1
|
Zarco N, Dovas A, de Araujo Farias V, Nagaiah NK, Haddock A, Sims PA, Hambardzumyan D, Meyer CT, Canoll P, Rosenfeld SS, Kenchappa RS. Resistance to spindle inhibitors in glioblastoma depends on STAT3 and therapy induced senescence. iScience 2024; 27:111311. [PMID: 39640583 PMCID: PMC11617384 DOI: 10.1016/j.isci.2024.111311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/16/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
While mitotic spindle inhibitors specifically kill proliferating tumor cells without the toxicities of microtubule poisons, resistance has limited their clinical utility. Treating glioblastomas with the spindle inhibitors ispinesib, alisertib, or volasertib creates a subpopulation of therapy induced senescent cells that resist these drugs by relying upon the anti-apoptotic and metabolic effects of activated STAT3. Furthermore, these senescent cells expand the repertoire of cells resistant to these drugs by secreting an array of factors, including TGFβ, which induce proliferating cells to exit mitosis and become quiescent-a state that also resists spindle inhibitors. Targeting STAT3 restores sensitivity to each of these drugs by depleting the senescent subpopulation and inducing quiescent cells to enter the mitotic cycle. These results support a therapeutic strategy of targeting STAT3-dependent therapy-induced senescence to enhance the efficacy of spindle inhibitors for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Natanael Zarco
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | | | - Ashley Haddock
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Dolores Hambardzumyan
- Departments of Oncological Sciences and Neurosurgery, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Steven S. Rosenfeld
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rajappa S. Kenchappa
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
2
|
Ghorai A, Singh B, Dutt S. Biphasic DNA damage and non-canonical replication stress response govern radiation-induced senescence in glioblastoma. J Cell Sci 2024; 137:jcs261844. [PMID: 39568404 DOI: 10.1242/jcs.261844] [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: 11/28/2023] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Therapy-induced senescence (TIS) in glioblastoma (GBM) residual disease and escape from TIS account for resistance and recurrence, but the mechanism of TIS manifestation remains obscure. Here, we demonstrate that replication stress (RS) is critical for the induction of TIS in residual cells by employing an in vitro GBM therapy-resistance cellular model. Interestingly, we found a 'biphasic' mode of DNA damage after radiation treatment and reveal that the second phase of DNA damage arises majorly in the S phase of residual cells due to RS. Mechanistically, we show that persistent phosphorylated ATR is a safeguard for radiation resilience, whereas the other canonical RS molecules remain unaltered during the second phase of DNA damage. Importantly, RS preceded the induction of senescence, and ATR inhibition resulted in TIS reduction, leading to apoptosis. Moreover, ATR inhibition sensitized PARP-1 inhibitor-induced enhanced TIS-mediated resistance, leading to cell death. Our study demonstrates the crucial role of RS in TIS induction and maintenance in GBM residual cells, and targeting ATR alone or in combination with a PARP-1 inhibitor will be an effective strategy to eliminate TIS for better treatment outcomes.
Collapse
Affiliation(s)
- Atanu Ghorai
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
| | - Bhawna Singh
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
- Shilpee Dutt Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India
| |
Collapse
|
3
|
Czajkowski K, Herbet M, Murias M, Piątkowska-Chmiel I. Senolytics: charting a new course or enhancing existing anti-tumor therapies? Cell Oncol (Dordr) 2024:10.1007/s13402-024-01018-5. [PMID: 39633108 DOI: 10.1007/s13402-024-01018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
4
|
He Y, Qiu Y, Yang X, Lu G, Zhao SS. Remodeling of tumor microenvironment by cellular senescence and immunosenescence in cervical cancer. Semin Cancer Biol 2024; 108:17-32. [PMID: 39586414 DOI: 10.1016/j.semcancer.2024.11.002] [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: 09/06/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
Cellular senescence is a response to various stress signals, which is characterized by stable cell cycle arrest, alterations in cellular morphology, metabolic reprogramming and production of senescence-associated secretory phenotype (SASP). When it occurs in the immune system, it is called immunosenescence. Cervical cancer is a common gynecological malignancy, and cervical cancer screening is generally recommended before the age of 65. Elderly women (≥65 years) are more often diagnosed with advanced disease and have poorer prognosis compared to younger patients. Despite extensive research, the tumor microenvironment requires more in-depth exploration, particularly in elderly patients. In cervical cancer, senescent cells have a double-edged sword effect on tumor progression. Induction of preneoplastic cell senescence prevents tumor initiation, and several treatment approaches of cervical cancer act in part by inducing cancer cell senescence. However, senescent immune cell populations within the tumor microenvironment facilitate tumor development, recurrence, treatment resistance, etc. Amplification of beneficial effects and inhibition of aging-related pro-tumorigenic pathways contribute to improving antitumor effects. This review discusses senescent cancer and immune cells present in the tumor microenvironment of cervical cancer and how these senescent cells and their SASP remodel the tumor microenvironment, influence antitumor immunity and tumor initiation and development. Moreover, we discuss the significance of senotherapeutics that enable to eliminate senescent cells and prevent tumor progression and development through improving antitumor immunity and affecting the tumor microenvironment.
Collapse
Affiliation(s)
- Yijiang He
- Abdominal Radiation Oncology Ward II, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yue Qiu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Xiansong Yang
- Department of Day Chemotherapy Ward, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, Shandong 266042, China
| | - Guimei Lu
- Department of Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Shan-Shan Zhao
- Department of Gynecology Surgery 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
5
|
Chowdhury SR, Murphy KC, Parikh CN, DeMarco KD, Zhou L, Ruscetti M. Measuring the impact of therapy-induced senescence on NK cell phenotypes in cancer. Methods Cell Biol 2024; 190:171-201. [PMID: 39515879 DOI: 10.1016/bs.mcb.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Cellular senescence is a damage-induced condition characterized by enduring cell cycle arrest and a heightened secretory profile known as the senescence-associated secretory phenotype (SASP). The SASP consists not only of release of inflammatory cytokines and chemokines that attract and activate a diverse repertoire of innate and adaptive immune cells, but also the upregulation of immunomodulatory cell surface molecules that promote immune clearance of senescent cells. Natural Killer (NK) cells are particularly adept at sensing and eliminating senescent cells. In the setting of cancer, commonly administered cytotoxic and cytostatic therapies can elicit senescence and in turn reactivate NK cell immune surveillance against tumors. Here, we detail a series of in vivo, ex vivo, and in vitro assays to assess the impact of therapy-induced senescence on NK cell phenotypes, including their activation, exhaustion, migration, and killing capacity in the context of pancreatic cancer. Importantly, this methodology can be adapted to investigate NK cell biology across various disease states and treatment modalities and help inform NK cell-based immunotherapies for cancer.
