao Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Tianjin Institute of Urology, Tianjin, 300211, China
- Department of Urology, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Taipeng Li
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Tianjin Institute of Urology, Tianjin, 300211, China
| | - Wenlong Gao
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Tianjin Institute of Urology, Tianjin, 300211, China
| | - Zhao Yang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Tianjin Institute of Urology, Tianjin, 300211, China
| | - Keruo Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Tianjin Institute of Urology, Tianjin, 300211, China
| | - Zihao Liu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Tianjin Institute of Urology, Tianjin, 300211, China
| | - Shimiao Zhu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Tianjin Institute of Urology, Tianjin, 300211, China
| | - Simeng Wen
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Tianjin Institute of Urology, Tianjin, 300211, China
| | - Yang Zhao
- Tianjin Institute of Urology, Tianjin, 300211, China
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Qiliang Cai
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Tianjin Institute of Urology, Tianjin, 300211, China.
| | - Zhiqun Shang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Tianjin Institute of Urology, Tianjin, 300211, China.
| | - Yong Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Tianjin Institute of Urology, Tianjin, 300211, China.
| | - Yuanjie Niu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Tianjin Institute of Urology, Tianjin, 300211, China.
| |
Collapse
|
20
|
Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Biomarkers of Cellular Senescence and Aging: Current State-of-the-Art, Challenges and Future Perspectives. Adv Biol (Weinh) 2024; 8:e2400079. [PMID: 38935557 DOI: 10.1002/adbi.202400079] [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: 02/13/2024] [Revised: 05/29/2024] [Indexed: 06/29/2024]
Abstract
Population aging has increased the global prevalence of aging-related diseases, including cancer, sarcopenia, neurological disease, arthritis, and heart disease. Understanding aging, a fundamental biological process, has led to breakthroughs in several fields. Cellular senescence, evinced by flattened cell bodies, vacuole formation, and cytoplasmic granules, ubiquitously plays crucial roles in tissue remodeling, embryogenesis, and wound repair as well as in cancer therapy and aging. The lack of universal biomarkers for detecting and quantifying senescent cells, in vitro and in vivo, constitutes a major limitation. The applications and limitations of major senescence biomarkers, including senescence-associated β-galactosidase staining, telomere shortening, cell-cycle arrest, DNA methylation, and senescence-associated secreted phenotypes are discussed. Furthermore, explore senotherapeutic approaches for aging-associated diseases and cancer. In addition to the conventional biomarkers, this review highlighted the in vitro, in vivo, and disease models used for aging studies. Further, technologies from the current decade including multi-omics and computational methods used in the fields of senescence and aging are also discussed in this review. Understanding aging-associated biological processes by using cellular senescence biomarkers can enable therapeutic innovation and interventions to improve the quality of life of older adults.
Collapse
Affiliation(s)
- Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Hyun-Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu, 41062, Republic of Korea
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Jeollanam-do, Naju, 58245, Republic of Korea
- Korean Convergence Medicine Major, University of Science & Technology (UST), KIOM Campus, Daejeon, 34054, Republic of Korea
| |
Collapse
|
21
|
Kotani H, Han W, Iida Y, Tanino R, Katakawa K, Okimoto T, Tsubata Y, Isobe T, Harada M. Therapeutic Senolysis of Axitinib-Induced Senescent Human Lung Cancer Cells. Cancers (Basel) 2024; 16:2782. [PMID: 39199555 PMCID: PMC11352446 DOI: 10.3390/cancers16162782] [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: 07/15/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) inhibit receptor-mediated signals in cells. Axitinib is a TKI with high specificity for vascular endothelial growth factor receptors (VEGFRs). AIM We determined whether axitinib could induce senescence in human cancer cells and be lysed by the senolytic drug ABT-263. METHODS Human lung and breast adenocarcinoma cell lines were used. These cells were cultured with axitinib or a multi-target TKI lenvatinib. The expression of β-galactosidase, VEGFRs, Ki-67, reactive oxygen species (ROS) of cancer cells, and their BrdU uptake were evaluated by flow cytometry. The mRNA expression of p21 and IL-8 was examined by quantitative PCR. The effects of TKIs on phosphorylation of Akt and Erk1/2, as downstream molecules of VEGFR signaling, were examined by immunoblot. The in vivo anti-cancer effect was examined using a xenograft mice model. RESULTS Axitinib, but not lenvatinib, induced cellular senescence (increased cell size and enhanced expression of β-galactosidase) in all adenocarcinoma cell lines. Axitinib-induced senescence was unrelated to the expression of VEGFRs on cancer cells. ROS were involved in axitinib-induced senescence. Axitinib-induced senescent lung adenocarcinoma A549 cells were drastically lysed by ABT-263. In A549-xenografted mice, combination therapy with axitinib and ABT-263 significantly suppressed tumor growth with the induction of apoptotic cancer cells.
Collapse
Affiliation(s)
- Hitoshi Kotani
- Department of Immunology, Faculty of Medicine, Shimane University, Izumo 693-8501, Shimane, Japan; (H.K.); (W.H.); (Y.I.)
| | - Wei Han
- Department of Immunology, Faculty of Medicine, Shimane University, Izumo 693-8501, Shimane, Japan; (H.K.); (W.H.); (Y.I.)
| | - Yuichi Iida
- Department of Immunology, Faculty of Medicine, Shimane University, Izumo 693-8501, Shimane, Japan; (H.K.); (W.H.); (Y.I.)
| | - Ryosuke Tanino
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Shimane, Japan; (R.T.); (T.O.); (Y.T.); (T.I.)
| | - Kazuaki Katakawa
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Shonan University of Medical Sciences, Yokohama 244-0806, Kanagawa, Japan;
| | - Tamio Okimoto
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Shimane, Japan; (R.T.); (T.O.); (Y.T.); (T.I.)
| | - Yukari Tsubata
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Shimane, Japan; (R.T.); (T.O.); (Y.T.); (T.I.)
| | - Takeshi Isobe
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Shimane, Japan; (R.T.); (T.O.); (Y.T.); (T.I.)
| | - Mamoru Harada
- Department of Immunology, Faculty of Medicine, Shimane University, Izumo 693-8501, Shimane, Japan; (H.K.); (W.H.); (Y.I.)
| |
Collapse
|
22
|
Tanaka H, Sugawara S, Tanaka Y, Loo TM, Tachibana R, Abe A, Kamiya M, Urano Y, Takahashi A. Dipeptidylpeptidase-4-targeted activatable fluorescent probes visualize senescent cells. Cancer Sci 2024; 115:2762-2773. [PMID: 38802068 PMCID: PMC11309953 DOI: 10.1111/cas.16229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Senescent cells promote cancer development and progression through chronic inflammation caused by a senescence-associated secretory phenotype (SASP). Although various senotherapeutic strategies targeting senescent cells have been developed for the prevention and treatment of cancers, technology for the in vivo detection and evaluation of senescent cell accumulation has not yet been established. Here, we identified activatable fluorescent probes targeting dipeptidylpeptidase-4 (DPP4) as an effective probe for detecting senescent cells through an enzymatic activity-based screening of fluorescent probes. We also determined that these probes were highly, selectively, and rapidly activated in senescent cells during live cell imaging. Furthermore, we successfully visualized senescent cells in the organs of mice using DPP4-targeted probes. These results are expected to lead to the development of a diagnostic technology for noninvasively detecting senescent cells in vivo and could play a role in the application of DPP4 prodrugs for senotherapy.
Collapse
Affiliation(s)
- Hisamichi Tanaka
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
- Department of JFCR Cancer Biology, Graduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Sho Sugawara
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Yoko Tanaka
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Tze Mun Loo
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Ryo Tachibana
- Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Atsuki Abe
- Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Mako Kamiya
- Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
- Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Akiko Takahashi
- Division of Cellular SenescenceCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
- Cancer Cell Communication Project, NEXT‐Ganken ProgramJapanese Foundation for Cancer ResearchTokyoJapan
| |
Collapse
|
23
|
Tang J, Yang Y, Yin HY, Ma B, Zhu M, Yang ZS, Peng XX, Jia F, Zhao Y, Wang F, Chen T, Zhang JL. A Platinum-Aluminum Bimetallic Salen Complex for Pro-senescence Cancer Therapy. Chembiochem 2024; 25:e202400105. [PMID: 38639074 DOI: 10.1002/cbic.202400105] [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: 02/01/2024] [Revised: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 04/20/2024]
Abstract
Cell senescence is defined as irreversible cell cycle arrest, which can be triggered by telomere shortening or by various types of genotoxic stress. Induction of senescence is emerging as a new strategy for the treatment of cancer, especially when sequentially combined with a second senolytic drug capable of killing the resulting senescent cells, however severely suffering from the undesired off-target side effects from the senolytic drugs. Here, we prepare a bimetalic platinum-aluminum salen complex (Alumiplatin) for cancer therapy-a combination of pro-senesence chemotherapy with in situ senotherapy to avoid the side effects. The aluminum salen moiety, as a G-quadruplex stabilizer, enhances the salen's ability to induce cancer cell senescence and this phenotype is in turn sensitive to the cytotoxic activity of the monofunctional platinum moiety. It exhibits an excellent capability for inducing senescence, a potent cytotoxic activity against cancer cells both in vitro and in vivo, and an improved safety profile compared to cisplatin. Therefore, Alumiplatin may be a good candidate to be further developed into safe and effective anticancer agents. This novel combination of cell senescence inducers with genotoxic drugs revolutionizes the therapy options of designing multi-targeting anticancer agents to improve the efficacy of anticancer therapies.