Collapse
Affiliation(s)
- Shreya R Chowdhury
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Katherine C Murphy
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Chaitanya N Parikh
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Kelly D DeMarco
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Lin Zhou
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States.
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States.
| |
Collapse
|
6
|
Zarco N, Dovas A, de Araujo Farias V, Nagaiah NK, Haddock A, Sims PA, Hambardzumyan D, Meyer CT, Canoll P, Rosenfeld SS, Kenchappa RS. Resistance to Spindle Inhibitors in Glioblastoma Depends on STAT3 and Therapy Induced Senescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598115. [PMID: 38895402 PMCID: PMC11185785 DOI: 10.1101/2024.06.09.598115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
While mitotic spindle inhibitors specifically kill proliferating tumor cells without the toxicities of microtubule poisons, resistance has limited their clinical utility. Treating glioblastomas with the spindle inhibitors ispinesib, alisertib, or volasertib creates a subpopulation of therapy induced senescent cells that resist these drugs by relying upon the anti-apoptotic and metabolic effects of activated STAT3. Furthermore, these senescent cells expand the repertoire of cells resistant to these drugs by secreting an array of factors, including TGFβ, which induce proliferating cells to exit mitosis and become quiescent-a state that also resists spindle inhibitors. Targeting STAT3 restores sensitivity to each of these drugs by depleting the senescent subpopulation and inducing quiescent cells to enter the mitotic cycle. These results support a therapeutic strategy of targeting STAT3-dependent therapy-induced senescence to enhance the efficacy of spindle inhibitors for the treatment of glioblastoma. Highlights • Resistance to non-microtubule spindle inhibitors limits their efficacy in glioblastoma and depends on STAT3.• Resistance goes hand in hand with development of therapy induced senescence (TIS).• Spindle inhibitor resistant glioblastomas consist of three cell subpopulations-proliferative, quiescent, and TIS-with proliferative cells sensitive and quiescent and TIS cells resistant.• TIS cells secrete TGFβ, which induces proliferative cells to become quiescent, thereby expanding the population of resistant cells in a spindle inhibitor resistant glioblastoma• Treatment with a STAT3 inhibitor kills TIS cells and restores sensitivity to spindle inhibitors.
Collapse
|
7
|
Wang L, Xiong B, Lu W, Cheng Y, Zhu J, Ai G, Zhang X, Liu X, Cheng Z. Senolytic drugs dasatinib and quercetin combined with Carboplatin or Olaparib reduced the peritoneal and adipose tissue metastasis of ovarian cancer. Biomed Pharmacother 2024; 174:116474. [PMID: 38518604 DOI: 10.1016/j.biopha.2024.116474] [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: 10/29/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024] Open
Abstract
Chemotherapy and targeted drugs-induced senescent ovarian cancer cells that accumulate in peritoneal adipose tissue contribute significantly to chronic inflammation, disrupt homeostasis, and may fuel various aspects of cancer progression. However, the pro-senescence effects of chemotherapy and targeted drugs on adipose derived stem cells (ADSCs) within peritoneal adipose tissue remain poorly understood. In this study, we show that the first-line chemotherapy and targeted drugs can induce the cellular senescence of ADSCs in vitro and increase the aging of peritoneal adipose tissue in vivo. These treatments significantly promoted the dysregulation of glucose and lipid metabolism, including insulin resistance and liver lipid accumulation. Our study shows that dasatinib and quercetin, as senolytics, effectively restore glucose homeostasis in mice with ovarian cancer and significantly reduce adipose tissue aging. Importantly, combining these drugs with Carboplatin or Olaparib results in a marked decrease in both peritoneal and adipose tissue metastasis of ovarian cancer cells. Mechanistically, we revealed that there is crosstalk between ovarian cancer cells and senescent ADSCs. The crosstalk increases inflammatory cytokines and chemokines production in ADSCs and notably upregulates chemokine receptors on cancer cells. Collectively, these data indicate that senescent ADSCs induced by chemotherapy and targeted therapy drugs impair adipose tissue function. However, the senolytic drugs dasatinib and quercetin, can significantly ameliorate organ aging and damage induced by these treatments. Notably, dasatinib and quercetin combined with Carboplatin or Olaparib reduced the peritoneal and adipose tissue metastasis of ovarian cancer, ultimately benefiting the mice undergoing chemotherapy and targeted therapy.
Collapse
Affiliation(s)
- Lian Wang
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Tongji University, School of Medicine, Shanghai, China
| | - Bing Xiong
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Wei Lu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; School of medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Yujie Cheng
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; School of medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Jihui Zhu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Guihai Ai
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaojie Zhang
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Gynecology, Jing'an District Hospital of Traditional Chinese Medicine, Shanghai 200072, China.
| | - Xiuni Liu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Zhongping Cheng
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Tongji University, School of Medicine, Shanghai, China; School of medicine, Anhui University of Science and Technology, Huainan 232001, China.
| |
Collapse
|
8
|
Elshazly AM, Shahin U, Al Shboul S, Gewirtz DA, Saleh T. A Conversation with ChatGPT on Contentious Issues in Senescence and Cancer Research. Mol Pharmacol 2024; 105:313-327. [PMID: 38458774 PMCID: PMC11026153 DOI: 10.1124/molpharm.124.000871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/10/2024] Open
Abstract
Artificial intelligence (AI) platforms, such as Generative Pretrained Transformer (ChatGPT), have achieved a high degree of popularity within the scientific community due to their utility in providing evidence-based reviews of the literature. However, the accuracy and reliability of the information output and the ability to provide critical analysis of the literature, especially with respect to highly controversial issues, has generally not been evaluated. In this work, we arranged a question/answer session with ChatGPT regarding several unresolved questions in the field of cancer research relating to therapy-induced senescence (TIS), including the topics of senescence reversibility, its connection to tumor dormancy, and the pharmacology of the newly emerging drug class of senolytics. ChatGPT generally provided responses consistent with the available literature, although occasionally overlooking essential components of the current understanding of the role of TIS in cancer biology and treatment. Although ChatGPT, and similar AI platforms, have utility in providing an accurate evidence-based review of the literature, their outputs should still be considered carefully, especially with respect to unresolved issues in tumor biology. SIGNIFICANCE STATEMENT: Artificial Intelligence platforms have provided great utility for researchers to investigate biomedical literature in a prompt manner. However, several issues arise when it comes to certain unresolved biological questions, especially in the cancer field. This work provided a discussion with ChatGPT regarding some of the yet-to-be-fully-elucidated conundrums of the role of therapy-induced senescence in cancer treatment and highlights the strengths and weaknesses in utilizing such platforms for analyzing the scientific literature on this topic.
Collapse
Affiliation(s)
- Ahmed M Elshazly
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (A.M.E., D.A.G.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt (A.M.E.); and Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan (U.S., S.A.S., T.S.)
| | - Uruk Shahin
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (A.M.E., D.A.G.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt (A.M.E.); and Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan (U.S., S.A.S., T.S.)
| | - Sofian Al Shboul
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (A.M.E., D.A.G.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt (A.M.E.); and Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan (U.S., S.A.S., T.S.)
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (A.M.E., D.A.G.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt (A.M.E.); and Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan (U.S., S.A.S., T.S.)
| | - Tareq Saleh
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (A.M.E., D.A.G.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt (A.M.E.); and Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan (U.S., S.A.S., T.S.)
| |
Collapse
|
9
|
Xiang X, Dong C, Zhou L, Liu J, Rabinowitz ZM, Zhang Y, Guo H, He F, Chen X, Wang Y, Cui L, Ma X. Novel PET Imaging Probe for Quantitative Detection of Senescence In Vivo. J Med Chem 2024; 67:5924-5934. [PMID: 38507820 PMCID: PMC11017977 DOI: 10.1021/acs.jmedchem.4c00179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
Real-time detection of cellular senescence remains a clinical challenge. Here, we aimed to develop a positron emission tomography (PET) imaging probe targeting senescence-associated β-galactosidase (SA-β-Gal), the most widely used biomarker of cellular senescence, and investigate its performance for real-time in vivo quantitative detection of cellular senescence. A stable PET imaging agent [68Ga]Ga-BGal was obtained with a high labeling yield (90.0 ± 4.3%) and a radiochemical purity (>95%). [68Ga]Ga-BGal displayed high sensitivity and specificity for β-Gal both in vitro and in vivo. The reaction and uptake of the probe correlated with the β-Gal concentration and reaction time. In PET imaging, high β-Gal-expressing CT26.CL25 tumors and doxorubicin-treated HeLa tumors showed high signals from [68Ga]Ga-BGal, while a low signal was observed in CT26.WT and untreated HeLa tumors. In summary, we showcased successful PET imaging of senescence in preclinical models using probe [68Ga]Ga-BGal. This finding holds the potential for translating senescence imaging into clinical applications.
Collapse
Affiliation(s)
- Xin Xiang
- Department
of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Chuning Dong
- Department
of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Lianbo Zhou
- Department
of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jun Liu
- Department
of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, University of Florida, Gainesville, Florida 32610, United States
| | - Zachary M. Rabinowitz
- Department
of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, University of Florida, Gainesville, Florida 32610, United States
| | - Yuzhao Zhang
- Department
of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, University of Florida, Gainesville, Florida 32610, United States
| | - Honghui Guo
- Department
of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Feng He
- Department
of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xingdou Chen
- Department
of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yunhua Wang
- Department
of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Lina Cui
- Department
of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, University of Florida, Gainesville, Florida 32610, United States
| | - Xiaowei Ma
- Department
of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
10
|
McGrath MK, Abolhassani A, Guy L, Elshazly AM, Barrett JT, Mivechi NF, Gewirtz DA, Schoenlein PV. Autophagy and senescence facilitate the development of antiestrogen resistance in ER positive breast cancer. Front Endocrinol (Lausanne) 2024; 15:1298423. [PMID: 38567308 PMCID: PMC10986181 DOI: 10.3389/fendo.2024.1298423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Estrogen receptor positive (ER+) breast cancer is the most common breast cancer diagnosed annually in the US with endocrine-based therapy as standard-of-care for this breast cancer subtype. Endocrine therapy includes treatment with antiestrogens, such as selective estrogen receptor modulators (SERMs), selective estrogen receptor downregulators (SERDs), and aromatase inhibitors (AIs). Despite the appreciable remission achievable with these treatments, a substantial cohort of women will experience primary tumor recurrence, subsequent metastasis, and eventual death due to their disease. In these cases, the breast cancer cells have become resistant to endocrine therapy, with endocrine resistance identified as the major obstacle to the medical oncologist and patient. To combat the development of endocrine resistance, the treatment options for ER+, HER2 negative breast cancer now include CDK4/6 inhibitors used as adjuvants to antiestrogen treatment. In addition to the dysregulated activity of CDK4/6, a plethora of genetic and biochemical mechanisms have been identified that contribute to endocrine resistance. These mechanisms, which have been identified by lab-based studies utilizing appropriate cell and animal models of breast cancer, and by clinical studies in which gene expression profiles identify candidate endocrine resistance genes, are the subject of this review. In addition, we will discuss molecular targeting strategies now utilized in conjunction with endocrine therapy to combat the development of resistance or target resistant breast cancer cells. Of approaches currently being explored to improve endocrine treatment efficacy and patient outcome, two adaptive cell survival mechanisms, autophagy, and "reversible" senescence, are considered molecular targets. Autophagy and/or senescence induction have been identified in response to most antiestrogen treatments currently being used for the treatment of ER+ breast cancer and are often induced in response to CDK4/6 inhibitors. Unfortunately, effective strategies to target these cell survival pathways have not yet been successfully developed. Thus, there is an urgent need for the continued interrogation of autophagy and "reversible" senescence in clinically relevant breast cancer models with the long-term goal of identifying new molecular targets for improved treatment of ER+ breast cancer.