Collapse
Affiliation(s)
- Juan Tang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
- Key Laboratory of Medicinal Molecule Science and pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 102488, P. R. China
| | - Yahui Yang
- Department of Chemistry, Jinan University, Guangzhou, 510632, P. R. China
| | - Hao-Yan Yin
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Bin Ma
- Department of Chemistry, Jinan University, Guangzhou, 510632, P. R. China
| | - Mengliang Zhu
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Zi-Shu Yang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Xin-Xin Peng
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Feifei Jia
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, National Centre for Mass Spectrometry in Beijing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yao Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, National Centre for Mass Spectrometry in Beijing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, National Centre for Mass Spectrometry in Beijing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, 510632, P. R. China
| | - Jun-Long Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Rare Earth Materials Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| |
Collapse
|
24
|
Nehme J, Mesilmany L, Varela-Eirin M, Brandenburg S, Altulea A, Lin Y, Gaya da Costa M, Seelen M, Hillebrands JL, van Goor H, Saab R, Akl H, Prevarskaya N, Farfariello V, Demaria M. Converting cell death into senescence by PARP1 inhibition improves recovery from acute oxidative injury. NATURE AGING 2024; 4:771-782. [PMID: 38724734 DOI: 10.1038/s43587-024-00627-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/05/2024] [Indexed: 06/21/2024]
Abstract
Excessive amounts of reactive oxygen species (ROS) lead to macromolecular damage and high levels of cell death with consequent pathological sequelae. We hypothesized that switching cell death to a tissue regenerative state could potentially improve the short-term and long-term detrimental effects of ROS-associated acute tissue injury, although the mechanisms regulating oxidative stress-induced cell fate decisions and their manipulation for improving repair are poorly understood. Here, we show that cells exposed to high oxidative stress enter a poly (ADP-ribose) polymerase 1 (PARP1)-mediated regulated cell death, and that blocking PARP1 activation promotes conversion of cell death into senescence (CODIS). We demonstrate that this conversion depends on reducing mitochondrial Ca2+ overload as a consequence of retaining the hexokinase II on mitochondria. In a mouse model of kidney ischemia-reperfusion damage, PARP inhibition reduces necrosis and increases transient senescence at the injury site, alongside improved recovery from damage. Together, these data provide evidence that converting cell death into transient senescence can therapeutically benefit tissue regeneration.
Collapse
Affiliation(s)
- Jamil Nehme
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen (RUG), University Medical Center Groningen, Groningen, the Netherlands
- Department of Biology, Lebanese University, Beirut, Lebanon
| | - Lina Mesilmany
- Department of Biology, Lebanese University, Beirut, Lebanon
- Université de Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Villeneuve d'Ascq, France
| | - Marta Varela-Eirin
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen (RUG), University Medical Center Groningen, Groningen, the Netherlands
| | - Simone Brandenburg
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen (RUG), University Medical Center Groningen, Groningen, the Netherlands
| | - Abdullah Altulea
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen (RUG), University Medical Center Groningen, Groningen, the Netherlands
| | - Yao Lin
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen (RUG), University Medical Center Groningen, Groningen, the Netherlands
| | - Mariana Gaya da Costa
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marc Seelen
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology & Medical Biology, Pathology Division, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology & Medical Biology, Pathology Division, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Raya Saab
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Haidar Akl
- Department of Biology, Lebanese University, Beirut, Lebanon
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Natacha Prevarskaya
- Université de Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Villeneuve d'Ascq, France
| | - Valerio Farfariello
- Université de Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Villeneuve d'Ascq, France
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen (RUG), University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
25
|
Neuwahl J, Neumann CA, Fitz AC, Biermann AD, Magel M, Friedrich A, Sellin L, Stork B, Piekorz RP, Proksch P, Budach W, Jänicke RU, Sohn D. Combined inhibition of class 1-PI3K-alpha and delta isoforms causes senolysis by inducing p21 WAF1/CIP1 proteasomal degradation in senescent cells. Cell Death Dis 2024; 15:373. [PMID: 38811535 PMCID: PMC11136996 DOI: 10.1038/s41419-024-06755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
The targeted elimination of radio- or chemotherapy-induced senescent cells by so-called senolytic substances represents a promising approach to reduce tumor relapse as well as therapeutic side effects such as fibrosis. We screened an in-house library of 178 substances derived from marine sponges, endophytic fungi, and higher plants, and determined their senolytic activities towards DNA damage-induced senescent HCT116 colon carcinoma cells. The Pan-PI3K-inhibitor wortmannin and its clinical derivative, PX-866, were identified to act as senolytics. PX-866 potently induced apoptotic cell death in senescent HCT116, MCF-7 mammary carcinoma, and A549 lung carcinoma cells, independently of whether senescence was induced by ionizing radiation or by chemotherapeutics, but not in proliferating cells. Other Pan-PI3K inhibitors, such as the FDA-approved drug BAY80-6946 (Copanlisib, Aliqopa®), also efficiently and specifically eliminated senescent cells. Interestingly, only the simultaneous inhibition of both PI3K class I alpha (with BYL-719 (Alpelisib, Piqray®)) and delta (with CAL-101 (Idelalisib, Zydelig®)) isoforms was sufficient to induce senolysis, whereas single application of these inhibitors had no effect. On the molecular level, inhibition of PI3Ks resulted in an increased proteasomal degradation of the CDK inhibitor p21WAF1/CIP1 in all tumor cell lines analyzed. This led to a timely induction of apoptosis in senescent tumor cells. Taken together, the senolytic properties of PI3K-inhibitors reveal a novel dimension of these promising compounds, which holds particular potential when employed alongside DNA damaging agents in combination tumor therapies.
Collapse
Affiliation(s)
- Judith Neuwahl
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Chantal A Neumann
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Annika C Fitz
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anica D Biermann
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Experimental Nephrology, Clinic for Nephrology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Maja Magel
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH, Aachen, Germany
| | - Annabelle Friedrich
- Institute of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Lorenz Sellin
- Experimental Nephrology, Clinic for Nephrology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Björn Stork
- Institute of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Roland P Piekorz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Wilfried Budach
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Reiner U Jänicke
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Dennis Sohn
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
26
|
Silva AO, Bitencourt TC, Vargas JE, Fraga LR, Filippi-Chiela E. Modulation of tumor plasticity by senescent cells: Deciphering basic mechanisms and survival pathways to unravel therapeutic options. Genet Mol Biol 2024; 47Suppl 1:e20230311. [PMID: 38805699 PMCID: PMC11132560 DOI: 10.1590/1678-4685-gmb-2023-0311] [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: 11/02/2023] [Accepted: 03/21/2024] [Indexed: 05/30/2024] Open
Abstract
Senescence is a cellular state in which the cell loses its proliferative capacity, often irreversibly. Physiologically, it occurs due to a limited capacity of cell division associated with telomere shortening, the so-called replicative senescence. It can also be induced early due to DNA damage, oncogenic activation, oxidative stress, or damage to other cellular components (collectively named induced senescence). Tumor cells acquire the ability to bypass replicative senescence, thus ensuring the replicative immortality, a hallmark of cancer. Many anti-cancer therapies, however, can lead tumor cells to induced senescence. Initially, this response leads to a slowdown in tumor growth. However, the longstanding accumulation of senescent cells (SnCs) in tumors can promote neoplastic progression due to the enrichment of numerous molecules and extracellular vesicles that constitutes the senescence-associated secretory phenotype (SASP). Among other effects, SASP can potentiate or unlock the tumor plasticity and phenotypic transitions, another hallmark of cancer. This review discusses how SnCs can fuel mechanisms that underlie cancer plasticity, like cell differentiation, stemness, reprogramming, and epithelial-mesenchymal transition. We also discuss the main molecular mechanisms that make SnCs resistant to cell death, and potential strategies to target SnCs. At the end, we raise open questions and clinically relevant perspectives in the field.
Collapse
Affiliation(s)
- Andrew Oliveira Silva
- Faculdade Estácio, Porto Alegre, RS, Brazil
- Centro de Pesquisa Experimental, Hospital de Clínicas de Porto
Alegre, Porto Alegre, RS, Brazil
| | - Thais Cardoso Bitencourt
- Centro de Pesquisa Experimental, Hospital de Clínicas de Porto
Alegre, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação
em Biologia Celular e Molecular, Porto Alegre, RS, Brazil
| | - Jose Eduardo Vargas
- Universidade Federal do Paraná, Departamento de Biologia Celular,
Curitiba, PR, Brazil
| | - Lucas Rosa Fraga
- Centro de Pesquisa Experimental, Hospital de Clínicas de Porto
Alegre, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Ciências
Morfológicas, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação
em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil
| | - Eduardo Filippi-Chiela
- Centro de Pesquisa Experimental, Hospital de Clínicas de Porto
Alegre, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Ciências
Morfológicas, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Centro de Biotecnologia,
Porto Alegre, RS, Brazil
| |
Collapse
|
27
|
Favaretto G, Rossi MN, Cuollo L, Laffranchi M, Cervelli M, Soriani A, Sozzani S, Santoni A, Antonangeli F. Neutrophil-activating secretome characterizes palbociclib-induced senescence of breast cancer cells. Cancer Immunol Immunother 2024; 73:113. [PMID: 38693312 PMCID: PMC11063017 DOI: 10.1007/s00262-024-03695-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/30/2024] [Indexed: 05/03/2024]
Abstract
Senescent cells have a profound impact on the surrounding microenvironment through the secretion of numerous bioactive molecules and inflammatory factors. The induction of therapy-induced senescence by anticancer drugs is known, but how senescent tumor cells influence the tumor immune landscape, particularly neutrophil activity, is still unclear. In this study, we investigate the induction of cellular senescence in breast cancer cells and the subsequent immunomodulatory effects on neutrophils using the CDK4/6 inhibitor palbociclib, which is approved for the treatment of breast cancer and is under intense investigation for additional malignancies. Our research demonstrates that palbociclib induces a reversible form of senescence endowed with an inflammatory secretome capable of recruiting and activating neutrophils, in part through the action of interleukin-8 and acute-phase serum amyloid A1. The activation of neutrophils is accompanied by the release of neutrophil extracellular trap and the phagocytic removal of senescent tumor cells. These findings may be relevant for the success of cancer therapy as neutrophils, and neutrophil-driven inflammation can differently affect tumor progression. Our results reveal that neutrophils, as already demonstrated for macrophages and natural killer cells, can be recruited and engaged by senescent tumor cells to participate in their clearance. Understanding the interplay between senescent cells and neutrophils may lead to innovative strategies to cope with chronic or tumor-associated inflammation.
Collapse
Affiliation(s)
- Gabriele Favaretto
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy
| | | | - Lorenzo Cuollo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Mattia Laffranchi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Fabrizio Antonangeli
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy.
| |
Collapse
|
28
|
Zheng H, Liu M, Shi S, Huang H, Yang X, Luo Z, Song Y, Xu Q, Li T, Xue L, Lu F, Wang J. MAP4K4 and WT1 mediate SOX6-induced cellular senescence by synergistically activating the ATF2-TGFβ2-Smad2/3 signaling pathway in cervical cancer. Mol Oncol 2024; 18:1327-1346. [PMID: 38383842 PMCID: PMC11076992 DOI: 10.1002/1878-0261.13613] [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: 07/27/2023] [Revised: 01/18/2024] [Accepted: 02/04/2024] [Indexed: 02/23/2024] Open
Abstract
SRY-box transcription factor 6 (SOX6) is a member of the SOX gene family and inhibits the proliferation of cervical cancer cells by inducing cell cycle arrest. However, the final cell fate and significance of these cell-cycle-arrested cervical cancer cells induced by SOX6 remains unclear. Here, we report that SOX6 inhibits the proliferation of cervical cancer cells by inducing cellular senescence, which is mainly mediated by promoting transforming growth factor beta 2 (TGFB2) gene expression and subsequently activating the TGFβ2-Smad2/3-p53-p21WAF1/CIP1-Rb pathway. SOX6 promotes TGFB2 gene expression through the MAP4K4-MAPK (JNK/ERK/p38)-ATF2 and WT1-ATF2 pathways, which is dependent on its high-mobility group (HMG) domain. In addition, the SOX6-induced senescent cervical cancer cells are resistant to cisplatin treatment. ABT-263 (navitoclax) and ABT-199 (venetoclax), two classic senolytics, can specifically eliminate the SOX6-induced senescent cervical cancer cells, and thus significantly improve the chemosensitivity of cisplatin-resistant cervical cancer cells. This study uncovers that the MAP4K4/WT1-ATF2-TGFβ2 axis mediates SOX6-induced cellular senescence, which is a promising therapeutic target in improving the chemosensitivity of cervical cancer.
Collapse
Affiliation(s)
- Han Zheng
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
- NHC Key Laboratory of Medical ImmunologyPeking UniversityBeijingChina
| | - Mingchen Liu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Shu Shi
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Hongxin Huang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Xingwen Yang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
- NHC Key Laboratory of Medical ImmunologyPeking UniversityBeijingChina
| | - Ziheng Luo
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
- NHC Key Laboratory of Medical ImmunologyPeking UniversityBeijingChina
| | - Yarong Song
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
- NHC Key Laboratory of Medical ImmunologyPeking UniversityBeijingChina
| | - Qiang Xu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Tingting Li
- Department of Biomedical Informatics, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Lixiang Xue
- Department of Radiation OncologyCancer Center of Peking University Third Hospital, Peking University Third HospitalBeijingChina
| | - Fengmin Lu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Jie Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
- NHC Key Laboratory of Medical ImmunologyPeking UniversityBeijingChina
| |
Collapse
|
29
|
Rutecki S, Pakuła-Iwańska M, Leśniewska-Bocianowska A, Matuszewska J, Rychlewski D, Uruski P, Stryczyński Ł, Naumowicz E, Szubert S, Tykarski A, Mikuła-Pietrasik J, Książek K. Mechanisms of carboplatin- and paclitaxel-dependent induction of premature senescence and pro-cancerogenic conversion of normal peritoneal mesothelium and fibroblasts. J Pathol 2024; 262:198-211. [PMID: 37941520 DOI: 10.1002/path.6223] [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: 05/22/2023] [Revised: 09/08/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023]
Abstract
Carboplatin (CPT) and paclitaxel (PCT) are the optimal non-surgical treatment of epithelial ovarian cancer (EOC). Although their growth-restricting influence on EOC cells is well known, their impact on normal peritoneal cells, including mesothelium (PMCs) and fibroblasts (PFBs), is poorly understood. Here, we investigated whether, and if so, by what mechanism, CPT and PCT induce senescence of omental PMCs and PFBs. In addition, we tested whether PMC and PFB exposure to the drugs promotes the development of a pro-cancerogenic phenotype. The results showed that CPT and PCT induce G2/M growth arrest-associated senescence of normal peritoneal cells and that the strongest induction occurs when the drugs act together. PMCs senesce telomere-independently with an elevated p16 level and via activation of AKT and STAT3. In PFBs, telomeres shorten along with an induction of p21 and p53, and their senescence proceeds via the activation of ERK1/2. Oxidative stress in CPT + PCT-treated PMCs and PFBs is extensive and contributes causatively to their premature senescence. Both PMCs and PFBs exposed to CPT + PCT fuel the proliferation, migration, and invasion of established (A2780, OVCAR-3, SKOV-3) and primary EOCs, and this activity is linked with an overproduction of multiple cytokines altering the cancer cell transcriptome and controlled by p38 MAPK, NF-κB, STAT3, Notch1, and JAK1. Collectively, our findings indicate that CPT and PCT lead to iatrogenic senescence of normal peritoneal cells, which paradoxically and opposing therapeutic needs alters their phenotype towards pro-cancerogenic. It cannot be excluded that these adverse outcomes of chemotherapy may contribute to EOC relapse in the case of incomplete tumor eradication and residual disease initiation. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Szymon Rutecki
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
- Poznań University of Medical Sciences Doctoral School, Poznań, Poland
| | | | | | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Daniel Rychlewski
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Łukasz Stryczyński
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Eryk Naumowicz
- General Surgery Ward, Medical Centre HCP, Poznań, Poland
| | - Sebastian Szubert
- Department of Gynecology, Division of Gynecologic Oncology, Poznań University of Medical Sciences, Poznań, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
30
|
Hou J, Zheng Y, Gao C. Regulation of cellular senescence by innate immunity. BIOPHYSICS REPORTS 2023; 9:338-351. [PMID: 38524701 PMCID: PMC10960571 DOI: 10.52601/bpr.2023.230032] [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: 10/30/2023] [Accepted: 01/12/2024] [Indexed: 03/26/2024] Open
Abstract
During the COVID-19 pandemic, the interplay between the processes of immunity and senescence is drawing more and more intensive attention. SARS-CoV-2 infection induces senescence in lung cells, failure to clear infected cells and increased presence of inflammatory factors could lead to a cytokine storm and acute respiratory disease syndrome (ARDS), which together with aging and age-associated disease lead to 70% of COVID-19-related deaths. Studies on how senescence initiates upon viral infection and how to restrict excessive accumulation of senescent cells to avoid harmful inflammation are crucially important. Senescence can induce innate immune signaling, and innate immunity can engage cell senescence. Here, we mainly review the innate immune pathways, such as cGAS-STING, TLRs, NF-κB, and NLRP3 inflammasome, participating in the senescence process. In these pathways, IFN-I and inflammatory factors play key roles. At the end of the review, we propose the strategies by which we can improve the immune function and reduce inflammation based on these findings.