Collapse
Affiliation(s)
- Michael K. McGrath
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Cellular Biology & Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Ali Abolhassani
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Cellular Biology & Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Luke Guy
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Cellular Biology & Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Ahmed M. Elshazly
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, United States
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - John T. Barrett
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Radiation Oncology, Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Nahid F. Mivechi
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Radiation Oncology, Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - David A. Gewirtz
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, United States
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Patricia V. Schoenlein
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Cellular Biology & Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
11
|
Wang L, Li J, Zhao Z, Xia Y, Xie Y, Hong D, Liu Y, Tan W. Aptamer Conjugate-Based Ratiometric Fluorescent Probe for Precise Imaging of Therapy-Induced Cancer Senescence. Anal Chem 2024; 96:154-162. [PMID: 38113452 DOI: 10.1021/acs.analchem.3c03435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Therapy-induced cellular senescence has been increasingly recognized as a key mechanism to promote various aspects of carcinogenesis in a nonautonomous manner. Thus, real-time imaging monitoring of cellular senescence during cancer therapy is imperative not only to further elucidate its roles in cancer progression but also to provide guidance for medical management of cancer. However, it has long been a challenging task due to the lack of effective imaging molecule tools with high specificity and accuracy toward cancer senescence. Herein, we report the rational design, synthesis, and evaluation of an aptamer conjugate-based ratiometric fluorescent probe for precise imaging of therapy-induced cancer senescence. Unlike traditional senescence imaging systems, our probe targets two senescence-associated markers at both cellular and subcellular dimensions, namely, aptamer-mediated membrane marker recognition for active cell targeting and lysosomal marker-triggered ratiometric fluorescence changes of two cyanine dyes for site-specific, high-contrast imaging. Moreover, such a two-channel fluorescence response is activated after a one-step reaction and at the same location, avoiding the diffusion-caused signal decay previously encountered in dual-marker activated probes, contributing to spatiotemporally specific imaging of therapy-induced cancer senescence in living cells and three-dimensional multicellular tumor spheroids. This work may offer a valuable tool for a basic understanding of cellular senescence in cancer biology and interventions.
Collapse
Affiliation(s)
- Linlin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Jili Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Zhihui Zhao
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yinghao Xia
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yuqi Xie
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Donghui Hong
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
12
|
Alsalem M, Ellaithy A, Bloukh S, Haddad M, Saleh T. Targeting therapy-induced senescence as a novel strategy to combat chemotherapy-induced peripheral neuropathy. Support Care Cancer 2024; 32:85. [PMID: 38177894 DOI: 10.1007/s00520-023-08287-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a treatment-limiting adverse effect of anticancer therapy that complicates the lifestyle of many cancer survivors. There is currently no gold-standard for the assessment or management of CIPN. Subsequently, understanding the underlying mechanisms that lead to the development of CIPN is essential for finding better pharmacological therapy. Therapy-induced senescence (TIS) is a form of senescence that is triggered in malignant and non-malignant cells in response to the exposure to chemotherapy. Recent evidence has also suggested that TIS develops in the dorsal root ganglia of rodent models of CIPN. Interestingly, several components of the senescent phenotype are commensurate with the currently established primary processes implicated in the pathogenesis of CIPN including mitochondrial dysfunction, oxidative stress, and neuroinflammation. In this article, we review the literature that supports the hypothesis that TIS could serve as a holistic mechanism leading to CIPN, and we propose the potential for investigating senotherapeutics as means to mitigate CIPN in cancer survivors.
Collapse
Affiliation(s)
- Mohammad Alsalem
- Department of Anatomy and Histology, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Amr Ellaithy
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah Bloukh
- Department of Anatomy and Histology, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Mansour Haddad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan.
| |
Collapse
|
13
|
Gonzalez-Meljem JM, Martinez-Barbera JP. Implications of cellular senescence in paediatric pituitary tumours. EBioMedicine 2024; 99:104905. [PMID: 38043401 PMCID: PMC10730348 DOI: 10.1016/j.ebiom.2023.104905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023] Open
Abstract
The long-standing view of senescent cells as passive and dysfunctional biological remnants has recently shifted into a new paradigm where they are main players in the development of many diseases, including cancer. The senescence programme represents a first line of defence that prevents tumour cell growth but also leads to the secretion of multiple pro-inflammatory and pro-tumourigenic factors that fuel tumour initiation, growth, and progression. Here, we review the main molecular features and biological functions of senescent cells in cancer, including the outcomes of inducing or targeting senescence. We discuss evidence on the role of cellular senescence in pituitary tumours, with an emphasis on adamantinomatous craniopharyngioma (ACP) and pituitary adenomas. Although senescence has been proposed to be a tumour-preventing mechanism in pituitary adenomas, research in ACP has shown that senescent cells are tumour-promoting in both murine models and human tumours. Future studies characterizing the impact of targeting senescent cells may result in novel therapies against pituitary tumours.
Collapse
Affiliation(s)
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London, UK.
| |
Collapse
|
14
|
Soto-Gamez A, van Es M, Hageman E, Serna-Salas SA, Moshage H, Demaria M, Pringle S, Coppes RP. Mesenchymal stem cell-derived HGF attenuates radiation-induced senescence in salivary glands via compensatory proliferation. Radiother Oncol 2024; 190:109984. [PMID: 37926332 DOI: 10.1016/j.radonc.2023.109984] [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: 06/15/2023] [Revised: 10/21/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND & AIM Irradiation of the salivary glands during head and neck cancer treatment induces cellular senescence in response to DNA damage and contributes to radiation-induced hyposalivation by affecting the salivary gland stem/progenitor cell (SGSC) niche. Cellular senescence, such as that induced by radiation, is a state of cell-cycle arrest, accompanied by an altered pro-inflammatory secretome known as the senescence-associated secretory phenotype (SASP) with potential detrimental effects on the surrounding microenvironment. We hypothesized that the pro-regenerative properties of mesenchymal stem cells (MSCs) may attenuate cellular senescence post-irradiation. Therefore, here we evaluated the effects of adipose-derived MSCs (ADSCs) on the radiation-induced response of salivary gland organoids (SGOs). METHODS Proteomic analyses to identify soluble mediators released by ADSCs co-cultured with SGOS revealed secretion of hepatocyte growth factor (HGF) in ADSCs, suggesting a possible role in the stem cell crosstalk. Next, the effect of recombinant HGF in the culture media of ex vivo grown salivary gland cells was tested in 2D monolayers and 3D organoid models. RESULTS Treatment with HGF robustly increased salivary gland cell proliferation. Importantly, HGF supplementation post-irradiation enhanced proliferation at lower doses of radiation (0, 3, 7 Gy), but not at higher doses (10, 14 Gy) where most cells stained positive for senescence-associated beta-galactosidase. Furthermore, HGF had no effect on the senescence-associated secretory phenotype (SASP) of irradiated SGOs, suggesting there may be compensatory proliferation by cell-division competent cells instead of a reversal of cellular senescence after irradiation. CONCLUSION ADSCs may positively influence radiation recovery through HGF secretion and can promote the ex vivo expansion of salivary gland stem/progenitor cells to enhance the effects of co-transplanted SGSC.
Collapse
Affiliation(s)
- A Soto-Gamez
- Dept. of Biomedical Sciences of Cells & Systems, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Dept. of Radiation Oncology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - M van Es
- Dept. of Biomedical Sciences of Cells & Systems, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Dept. of Radiation Oncology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - E Hageman
- Dept. of Biomedical Sciences of Cells & Systems, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Dept. of Radiation Oncology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - S A Serna-Salas
- Dept of Gastroenterology and Hepatology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - H Moshage
- Dept of Gastroenterology and Hepatology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - M Demaria
- European Research Institute for the Biology of Ageing, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - S Pringle
- Dept. of Rheumatology and Clinical Immunology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - R P Coppes
- Dept. of Biomedical Sciences of Cells & Systems, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Dept. of Radiation Oncology, University of Groningen (RUG) and University Medical Center Groningen (UMCG), Groningen, the Netherlands.
| |
Collapse
|
15
|
Wilczyński J, Paradowska E, Wilczyńska J, Wilczyński M. Prediction of Chemoresistance-How Preclinical Data Could Help to Modify Therapeutic Strategy in High-Grade Serous Ovarian Cancer. Curr Oncol 2023; 31:229-249. [PMID: 38248100 PMCID: PMC10814576 DOI: 10.3390/curroncol31010015] [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: 11/13/2023] [Revised: 12/12/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is one of the most lethal tumors generally and the most fatal cancer of the female genital tract. The approved standard therapy consists of surgical cytoreduction and platinum/taxane-based chemotherapy, and of targeted therapy in selected patients. The main therapeutic problem is chemoresistance of recurrent and metastatic HGSOC tumors which results in low survival in the group of FIGO III/IV. Therefore, the prediction and monitoring of chemoresistance seems to be of utmost importance for the improvement of HGSOC management. This type of cancer has genetic heterogeneity with several subtypes being characterized by diverse gene signatures and disturbed peculiar epigenetic regulation. HGSOC develops and metastasizes preferentially in the specific intraperitoneal environment composed mainly of fibroblasts, adipocytes, and immune cells. Different HGSOC subtypes could be sensitive to distinct sets of drugs. Moreover, primary, metastatic, and recurrent tumors are characterized by an individual biology, and thus diverse drug responsibility. Without a precise identification of the tumor and its microenvironment, effective treatment seems to be elusive. This paper reviews tumor-derived genomic, mutational, cellular, and epigenetic biomarkers of HGSOC drug resistance, as well as tumor microenvironment-derived biomarkers of chemoresistance, and discusses their possible use in the novel complex approach to ovarian cancer therapy and monitoring.
Collapse
Affiliation(s)
- Jacek Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| | - Justyna Wilczyńska
- Department of Tele-Radiotherapy, Mikolaj Kopernik Provincial Multi-Specialized Oncology and Traumatology Center, 62 Pabianicka Str., 93-513 Lodz, Poland;
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| |
Collapse
|
16
|
Thapa BV, Banerjee M, Glimm T, Saini DK, Bhat R. The senescent mesothelial matrix accentuates colonization by ovarian cancer cells. Cell Mol Life Sci 2023; 81:2. [PMID: 38043093 PMCID: PMC10694112 DOI: 10.1007/s00018-023-05017-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 12/05/2023]
Abstract
Ovarian cancer is amongst the most morbid of gynecological malignancies due to its diagnosis at an advanced stage, a transcoelomic mode of metastasis, and rapid transition to chemotherapeutic resistance. Like all other malignancies, the progression of ovarian cancer may be interpreted as an emergent outcome of the conflict between metastasizing cancer cells and the natural defense mounted by microenvironmental barriers to such migration. Here, we asked whether senescence in coelom-lining mesothelia, brought about by drug exposure, affects their interaction with disseminated ovarian cancer cells. We observed that cancer cells adhered faster on senescent human and murine mesothelial monolayers than on non-senescent controls. Time-lapse epifluorescence microscopy showed that mesothelial cells were cleared by a host of cancer cells that surrounded the former, even under sub-confluent conditions. A multiscale computational model predicted that such colocalized mesothelial clearance under sub-confluence requires greater adhesion between cancer cells and senescent mesothelia. Consistent with the prediction, we observed that senescent mesothelia expressed an extracellular matrix with higher levels of fibronectin, laminins and hyaluronan than non-senescent controls. On senescent matrix, cancer cells adhered more efficiently, spread better, and moved faster and persistently, aiding the spread of cancer. Inhibition assays using RGD cyclopeptides suggested the adhesion was predominantly contributed by fibronectin and laminin. These findings led us to propose that the senescence-associated matrisomal phenotype of peritoneal barriers enhances the colonization of invading ovarian cancer cells contributing to the metastatic burden associated with the disease.