Collapse
Affiliation(s)
- Jinxiu Hou
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity, Shandong Province & Key Laboratory for Experimental Teratology, Ministry of Education, Shandong University, Jinan 250012, China
- Department of Immunology, the School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
31
|
Scheper J, Hildebrand LS, Faulhaber EM, Deloch L, Gaipl US, Symank J, Fietkau R, Distel LV, Hecht M, Jost T. Tumor-specific radiosensitizing effect of the ATM inhibitor AZD0156 in melanoma cells with low toxicity to healthy fibroblasts. Strahlenther Onkol 2023; 199:1128-1139. [PMID: 36229655 PMCID: PMC10673781 DOI: 10.1007/s00066-022-02009-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Despite new treatment options, melanoma continues to have an unfavorable prognosis. DNA damage response (DDR) inhibitors are a promising drug class, especially in combination with chemotherapy (CT) or radiotherapy (RT). Manipulating DNA damage repair during RT is an opportunity to exploit the genomic instability of cancer cells and may lead to radiosensitizing effects in tumors that could improve cancer therapy. METHODS A panel of melanoma-derived cell lines of different origin were used to investigate toxicity-related clonogenic survival, cell death, and cell cycle distribution after treatment with a kinase inhibitor (KI) against ATM (AZD0156) or ATR (VE-822, berzosertib), irradiation with 2 Gy, or a combination of KI plus ionizing radiation (IR). Two fibroblast cell lines generated from healthy skin tissue were used as controls. RESULTS Clonogenic survival indicated a clear radiosensitizing effect of the ATM inhibitor (ATMi) AZD0156 in all melanoma cells in a synergistic manner, but not in healthy tissue fibroblasts. In contrast, the ATR inhibitor (ATRi) VE-822 led to additive enhancement of IR-related toxicity in most of the melanoma cells. Both inhibitors mainly increased cell death induction in combination with IR. In healthy fibroblasts, VE-822 plus IR led to higher cell death rates compared to AZD0156. A significant G2/M block was particularly induced in cancer cells when combining AZD0156 with IR. CONCLUSION ATMi, in contrast to ATRi, resulted in synergistic radiosensitization regarding colony formation in melanoma cancer cells, while healthy tissue fibroblasts were merely affected with respect to cell death induction. In connection with an increased number of melanoma cells in the G2/M phase after ATMi plus IR treatment, ATMi seems to be superior to ATRi in melanoma cancer cell treatments when combined with RT.
Collapse
Affiliation(s)
- Julian Scheper
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Laura S Hildebrand
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Eva-Maria Faulhaber
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
- Translational Radiobiology, Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
- Translational Radiobiology, Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Julia Symank
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Luitpold V Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Markus Hecht
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
| | - Tina Jost
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany.
- Translational Radiobiology, Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
32
|
Song KX, Wang JX, Huang D. Therapy-induced senescent tumor cells in cancer relapse. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:273-278. [PMID: 39036667 PMCID: PMC11256611 DOI: 10.1016/j.jncc.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 07/23/2024] Open
Abstract
Cellular senescence is characterized by a generally irreversible cell cycle arrest and the secretion of bioactive factors known as the senescence-associated secretory phenotype (SASP). In an oncogenic context, senescence is considered a tumor suppressive mechanism as it prevents cell proliferation and inhibits the progression from pre-malignant to malignant disease. However, recent studies have demonstrated that senescent tumor cells, which could spontaneously exist within cancer tissues or arise in response to various cancer interventions (the so-called therapy-induced senescence, TIS), can acquire pro-tumorigenic properties and are capable of driving local and metastatic relapse. This highlights the complex and multifaceted nature of cellular senescence in cancer biology. Here, we summarize the current knowledge of the pathological function of therapy-induced senescent tumor cells and discuss possible mechanisms by which tumor cell senescence contributes to cancer relapse. We also discuss implications for future studies toward targeting these less appreciated cells.
Collapse
Affiliation(s)
- Ke-Xin Song
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jun-Xian Wang
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - De Huang
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
33
|
Wang K, Jiang X, Jiang Y, Liu J, Du Y, Zhang Z, Li Y, Zhao X, Li J, Zhang R. EZH2-H3K27me3-mediated silencing of mir-139-5p inhibits cellular senescence in hepatocellular carcinoma by activating TOP2A. J Exp Clin Cancer Res 2023; 42:320. [PMID: 38008711 PMCID: PMC10680220 DOI: 10.1186/s13046-023-02855-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/08/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Epigenetic alterations play an important role in hepatocellular carcinoma (HCC) development. Enhancer of zeste homolog 2 (EZH2) is a well-known epigenetic modifier that functions as an oncogene in tumors by promoting the H3K27me3-mediated transcriptional repression of tumor suppressor genes. "Senescent cells" has been proposed as a possible core component of the hallmarks of cancer conceptualization. Induction of cell senescence and targeted elimination of these senescent tumor cells are new strategies for tumor therapy. However, the role of EZH2 in regulating cellular senescence remains poorly understood. METHODS Bioinformatics analyses suggested that EZH2 and DNA topoisomerase II alpha (TOP2A) are coexpressed in tumors, including HCC. Kyoto Encyclopedia of Genes and Genome (KEGG) pathway enrichment analyses and gene set enrichment analyses (GSEA) suggests a correlation of EZH2 and TOP2A expression with cellular senescence in HCC. MicroRNA (miRNA) inhibitor and mimics, siRNA, PLKO-shRNA, and plenti6.3-miR-139 were used to upregulate or downregulate the expression of target genes. CCK8, EdU, clone formation, and senescence-associated β-galactosidase (SA-β-gal) staining assays were performed to assess cell proliferation and cellular senescence phenotypes. Dual-luciferase reporter and chromatin immunoprecipitation assays were performed to investigate the targeted binding and inhibition of TOP2A 3' untranslated region (UTR) by miR-139-5p and the DNA enrichment of miR139-5p by EZH2 and H3K27me3. BALB/c nude mice were used to establish a xenograft tumor model and verify the phenotypes upon EZH2 and TOP2A silencing and miR-139 overexpression in vivo. In addition, tissue microarrays were used to analyze the expression patterns and correlations among EZH2, TOP2A, and miR-139-5p expression in HCC. RESULTS Bioinformatics analysis revealed that EZH2 and TOP2A are coexpressed in HCC. In vitro gain- and loss-of-function experiments showed that inhibition of EZH2 and TOP2A induces cellular senescence and inhibits proliferation of HCC cells. In vivo tumorigenesis assays indicated that EZH2 and TOP2A knockdown inhibits tumorigenesis by inducing cellular senescence. Mechanistically, EZH2 promotes TOP2A expression by regulating the H3K27me3-mediated epigenetic silencing of miR-139-5p. TOP2A is a direct target of miR-139-5p, and inhibition of miR-139-5p can reverse the promotion by EZH2 of TOP2A expression. The overexpression of miR-139-5p induces cellular senescence and inhibits proliferation of HCC cells both in vitro and in vivo. Clinically, expression of EZH2 and TOP2A are higher in HCC tissues than in normal tissues, and this high coexpression indicates a worse outcome of patients with HCC. Moreover, expression of EZH2 and TOP2A is significantly correlated with tumor differentiation grade, tumor invasion, and TNM stage in HCC. miR-139-5p expression is lower in HCC tumors than in normal tissues and is correlated with better prognosis of HCC patients. CONCLUSIONS Our study revealed the role of the EZH2/miR-139-5p/TOP2A axis in regulating cellular senescence and cell proliferation in HCC, enriching the molecular mechanisms of EZH2-mediated epigenetic regulation in HCC. Therefore, our results provide insight into the therapeutic potential of targeting EZH2 to induce cellular senescence and then destroy senescent cells for HCC.
Collapse
Affiliation(s)
- Ke Wang
- Department of digestive surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Department of General Surgery, Central Theater Command General Hospital of the Chinese People's Liberation Army, Wuhan, 430064, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xunliang Jiang
- Department of digestive surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu Jiang
- Department of Hepatobiliary Surgery, XI'AN DAXING hospital, Xi'an, 710032, China
| | - Jun Liu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yongtao Du
- Department of digestive surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Zecheng Zhang
- Department of digestive surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yunlong Li
- Department of digestive surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xinhui Zhao
- Department of Thyroid and Breast Surgery, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, 710018, China
| | - Jipeng Li
- Department of digestive surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Experimental Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China.