Collapse
Affiliation(s)
- Bharat Vivan Thapa
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
- Undergraduate Program, Indian Institute of Science, Bangalore, 560012, India
| | - Mallar Banerjee
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Tilmann Glimm
- Department of Mathematics, Western Washington University, Bellingham, WA, 98229, USA
| | - Deepak K Saini
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India.
| | - Ramray Bhat
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
17
|
Liu X, Lin L, Cai Q, Sheng H, Zeng R, Zhao Y, Qiu X, Liu H, Huang L, Liang W, He J. Construction and Validation of a Prognostic Model Based on Novel Senescence-Related Genes in Non-Small Cell Lung Cancer Patients with Drug Sensitivity and Tumor Microenvironment. Adv Biol (Weinh) 2023; 7:e2300190. [PMID: 37518773 DOI: 10.1002/adbi.202300190] [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: 05/23/2023] [Revised: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Cellular senescence contributes to cancer pathogenesis and immune regulation. Using the LASSO Cox regression, we developed a 12-gene prognostic signature for lung adenocarcinoma (LUAD) from The Cancer Genome Atlas (TCGA) and a Gene Expression Omnibus (GEO) dataset. We assessed gene expression, drug sensitivity, immune infiltration, and conducted cell line experiments. High-risk LUAD patients showed increased mortality risk and shorter survival (P < 0.001). Senescence-related gene analysis indicated differences in protein phosphorylation and DNA methylation between normal individuals and LUAD patients. The high-risk group showed a positive association with PD-L1 expression (P = 0.003). Single-cell sequencing data suggested PEBP1 might significantly impact T cell infiltration. We predicted potential sensitive compounds for 12 senescence genes and found GAPDH promoted cell line proliferation. We established a novel prognostic system based on a newly identified senescence gene. High-risk patients had elevated immunosuppressive markers, and PEBP1 might influence T cell infiltration significantly. GAPDH, expressed at higher levels in tumors, could affect cancer progression. Our drug prediction model may guide treatment selection.
Collapse
Affiliation(s)
- Xiwen Liu
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
| | - Lixuan Lin
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
- School of Clinical Medicine, Henan University, Kaifeng, 475000, China
| | - Qi Cai
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
| | - Hongxu Sheng
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
| | - Ruiqi Zeng
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Yi Zhao
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
| | - Xinyi Qiu
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
- First Clinical School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Huiting Liu
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
| | - Linchong Huang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
- The First People's Hospital of Zhaoqing, Zhaoqing, 526000, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, China
- Southern Medical University, Guangzhou, 510120, China
| |
Collapse
|
18
|
Ni H, Qian S, Lu J, Feng J, Mou XZ, Zhang J. Natural Polysaccharide Delivery Platforms with Multiscale Structure Used for Cancer Chemoimmunotherapy. Mol Pharm 2023; 20:5778-5789. [PMID: 37752866 DOI: 10.1021/acs.molpharmaceut.3c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Chemoimmunotherapy is an effective cancer treatment method. Drugs are always combined and used in treating cancer. However, the characteristic of drugs varies, making it challenging to control their release kinetics utilizing delivery devices with a single microstructure. In this study, we attempted to uniformly size drugs of varying molecular weights and confine them in a compartment where immune cells may be recruited and moved freely. Dextran microgels were created as modular drug libraries to address the cryogel burst release of small molecule drugs. Then, modular drug libraries and granulocyte-macrophage colony-stimulating factor (GM-CSF) were integrated into cryogels for a combined treatment. Herein, alginate was zwitterion modified to avoid the immune reaction generated by the material. Because of its macroporous structure, the cryogel could be injected into the body, eliminating invasive surgical procedures. Results demonstrated that multiscale delivery platforms could improve the synergistic effect of various medications on tumor treatment.
Collapse
Affiliation(s)
- Haifeng Ni
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Sunxiang Qian
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Jie Lu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Jie Feng
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Xiao-Zhou Mou
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang 310014, P. R. China
| | - Jing Zhang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| |
Collapse
|
19
|
Saleh T, Bloukh S, Hasan M, Al Shboul S. Therapy-induced senescence as a component of tumor biology: Evidence from clinical cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188994. [PMID: 37806641 DOI: 10.1016/j.bbcan.2023.188994] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/10/2023]
Abstract
Therapy-Induced Senescence (TIS) is an established response to anticancer therapy in a variety of cancer models. Ample evidence has characterized the triggers, hallmarks, and functional outcomes of TIS in preclinical studies; however, limited evidence delineates TIS in clinical cancer (human tumor samples). We examined the literature that investigated the induction of TIS in samples derived from human cancers and highlighted the major findings that suggested that TIS represents a main constituent of tumor biology. The most frequently utilized approach to identify TIS in human cancers was to investigate the protein expression of senescence-associated markers (such as cyclins, cyclin-dependent kinase inhibitors, Ki67, DNA damage repair response markers, DEC1, and DcR1) via immunohistochemical techniques using formalin-fixed paraffin-embedded (FFPE) tissue samples and/or testing the upregulation of Senescence-Associated β-galactosidase (SA-β-gal) in frozen sections of unfixed tumor samples. Collectively, and in studies where the extent of TIS was determined, TIS was detected in 31-66% of tumors exposed to various forms of chemotherapy. Moreover, TIS was not only limited to both malignant and non-malignant components of tumoral tissue but was also identified in samples of normal (non-transformed) tissue upon chemo- or radiotherapy exposure. Nevertheless, the available evidence continues to be limited and requires a more rigorous assessment of in vivo senescence based on novel approaches and more reliable molecular signatures. The accurate assessment of TIS will be beneficial for determining its relevant contribution to the overall outcome of cancer therapy and the potential translatability of senotherapeutics.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13115, Jordan.
| | - Sarah Bloukh
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Mira Hasan
- Department of Medicine, University of Connecticut Health Center, Farmington, USA
| | - Sofian Al Shboul
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13115, Jordan
| |
Collapse
|
20
|
Sienkiewicz M, Sroka K, Binienda A, Jurk D, Fichna J. A new face of old cells: An overview about the role of senescence and telomeres in inflammatory bowel diseases. Ageing Res Rev 2023; 91:102083. [PMID: 37802318 DOI: 10.1016/j.arr.2023.102083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
Cellular senescence is a pivotal factor contributing to aging and the pathophysiology of age-related diseases. Despite the presence of inflammation and abnormal immune system function in both inflammatory bowel diseases (IBD) and senescence, the relationship between the two remains largely unexplored. Therefore, our study aimed to investigate the intricate connection between cellular senescence, telomeres, and IBD. The review highlights the presence of senescence markers, particularly p16 and p21, in IBD patients, suggesting their potential association with disease progression and mucosal inflammation. We emphasize the critical role of macrophages in eliminating senescent cells and how disturbance in effective clearance may contribute to persistent senescence and inflammation in IBD. Additionally, we shed light on the involvement of telomeres in IBD, as their dysfunction impairs enterocyte function and disrupts colonic barrier integrity, potentially exacerbating the pathogenesis of the disease. Targeting senescence and telomere dysfunctions holds promise for the development of innovative therapeutic approaches to mitigate intestinal inflammation and alleviate symptoms in IBD patients. By unraveling the precise role of senescence in IBD, we can pave the way for the discovery of novel therapeutic interventions that effectively address the underlying mechanisms of intestinal inflammation, offering hope for improved management and treatment of IBD patients.
Collapse
Affiliation(s)
- Michał Sienkiewicz
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Kamila Sroka
- Department of Family Medicine and Public Health, University of Opole, Opole, Poland
| | - Agata Binienda
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Diana Jurk
- Robert and Arlene Kogod Center On Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
21
|
Rubin de Celis MF, Bonner-Weir S. Reversing and modulating cellular senescence in beta cells, a new field of opportunities to treat diabetes. Front Endocrinol (Lausanne) 2023; 14:1217729. [PMID: 37822597 PMCID: PMC10562723 DOI: 10.3389/fendo.2023.1217729] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Diabetes constitutes a world-wide pandemic that requires searching for new treatments to halt its progression. Cellular senescence of pancreatic beta cells has been described as a major contributor to development and worsening of diabetes. The concept of reversibility of cellular senescence is critical as is the timing to take actions against this "dormant" senescent state. The reversal of cellular senescence can be considered as rejuvenation of the specific cell if it returns to the original "healthy state" and doesn't behave aberrantly as seen in some cancer cells. In rodents, treatment with senolytics and senomorphics blunted or prevented disease progression, however their use carry drawbacks. Modulators of cellular senescence is a new area of research that seeks to reverse the senescence. More research in each of these modalities should lead to new treatments to stop diabetes development and progression.
Collapse
Affiliation(s)
- Maria F. Rubin de Celis
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Susan Bonner-Weir
- Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
22
|
Miller D, Kerkhofs K, Abbas-Aghababazadeh F, Madahar SS, Minden MD, Hébert J, Haibe-Kains B, Bayfield MA, Benchimol S. Heterogeneity in leukemia cells that escape drug-induced senescence-like state. Cell Death Dis 2023; 14:503. [PMID: 37543610 PMCID: PMC10404232 DOI: 10.1038/s41419-023-06015-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 07/06/2023] [Accepted: 07/25/2023] [Indexed: 08/07/2023]
Abstract
Erythropoietin (EPO) suppresses drug-induced apoptosis in EPO-receptor-positive leukemia cells and allows cells to persist after drug treatment by promoting cellular senescence. Importantly a small proportion of senescent cells can re-enter the cell cycle and resume proliferation after drug treatment, resulting in disease recurrence/persistence. Using a single-cell assay to track individual cells that exit a drug-induced senescence-like state, we show that cells exhibit asynchronous exit from a senescent-like state, and display different rates of proliferation. Escaped cells retain sensitivity to drug treatment, but display inter-clonal variability. We also find heterogeneity in gene expression with some of the escaped clones retaining senescence-associated gene expression. Senescent leukemia cells exhibit changes in gene expression that affect metabolism and senescence-associated secretory phenotype (SASP)-related genes. Herein, we generate a senescence gene signature and show that this signature is a prognostic marker of worse overall survival in AML and multiple other cancers. A portion of senescent leukemia cells depend on lysosome activity; chloroquine, an inhibitor of lysosome activity, promotes senolysis of some senescent leukemia cells. Our study indicates that the serious risks associated with the use of erythropoietin-stimulating agents (ESAs) in anemic cancer patients may be attributed to their ability to promote drug-tolerant cancer cells through the senescence program.
Collapse
Affiliation(s)
- David Miller
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
| | - Kyra Kerkhofs
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | | | | | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Canada
| | - Josée Hébert
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital and Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Canada
- Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute for Artificial Intelligence, Toronto, Canada
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Computer Science, University of Toronto, Toronto, Canada
- Department of Biostatistics, Dalla Lana School of Public Health, Toronto, Canada
| | - Mark A Bayfield
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | - Samuel Benchimol
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
23
|
Panovska D, Nazari P, Cole B, Creemers PJ, Derweduwe M, Solie L, Van Gassen S, Claeys A, Verbeke T, Cohen EF, Tolstorukov MY, Saeys Y, Van der Planken D, Bosisio FM, Put E, Bamps S, Clement PM, Verfaillie M, Sciot R, Ligon KL, De Vleeschouwer S, Antoranz A, De Smet F. Single-cell molecular profiling using ex vivo functional readouts fuels precision oncology in glioblastoma. Cell Mol Life Sci 2023; 80:147. [PMID: 37171617 PMCID: PMC11071868 DOI: 10.1007/s00018-023-04772-1] [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: 12/01/2022] [Revised: 03/06/2023] [Accepted: 03/29/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Functional profiling of freshly isolated glioblastoma (GBM) cells is being evaluated as a next-generation method for precision oncology. While promising, its success largely depends on the method to evaluate treatment activity which requires sufficient resolution and specificity. METHODS Here, we describe the 'precision oncology by single-cell profiling using ex vivo readouts of functionality' (PROSPERO) assay to evaluate the intrinsic susceptibility of high-grade brain tumor cells to respond to therapy. Different from other assays, PROSPERO extends beyond life/death screening by rapidly evaluating acute molecular drug responses at single-cell resolution. RESULTS The PROSPERO assay was developed by correlating short-term single-cell molecular signatures using mass cytometry by time-of-flight (CyTOF) to long-term cytotoxicity readouts in representative patient-derived glioblastoma cell cultures (n = 14) that were exposed to radiotherapy and the small-molecule p53/MDM2 inhibitor AMG232. The predictive model was subsequently projected to evaluate drug activity in freshly resected GBM samples from patients (n = 34). Here, PROSPERO revealed an overall limited capacity of tumor cells to respond to therapy, as reflected by the inability to induce key molecular markers upon ex vivo treatment exposure, while retaining proliferative capacity, insights that were validated in patient-derived xenograft (PDX) models. This approach also allowed the investigation of cellular plasticity, which in PDCLs highlighted therapy-induced proneural-to-mesenchymal (PMT) transitions, while in patients' samples this was more heterogeneous. CONCLUSION PROSPERO provides a precise way to evaluate therapy efficacy by measuring molecular drug responses using specific biomarker changes in freshly resected brain tumor samples, in addition to providing key functional insights in cellular behavior, which may ultimately complement standard, clinical biomarker evaluations.