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
34
|
Xiao S, Qin D, Hou X, Tian L, Yu Y, Zhang R, Lyu H, Guo D, Chen XZ, Zhou C, Tang J. Cellular senescence: a double-edged sword in cancer therapy. Front Oncol 2023; 13:1189015. [PMID: 37771436 PMCID: PMC10522834 DOI: 10.3389/fonc.2023.1189015] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023] Open
Abstract
Over the past few decades, cellular senescence has been identified in cancer patients undergoing chemotherapy and radiotherapy. Senescent cells are generally characterized by permanent cell cycle arrest as a response to endogenous and exogenous stresses. In addition to exiting the cell cycle process, cellular senescence also triggers profound phenotypic changes such as senescence-associated secretory phenotype (SASP), autophagy modulation, or metabolic reprograming. Consequently, cellular senescence is often considered as a tumor-suppressive mechanism that permanently arrests cells at risk of malignant transformation. However, accumulating evidence shows that therapy-induced senescence can promote epithelial-mesenchymal transition and tumorigenesis in neighboring cells, as well as re-entry into the cell cycle and activation of cancer stem cells, thereby promoting cancer cell survival. Therefore, it is particularly important to rapidly eliminate therapy-induced senescent cells in patients with cancer. Here we review the hallmarks of cellular senescence and the relationship between cellular senescence and cancer. We also discuss several pathways to induce senescence in tumor therapy, as well as strategies to eliminate senescent cells after cancer treatment. We believe that exploiting the intersection between cellular senescence and tumor cells is an important means to defeat tumors.
Collapse
Affiliation(s)
- Shuai Xiao
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Dongmin Qin
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Xueyang Hou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Lingli Tian
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Yeping Yu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Dong Guo
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cefan Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| |
Collapse
|
35
|
Billimoria R, Bhatt P. Senescence in cancer: Advances in detection and treatment modalities. Biochem Pharmacol 2023; 215:115739. [PMID: 37562510 DOI: 10.1016/j.bcp.2023.115739] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
Senescence is a form of irreversible cell cycle arrest. Senescence plays a dual role in cancer, as both a tumor suppressor by preventing the growth of damaged cells and a cancer promoter by creating an inflammatory milieu. Stress-induced premature senescence (SIPS) and replicative senescence are the two major sub-types of senescence. Senescence plays a dual role in cancer, depending on the context and kind of senescence involved. SIPS can cause cancer by nurturing an inflammatory environment, whereas replicative senescence may prevent cancer. Major pathways that are involved in senescence are the p53-p21, p16INK4A-Rb pathway along with mTOR, MAPK, and PI3K pathways. The lack of universal senescence markers makes it difficult to identify senescent cells in vivo. A combination of reliable detection methods of senescent cells in vivo is of utmost importance and will help in early detection and open new avenues for future treatment. New strategies that are being developed in order to tackle these shortcomings are in the field of fluorescent probes, nanoparticles, positron emission tomography probes, biosensors, and the detection of cell-free DNA from liquid biopsies. Along with detection, eradication of these senescent cells is also important to prevent cancer reoccurrence. Recently, the field of nano-senolytic and immunotherapy has also been emerging. This review provides up-to-date information on the various types of advancements made in the field of detection and treatment modalities for senescent cells that hold promise for the future treatment and prognosis of cancer, as well as their limitations and potential solutions.
Collapse
Affiliation(s)
- Rezina Billimoria
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Vile Parle (West), Mumbai, India
| | - Purvi Bhatt
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Vile Parle (West), Mumbai, India.
| |
Collapse
|
36
|
Yang K, Li X, Xie K. Senescence program and its reprogramming in pancreatic premalignancy. Cell Death Dis 2023; 14:528. [PMID: 37591827 PMCID: PMC10435572 DOI: 10.1038/s41419-023-06040-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
Tumor is a representative of cell immortalization, while senescence irreversibly arrests cell proliferation. Although tumorigenesis and senescence seem contrary to each other, they have similar mechanisms in many aspects. Pancreatic ductal adenocarcinoma (PDA) is highly lethal disease, which occurs and progresses through a multi-step process. Senescence is prevalent in pancreatic premalignancy, as manifested by decreased cell proliferation and increased clearance of pre-malignant cells by immune system. However, the senescent microenvironment cooperates with multiple factors and significantly contributes to tumorigenesis. Evidently, PDA progression requires to evade the effects of cellular senescence. This review will focus on dual roles that senescence plays in PDA development and progression, the signaling effectors that critically regulate senescence in PDA, the identification and reactivation of molecular targets that control senescence program for the treatment of PDA.
Collapse
Affiliation(s)
- Kailing Yang
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, China
| | - Xiaojia Li
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, China
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, China.
- The South China University of Technology Comprehensive Cancer Center, Guangdong, China.
- The Second Affiliated Hospital and Guangzhou First People's Hospital, South China University of Technology School of Medicine, Guangdong, China.
| |
Collapse
|
37
|
Lv M, Cai D, Li C, Chen J, Li G, Hu C, Gai B, Lei J, Lan P, Wu X, He X, Gao F. Senescence-based colorectal cancer subtyping reveals distinct molecular characteristics and therapeutic strategies. MedComm (Beijing) 2023; 4:e333. [PMID: 37502611 PMCID: PMC10369159 DOI: 10.1002/mco2.333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Cellular senescence has been listed as a hallmark of cancer, but its role in colorectal cancer (CRC) remains unclear. We comprehensively evaluated the transcriptome, genome, digital pathology, and clinical data from multiple datasets of CRC patients and proposed a novel senescence subtype for CRC. Multi-omics data was used to analyze the biological features, tumor microenvironment, and mutation landscape of senescence subtypes, as well as drug sensitivity and immunotherapy response. The senescence score was constructed to better quantify senescence in each patient for clinical use. Unsupervised learning revealed three transcriptome-based senescence subtypes. Cluster 1, characterized by low senescence and activated proliferative pathways, was sensitive to chemotherapeutic drugs. Cluster 2, characterized by intermediate senescence and high immune infiltration, exhibited significant immunotherapeutic advantages. Cluster 3, characterized by high senescence, high immune, and stroma infiltration, had a worse prognosis and maybe benefit from targeted therapy. We further constructed a senescence scoring system based on seven senescent genes through machine learning. Lower senescence scores were highly predictive of longer disease-free survival, and patients with low senescence scores may benefit from immunotherapy. We proposed the senescence subtypes of CRC and our findings provide potential treatment interventions for each CRC senescence subtype to promote precision treatment.
Collapse
Affiliation(s)
- Min‐Yi Lv
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Du Cai
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Cheng‐Hang Li
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Junguo Chen
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Guanman Li
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Chuling Hu
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Baowen Gai
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Jiaxin Lei
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Ping Lan
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Xiaojian Wu
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Xiaosheng He
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| | - Feng Gao
- Department of Genaral Surgery (Colorectal Surgery)The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Biomedical Innovation CenterThe Sixth Affiliated Hospital,Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
38
|
Malayaperumal S, Marotta F, Kumar MM, Somasundaram I, Ayala A, Pinto MM, Banerjee A, Pathak S. The Emerging Role of Senotherapy in Cancer: A Comprehensive Review. Clin Pract 2023; 13:838-852. [PMID: 37489425 PMCID: PMC10366900 DOI: 10.3390/clinpract13040076] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023] Open
Abstract
Senotherapy, a promising therapeutic strategy, has drawn a lot attention recently due to its potential for combating cancer. Senotherapy refers to the targeting of senescent cells to restore tissue homeostasis and mitigate the deleterious effects associated with senescence. Senolytic drugs represent a promising avenue in cancer treatment, with the potential to target and modulate senescent cells to improve patient outcomes. The review highlights the intricate interplay between the senescence-associated secretory phenotype (SASP) and the tumor microenvironment, emphasizing the role of senescent cells in promoting chronic inflammation, immune evasion, and tumor-cell proliferation. It then explores the potential of senotherapy as a novel strategy for cancer therapy. This review addresses the emerging evidence on the combination of senotherapy with conventional cancer treatments, such as chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Sarubala Malayaperumal
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, India; (S.M.); (M.M.K.); (A.B.)
| | - Francesco Marotta
- ReGenera R&D International for Aging Intervention, 20154 Milan, Italy
| | - Makalakshmi Murali Kumar
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, India; (S.M.); (M.M.K.); (A.B.)
| | | | - Antonio Ayala
- Department of Biochemistry and Molecular Biology, University of Seville, 41012 Seville, Spain; (A.A.); (M.M.P.)
| | - Mario Munoz Pinto
- Department of Biochemistry and Molecular Biology, University of Seville, 41012 Seville, Spain; (A.A.); (M.M.P.)
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, India; (S.M.); (M.M.K.); (A.B.)
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, India; (S.M.); (M.M.K.); (A.B.)
| |
Collapse
|
39
|
Gong Q, Jiang Y, Xiong J, Liu F, Guan J. Integrating scRNA and bulk-RNA sequencing develops a cell senescence signature for analyzing tumor heterogeneity in clear cell renal cell carcinoma. Front Immunol 2023; 14:1199002. [PMID: 37503331 PMCID: PMC10370498 DOI: 10.3389/fimmu.2023.1199002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/14/2023] [Indexed: 07/29/2023] Open
Abstract
Introduction Cellular senescence (CS) plays a critical role in cancer development, including clear cell renal cell carcinoma (ccRCC). Traditional RNA sequencing cannot detect precise molecular composition changes within tumors. This study aimed to analyze cellular senescence's biochemical characteristics in ccRCC using single RNA sequencing (ScRNA-seq) and traditional RNA sequencing (Bulk RNA-seq). Methods Researchers analyzed the biochemical characteristics of cellular senescence in ccRCC using ScRNA-seq and Bulk RNA-seq. They combined these approaches to identify differences between malignant and non-malignant phenotypes in ccRCC across three senescence-related pathways. Genes from these pathways were used to identify molecular subtypes associated with senescence, and a new risk model was constructed. The function of the gene DUSP1 in ccRCC was validated through biological experiments. Results The combined analysis of ScRNA-seq and Bulk RNA-seq revealed significant differences between malignant and non-malignant phenotypes in ccRCC across three senescence-related pathways. Researchers identified genes from these pathways to identify molecular subtypes associated with senescence, constructing a new risk model. Different subgroups showed significant differences in prognosis level, clinical stage and grade, immune infiltration, immunotherapy, and drug sensitivity. Discussion Senescence signature markers are practical biomarkers and predictors of molecular typing in ccRCC. Differences in prognosis level, clinical stage and grade, immune infiltration, immunotherapy, and drug sensitivity between different subgroups indicate that this approach could provide valuable insights into senescence-related treatment options and prognostic assessment for patients with ccRCC. The function of the gene DUSP1 in ccRCC was validated through biological experiments, confirming its feasibility as a novel biomarker for ccRCC. These findings suggest that targeted therapies based on senescence-related mechanisms could be an effective treatment option for ccRCC.
Collapse
Affiliation(s)
- Qiming Gong
- Department of Pediatric Oncology Surgery, Zhengzhou Key Laboratory of Precise Diagnosis and Treatment of Children’s Malignant Tumors, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Yan Jiang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Junfeng Xiong
- Department of Pediatric Oncology Surgery, Zhengzhou Key Laboratory of Precise Diagnosis and Treatment of Children’s Malignant Tumors, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Fahui Liu
- Department of Pediatric Oncology Surgery, Zhengzhou Key Laboratory of Precise Diagnosis and Treatment of Children’s Malignant Tumors, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jikui Guan
- Department of Pediatric Oncology Surgery, Zhengzhou Key Laboratory of Precise Diagnosis and Treatment of Children’s Malignant Tumors, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
40
|
Prieto LI, Sturmlechner I, Graves SI, Zhang C, Goplen NP, Yi ES, Sun J, Li H, Baker DJ. Senescent alveolar macrophages promote early-stage lung tumorigenesis. Cancer Cell 2023; 41:1261-1275.e6. [PMID: 37267954 PMCID: PMC10524974 DOI: 10.1016/j.ccell.2023.05.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 03/07/2023] [Accepted: 05/05/2023] [Indexed: 06/04/2023]
Abstract
Senescent cells play relevant but context-dependent roles during tumorigenesis. Here, in an oncogenic Kras-driven lung cancer mouse model, we found that senescent cells, specifically alveolar macrophages, accumulate early in neoplasia. These macrophages have upregulated expression of p16INK4a and Cxcr1, are distinct from previously defined subsets and are sensitive to senolytic interventions, and suppress cytotoxic T cell responses. Their removal attenuates adenoma development and progression in mice, indicating their tumorigenesis-promoting role. Importantly, we found that alveolar macrophages with these properties increase with normal aging in mouse lung and in human lung adenocarcinoma in situ. Collectively, our study indicates that a subset of tissue-resident macrophages can support neoplastic transformation through altering their local microenvironment, suggesting that therapeutic interventions targeting senescent macrophages may attenuate lung cancer progression during early stages of disease.
Collapse
Affiliation(s)
- Luis I Prieto
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Ines Sturmlechner
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Sara I Graves
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; Paul F. Glenn Center for Biology of Aging Research at Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Nick P Goplen
- Division of Pulmonary and Critical Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; The Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Eunhee S Yi
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Jie Sun
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; Division of Pulmonary and Critical Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; The Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; Paul F. Glenn Center for Biology of Aging Research at Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Darren J Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; Paul F. Glenn Center for Biology of Aging Research at Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; The Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA.
| |
Collapse
|
41
|
Lucas V, Cavadas C, Aveleira CA. Cellular Senescence: From Mechanisms to Current Biomarkers and Senotherapies. Pharmacol Rev 2023; 75:675-713. [PMID: 36732079 DOI: 10.1124/pharmrev.122.000622] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/29/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
An increase in life expectancy in developed countries has led to a surge of chronic aging-related diseases. In the last few decades, several studies have provided evidence of the prominent role of cellular senescence in many of these pathologies. Key traits of senescent cells include cell cycle arrest, apoptosis resistance, and secretome shift to senescence-associated secretory phenotype resulting in increased secretion of various intermediate bioactive factors important for senescence pathophysiology. However, cellular senescence is a highly phenotypically heterogeneous process, hindering the discovery of totally specific and accurate biomarkers. Also, strategies to prevent the pathologic effect of senescent cell accumulation during aging by impairing senescence onset or promoting senescent cell clearance have shown great potential during in vivo studies, and some are already in early stages of clinical translation. The adaptability of these senotherapeutic approaches to human application has been questioned due to the lack of proper senescence targeting and senescence involvement in important physiologic functions. In this review, we explore the heterogeneous phenotype of senescent cells and its influence on the expression of biomarkers currently used for senescence detection. We also discuss the current evidence regarding the efficacy, reliability, development stage, and potential for human applicability of the main existing senotherapeutic strategies. SIGNIFICANCE STATEMENT: This paper is an extensive review of what is currently known about the complex process of cellular senescence and explores its most defining features. The main body of the discussion focuses on how the multifeature fluctuation of the senescence phenotype and the physiological role of cellular senescence have both caused a limitation in the search for truly reliable senescence biomarkers and the progression in the development of senotherapies.
Collapse
Affiliation(s)
- Vasco Lucas
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| | - Célia Alexandra Aveleira
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
42
|
Kim JY, Rosenberger MG, Rutledge NS, Esser-Kahn AP. Next-Generation Adjuvants: Applying Engineering Methods to Create and Evaluate Novel Immunological Responses. Pharmaceutics 2023; 15:1687. [PMID: 37376133 PMCID: PMC10300703 DOI: 10.3390/pharmaceutics15061687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Adjuvants are a critical component of vaccines. Adjuvants typically target receptors that activate innate immune signaling pathways. Historically, adjuvant development has been laborious and slow, but has begun to accelerate over the past decade. Current adjuvant development consists of screening for an activating molecule, formulating lead molecules with an antigen, and testing this combination in an animal model. There are very few adjuvants approved for use in vaccines, however, as new candidates often fail due to poor clinical efficacy, intolerable side effects, or formulation limitations. Here, we consider new approaches using tools from engineering to improve next-generation adjuvant discovery and development. These approaches will create new immunological outcomes that will be evaluated with novel diagnostic tools. Potential improved immunological outcomes include reduced vaccine reactogenicity, tunable adaptive responses, and enhanced adjuvant delivery. Evaluations of these outcomes can leverage computational approaches to interpret "big data" obtained from experimentation. Applying engineering concepts and solutions will provide alternative perspectives, further accelerating the field of adjuvant discovery.
Collapse
Affiliation(s)
| | | | | | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA; (J.Y.K.); (M.G.R.); (N.S.R.)
| |
Collapse
|
43
|
Garbarino O, Lambroia L, Basso G, Marrella V, Franceschini B, Soldani C, Pasqualini F, Giuliano D, Costa G, Peano C, Barbarossa D, Annarita D, Salvati A, Terracciano L, Torzilli G, Donadon M, Faggioli F. Spatial resolution of cellular senescence dynamics in human colorectal liver metastasis. Aging Cell 2023:e13853. [PMID: 37157887 PMCID: PMC10352575 DOI: 10.1111/acel.13853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 05/10/2023] Open
Abstract
Hepatic metastasis is a clinical challenge for colorectal cancer (CRC). Senescent cancer cells accumulate in CRC favoring tumor dissemination. Whether this mechanism progresses also in metastasis is unexplored. Here, we integrated spatial transcriptomics, 3D-microscopy, and multicellular transcriptomics to study the role of cellular senescence in human colorectal liver metastasis (CRLM). We discovered two distinct senescent metastatic cancer cell (SMCC) subtypes, transcriptionally located at the opposite pole of epithelial (e) to mesenchymal (m) transition. SMCCs differ in chemotherapy susceptibility, biological program, and prognostic roles. Mechanistically, epithelial (e)SMCC initiation relies on nucleolar stress, whereby c-myc dependent oncogene hyperactivation induces ribosomal RPL11 accumulation and DNA damage response. In a 2D pre-clinical model, we demonstrated that RPL11 co-localized with HDM2, a p53-specific ubiquitin ligase, leading to senescence activation in (e)SMCCs. On the contrary, mesenchymal (m)SMCCs undergo TGFβ paracrine activation of NOX4-p15 effectors. SMCCs display opposing effects also in the immune regulation of neighboring cells, establishing an immunosuppressive environment or leading to an active immune workflow. Both SMCC signatures are predictive biomarkers whose unbalanced ratio determined the clinical outcome in CRLM and CRC patients. Altogether, we provide a comprehensive new understanding of the role of SMCCs in CRLM and highlight their potential as new therapeutic targets to limit CRLM progression.
Collapse
Affiliation(s)
| | - Luca Lambroia
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gianluca Basso
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Veronica Marrella
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Institute of Genetics and Biomedical Research, UoS of Milan, National Research Council, Milan, Italy
| | - Barbara Franceschini
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Cristiana Soldani
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Fabio Pasqualini
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele (MI), Italy
| | | | - Guido Costa
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele (MI), Italy
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, UoS of Milan, National Research Council, Milan, Italy
- Fondazione Human Technopole, Milan, Italy
| | | | - Destro Annarita
- Department of Pathology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Andreina Salvati
- Department of Pathology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luigi Terracciano
- Department of Biomedical Science, Humanitas University, Pieve Emanuele (MI), Italy
- Department of Pathology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Guido Torzilli
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele (MI), Italy
| | - Matteo Donadon
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele (MI), Italy
| | - Francesca Faggioli
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Institute of Genetics and Biomedical Research, UoS of Milan, National Research Council, Milan, Italy
| |
Collapse
|
44
|
Du PY, Gandhi A, Bawa M, Gromala J. The ageing immune system as a potential target of senolytics. OXFORD OPEN IMMUNOLOGY 2023; 4:iqad004. [PMID: 37255929 PMCID: PMC10191675 DOI: 10.1093/oxfimm/iqad004] [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: 01/29/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Ageing leads to a sharp decline in immune function, precipitating the development of inflammatory conditions. The combined impact of these processes renders older individuals at greater risk of inflammatory and immune-related diseases, such as cancer and infections. This is compounded by reduced efficacy in interventions aiming to limit disease impact, for instance vaccines being less effective in elderly populations. This state of diminished cellular function is driven by cellular senescence, a process where cells undergo stable growth arrest following exposure to stressful stimuli, and the associated pro-inflammatory secretory phenotype. Removing harmful senescent cells (SnCs) using senolytic therapies is an emerging field holding promise for patient benefit. Current senolytics have been developed either to specifically target SnCs, or repurposed from cancer therapies or vaccination protocols. Herein, we discuss recent developments in senolytic therapies, focusing on how senolytics could be used to combat the age-associated diminution of the immune system. In particular, exploring how these drugs may be used to promote immunity in the elderly, and highlighting recent trials of senolytics in idiopathic pulmonary fibrosis and diabetic kidney disease. Novel immunotherapeutic approaches including chimeric antigen receptor T-cells or monoclonal antibodies targeting SnCs are being investigated to combat the shortcomings of current senolytics and their adverse effects. The flexible nature of senolytic treatment modalities and their efficacy in safely removing harmful SnCs could have great potential to promote healthy immune function in ageing populations.
Collapse
Affiliation(s)
- Peter Yandi Du
- Correspondence address. Faculty of Medicine, Imperial College London, Level 2, Faculty Building, South Kensington Campus, London SW7 2AZ, UK. Tel: +44 (0)20 3313 8213, E-mail:
| | | | | | | |
Collapse
|
45
|
Koziej P, Kluszczynska K, Hartman ML, Czyz M. Trametinib-Resistant Melanoma Cells Displaying MITF high/NGFR low/IL-8 low Phenotype Are Highly Responsive to Alternating Periods of Drug Withdrawal and Drug Rechallenge. Int J Mol Sci 2023; 24:ijms24097891. [PMID: 37175614 PMCID: PMC10178474 DOI: 10.3390/ijms24097891] [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: 03/19/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Despite significant advances in targeted therapies against the hyperactivated BRAFV600/MEK pathway for patients with unresectable metastatic melanoma, acquired resistance remains an unsolved clinical problem. In this study, we focused on melanoma cells resistant to trametinib, an agent broadly used in combination therapies. Molecular and cellular changes were assessed during alternating periods of trametinib withdrawal and rechallenge in trametinib-resistant cell lines displaying either a differentiation phenotype (MITFhigh/NGFRlow) or neural crest stem-like dedifferentiation phenotype (NGFRhigh/MITFlow). Neither drug withdrawal nor drug rechallenge induced cell death, and instead of loss of fitness, trametinib-resistant melanoma cells adapted to altered conditions by phenotype switching. In resistant cells displaying a differentiation phenotype, trametinib withdrawal markedly decreased MITF level and activity, which was associated with reduced cell proliferation capacity, and induced stemness assessed as NGFR-positive cells and senescence features, including IL-8 expression and secretion. All these changes could be reversed by trametinib re-exposure, which emphasizes melanoma cell plasticity. Trametinib-resistant cells displaying a dedifferentiation phenotype were less responsive presumably due to the already low level of MITF, a master regulator of the melanoma phenotype. Considering new directions of the development of anti-melanoma treatment, our study suggests that the phenotype of melanomas resistant to targeted therapy might be a crucial determinant of the selection of second-line therapy for melanoma patients.
Collapse
Affiliation(s)
- Paulina Koziej
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Katarzyna Kluszczynska
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Mariusz L Hartman
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| |
Collapse
|
46
|
Zhang S, Zheng Y, Li X, Zhang S, Hu H, Kuang W. Cellular senescence-related gene signature as a valuable predictor of prognosis in hepatocellular carcinoma. Aging (Albany NY) 2023; 15:3064-3093. [PMID: 37059592 DOI: 10.18632/aging.204658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a lethal tumor. Its prognosis prediction remains a challenge. Meanwhile, cellular senescence, one of the hallmarks of cancer, and its related prognostic genes signature can provide critical information for clinical decision-making. METHOD Using bulk RNA sequencing and microarray data of HCC samples, we established a senescence score model via multi-machine learning algorithms to predict the prognosis of HCC. Single-cell and pseudo-time trajectory analyses were used to explore the hub genes of the senescence score model in HCC sample differentiation. RESULT A machine learning model based on cellular senescence gene expression profiles was identified in predicting HCC prognosis. The feasibility and accuracy of the senescence score model were confirmed in external validation and comparison with other models. Moreover, we analyzed the immune response, immune checkpoints, and sensitivity to immunotherapy drugs of HCC patients in different prognostic risk groups. Pseudo-time analyses identified four hub genes in HCC progression, including CDCA8, CENPA, SPC25, and TTK, and indicated related cellular senescence. CONCLUSIONS This study identified a prognostic model of HCC by cellular senescence-related gene expression and insight into novel potential targeted therapies.
Collapse
Affiliation(s)
- Shuqiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yilu Zheng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xinyu Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shijun Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao Hu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, The First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
47
|
Cherradi S, Garambois V, Marines J, Andrade AF, Fauvre A, Morand O, Fargal M, Mancouri F, Ayrolles-Torro A, Vezzo-Vié N, Jarlier M, Loussaint G, Huvelle S, Joubert N, Mazard T, Gongora C, Pourquier P, Boissière-Michot F, Rio MD. Improving the response to oxaliplatin by targeting chemotherapy-induced CLDN1 in resistant metastatic colorectal cancer cells. Cell Biosci 2023; 13:72. [PMID: 37041570 PMCID: PMC10091849 DOI: 10.1186/s13578-023-01015-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/15/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Tumor resistance is a frequent cause of therapy failure and remains a major challenge for the long-term management of colorectal cancer (CRC). The aim of this study was to determine the implication of the tight junctional protein claudin 1 (CLDN1) in the acquired resistance to chemotherapy. METHODS Immunohistochemistry was used to determine CLDN1 expression in post-chemotherapy liver metastases from 58 CRC patients. The effects of oxaliplatin on membrane CLDN1 expression were evaluated by flow cytometry, immunofluorescence and western blotting experiments in vitro and in vivo. Phosphoproteome analyses, proximity ligation and luciferase reporter assays were used to unravel the mechanism of CLDN1 induction. RNAseq experiments were performed on oxaliplatin-resistant cell lines to investigate the role of CLDN1 in chemoresistance. The "one-two punch" sequential combination of oxaliplatin followed by an anti-CLDN1 antibody-drug conjugate (ADC) was tested in both CRC cell lines and murine models. RESULTS We found a significant correlation between CLDN1 expression level and histologic response to chemotherapy, CLDN1 expression being the highest in resistant metastatic residual cells of patients showing minor responses. Moreover, in both murine xenograft model and CRC cell lines, CLDN1 expression was upregulated after exposure to conventional chemotherapies used in CRC treatment. CLDN1 overexpression was, at least in part, functionally related to the activation of the MAPKp38/GSK3β/Wnt/β-catenin pathway. Overexpression of CLDN1 was also observed in oxaliplatin-resistant CRC cell lines and was associated with resistance to apoptosis, suggesting an anti-apoptotic role for CLDN1. Finally, we demonstrated that the sequential treatment with oxaliplatin followed by an anti-CLDN1 ADC displayed a synergistic effect in vitro and in in vivo. CONCLUSION Our study identifies CLDN1 as a new biomarker of acquired resistance to chemotherapy in CRC patients and suggests that a "one-two punch" approach targeting chemotherapy-induced CLDN1 expression may represent a therapeutic opportunity to circumvent resistance and to improve the outcome of patients with advanced CRC.
Collapse
Affiliation(s)
- Sara Cherradi
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Véronique Garambois
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Johanna Marines
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Augusto Faria Andrade
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Alexandra Fauvre
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Olivia Morand
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Manon Fargal
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Ferial Mancouri
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Adeline Ayrolles-Torro
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Nadia Vezzo-Vié
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Marta Jarlier
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
- Biometry Department, ICM, Montpellier, France
| | - Gerald Loussaint
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Steve Huvelle
- GICC, Team IMT, University of Tours, Tours, 7501, F-37032, France
| | - Nicolas Joubert
- GICC, Team IMT, University of Tours, Tours, 7501, F-37032, France
| | - Thibault Mazard
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Céline Gongora
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Philippe Pourquier
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
| | - Florence Boissière-Michot
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France
- Translational Research Unit, ICM, Montpellier, France
| | - Maguy Del Rio
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier, 208 rue des Apothicaires, Montpellier Cedex 5, F-34298, France.
| |
Collapse
|
48
|
Zhao B, Wu B, Feng N, Zhang X, Zhang X, Wei Y, Zhang W. Aging microenvironment and antitumor immunity for geriatric oncology: the landscape and future implications. J Hematol Oncol 2023; 16:28. [PMID: 36945046 PMCID: PMC10032017 DOI: 10.1186/s13045-023-01426-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023] Open
Abstract
The tumor microenvironment (TME) has been extensively investigated; however, it is complex and remains unclear, especially in elderly patients. Senescence is a cellular response to a variety of stress signals, which is characterized by stable arrest of the cell cycle and major changes in cell morphology and physiology. To the best of our knowledge, senescence leads to consistent arrest of tumor cells and remodeling of the tumor-immune microenvironment (TIME) by activating a set of pleiotropic cytokines, chemokines, growth factors, and proteinases, which constitute the senescence-associated secretory phenotype (SASP). On the one hand, the SASP promotes antitumor immunity, which enhances treatment efficacy; on the other hand, the SASP increases immunosuppressive cell infiltration, including myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), M2 macrophages, and N2 neutrophils, contributing to TIME suppression. Therefore, a deeper understanding of the regulation of the SASP and components contributing to robust antitumor immunity in elderly individuals with different cancer types and the available therapies is necessary to control tumor cell senescence and provide greater clinical benefits to patients. In this review, we summarize the key biological functions mediated by cytokines and intercellular interactions and significant components of the TME landscape, which influence the immunotherapy response in geriatric oncology. Furthermore, we summarize recent advances in clinical practices targeting TME components and discuss potential senescent TME targets.
Collapse
Affiliation(s)
- Binghao Zhao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Departments of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100032, China
| | - Bo Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Nan Feng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiang Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xin Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| |
Collapse
|
49
|
Majeed Y, Madani AY, Altamimi AI, Courjaret R, Vakayil M, Fountain SJ, Machaca K, Mazloum NA. STAT1- and NFAT-independent amplification of purinoceptor function integrates cellular senescence with interleukin-6 production in preadipocytes. Br J Pharmacol 2023; 180:609-627. [PMID: 36321760 DOI: 10.1111/bph.15978] [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: 06/16/2022] [Revised: 09/03/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND AND PURPOSE Senescent preadipocytes promote adipose tissue dysfunction by secreting pro-inflammatory factors, although little is known about the mechanisms regulating their production. We investigated if up-regulated purinoceptor function sensitizes senescent preadipocytes to cognate agonists and how such sensitization regulates inflammation. EXPERIMENTAL APPROACH Etoposide was used to trigger senescence in 3T3-L1 preadipocytes. CRISPR/Cas9 technology or pharmacology allowed studies of transcription factor function. Fura-2 imaging was used for calcium measurements. Interleukin-6 levels were quantified using quantitative PCR and ELISA. Specific agonists and antagonists supported studies of purinoceptor coupling to interleukin-6 production. Experiments in MS1 VEGF angiosarcoma cells and adipose tissue samples from obese mice complemented preadipocyte experiments. KEY RESULTS DNA damage-induced senescence up-regulated purinoceptor expression levels in preadipocytes and MS1 VEGF angiosarcoma cells. ATP-evoked Ca2+ release was potentiated in senescent preadipocytes. ATP enhanced interleukin-6 production, an effect mimicked by ADP but not UTP, in a calcium-independent manner. Senescence-associated up-regulation and activation of the adenosine A3 receptor also enhanced interleukin-6 production. However, nucleotide hydrolysis was not essential because exposure to ATPγS also enhanced interleukin-6 secretion. Pharmacological experiments suggested coupling of P2X ion channels and P2Y12 -P2Y13 receptors to downstream interleukin-6 production. Interleukin-6 signalling exacerbated inflammation during senescence and compromised adipogenesis. CONCLUSIONS AND IMPLICATIONS We report a previously uncharacterized link between cellular senescence and purinergic signalling in preadipocytes and endothelial cancer cells, raising the possibility that up-regulated purinoceptors play key modulatory roles in senescence-associated conditions like obesity and cancer. There is potential for exploitation of specific purinoceptor antagonists as therapeutics in inflammatory disorders.
Collapse
Affiliation(s)
- Yasser Majeed
- Department of Microbiology and Immunology, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Aisha Y Madani
- Department of Microbiology and Immunology, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Ahmed I Altamimi
- Department of Microbiology and Immunology, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Raphael Courjaret
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Muneera Vakayil
- Department of Microbiology and Immunology, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Samuel J Fountain
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Nayef A Mazloum
- Department of Microbiology and Immunology, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| |
Collapse
|
50
|
Karakousis ND, Chrysavgis L, Papatheodoridi A, Legaki AI, Lembessis P, Cholongitas E, Chatzigeorgiou A, Papatheodoridis G. Significance of Circulating Cell-Free DNA Biomarkers in HBeAg-Negative Chronic Hepatitis B Virus Infection and Their Changes after Treatment Initiation. Pathogens 2023; 12:394. [PMID: 36986316 PMCID: PMC10053129 DOI: 10.3390/pathogens12030394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Chronic hepatitis B virus (HBV) infection is a common chronic liver disease that is closely associated with increased morbidity and mortality. Circulating cell-free DNA (cf-DNA) and global DNA methylation, expressed as circulating levels of 5-methyl-2'-deoxycytidine, are increasingly used to monitor chronic inflammatory diseases of several etiologies. This study attempts to investigate the serum levels of circulating cf-DNA and 5-methyl-2'-deoxycytidine in HBeAg-negative patients with chronic infection (carriers) and chronic hepatitis B (CHB), as well as their changes after treatment initiation in CHB. METHODS Serum samples from a total of 61 HBeAg-negative patients (30 carriers and 31 CHB patients) were included in order to quantify the levels of circulating cf-DNA and 5-methyl-2'-deoxycytidine. In addition, serum samples from 17 CHB patients in complete virological and biochemical remission after initiation of treatment with a nucleos(t)ide analogue were included. RESULTS Circulating cf-DNA concentration was significantly increased after the initiation of treatment (15 vs. 10 ng/mL, p = 0.022). There was a trend in higher mean levels of circulating 5-methyl-2'-deoxycytidine in carriers compared to CHB patients (211.02 vs. 175.66 ng/mL, p = 0.089), as well as a trend in increasing 5-methyl-2'-deoxycytidine levels after treatment initiation in CHB patients compared to pre-treatment levels (215 vs. 173 ng/mL, p = 0.079). CONCLUSIONS Both circulating levels of cf-DNA and 5-methyl-2'-deoxycytidine might be useful biomarkers in order to monitor liver disease activity and response to antiviral treatment in HBeAg-negative chronic HBV patients, but further studies are essential in order to validate these intriguing findings.
Collapse
Affiliation(s)
- Nikolaos D. Karakousis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, 11527 Athens, Greece
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Lampros Chrysavgis
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Alkistis Papatheodoridi
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, “Alexandra” General Hospital of Athens, 11528 Athens, Greece
| | - Aigli-Ioanna Legaki
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Panagiotis Lembessis
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Evangelos Cholongitas
- First Department of Internal Medicine, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, 11527 Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George Papatheodoridis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, 11527 Athens, Greece
| |
Collapse
|