Collapse
Affiliation(s)
- Dena Panovska
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Pouya Nazari
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Basiel Cole
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Pieter-Jan Creemers
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Marleen Derweduwe
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Lien Solie
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
- Department of Neurosurgery, University Hospitals (UZ) Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Sofie Van Gassen
- Data Mining and Modeling for Biomedicine Group, VIB Inflammation Research Center, Ghent University, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Annelies Claeys
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Tatjana Verbeke
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Elizabeth F Cohen
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Y Tolstorukov
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine Group, VIB Inflammation Research Center, Ghent University, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | | | - Francesca M Bosisio
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Eric Put
- Neurosurgery Department, Faculty of Medicine and Life Sciences UHasselt, Hasselt, Belgium
| | - Sven Bamps
- Neurosurgery Department, Faculty of Medicine and Life Sciences UHasselt, Hasselt, Belgium
| | - Paul M Clement
- Department of Oncology, KU Leuven/UZ Leuven, Leuven, Belgium
| | - Michiel Verfaillie
- Europaziekenhuizen, Cliniques de l'Europe, Sint-Elisabeth, Brussels, Belgium
| | - Raf Sciot
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Keith L Ligon
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Steven De Vleeschouwer
- Department of Neurosurgery, University Hospitals (UZ) Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Asier Antoranz
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Frederik De Smet
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium.
- Leuven Institute for single-cell omics (LISCO), Leuven, Belgium.
| |
Collapse
|
24
|
Shen Q, Liu Y, Deng X, Hu CD. PRMT5 promotes chemotherapy-induced neuroendocrine differentiation in NSCLC. Thorac Cancer 2023. [PMID: 37140020 DOI: 10.1111/1759-7714.14921] [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: 03/13/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND In response to therapeutic treatments, cancer cells can exhibit a variety of resistance phenotypes including neuroendocrine differentiation (NED). NED is a process by which cancer cells can transdifferentiate into neuroendocrine-like cells in response to treatments, and is now widely accepted as a key mechanism of acquired therapy resistance. Recent clinical evidence has suggested that non-small cell lung cancer (NSCLC) can also transform into small cell lung cancer (SCLC) in patients treated with EGFR inhibitors. However, whether chemotherapy induces NED to confer therapy resistance in NSCLC remains unknown. METHODS We evaluated whether NSCLC cells can undergo NED in response to chemotherapeutic agents etoposide and cisplatin. By Knock-down of PRMT5 or pharmacological inhibition of PRMT5 to identify its role in the NED process. RESULTS We observed that both etoposide and cisplatin can induce NED in multiple NSCLC cell lines. Mechanistically, we identified protein arginine methyltransferase 5 (PRMT5) as a critical mediator of chemotherapy-induced NED. Significantly, the knock-down of PRMT5 or pharmacological inhibition of PRMT5 suppressed the induction of NED and increased the sensitivity to chemotherapy. CONCLUSION Taken together, our results suggest that targeting PRMT5 may be explored as a chemosensitization approach by inhibiting chemotherapy-induced NED.
Collapse
Affiliation(s)
- Qi Shen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, Indiana, USA
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, Indiana, USA
- College of Chemical Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Xuehong Deng
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, Indiana, USA
| | - Chang-Deng Hu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, Indiana, USA
| |
Collapse
|
25
|
Sun Y, Weng J, Chen X, Ma S, Zhang Y, Zhang F, Zhang Z, Wang F, Shao J, Zheng S. Oroxylin A activates ferritinophagy to induce hepatic stellate cell senescence against hepatic fibrosis by regulating cGAS-STING pathway. Biomed Pharmacother 2023; 162:114653. [PMID: 37086511 DOI: 10.1016/j.biopha.2023.114653] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/24/2023] Open
Abstract
In recent study, the pathological mechanism of liver fibrosis has been associated with hepatic stellate cell (HSC) senescence. Targeted induction of HSC senescence is considered as a new strategy to remove activated HSC. Nevertheless, little is known about the role of ferritinophagy in cell senescence. In this study, we reported that Oroxylin A from Scutellaria baicalensis Georgi can regulate HSC senescence induced by ferritinophagy through the cGAS-STING pathway to reduce liver fibrosis. We first found that Oroxylin A treatment alleviated the pathological changes of liver fibrosis, reduced collagen deposition, and significantly inhibited liver fibrosis. Interestingly, Oroxylin A treatment can activate HSC ferritinophagy and further induce HSC senescence. It is noteworthy that ferritinophagy is mediated by nuclear receptor coactivator 4 (NCOA4), an important selective mediator for ferritin degradation. NCOA4 siRNA causes Oroxylin A to reduce the degree of telomerase activity in HSCs and induce the expression of senescence markers, such as SA-β-Gal and related marker proteins. Importantly, the cGAS-STING pathway is crucial to the activation of HSC ferritinophagy by Oroxylin A. Specifically, Oroxylin A can promote the secretion of cytokines like IFN-β by the cGAS-STING pathway to regulate ferritinophagy. cGAS siRNA resulted in a dose-dependent decrease in the expression of NCOA4, a significant reduction in the expression level of autophagy-related phenotype, and a decrease in the content of ROS and iron ions in HSCs. In conclusion, we identified the new role of ferritinophagy and the GAS-STING pathway in Oroxylin A -mediated anti-hepatic fibrosis.
Collapse
Affiliation(s)
- Ying Sun
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingdan Weng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaolei Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuyao Ma
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
26
|
Alhasan B, Mikeladze M, Guzhova I, Margulis B. Autophagy, molecular chaperones, and unfolded protein response as promoters of tumor recurrence. Cancer Metastasis Rev 2023; 42:217-254. [PMID: 36723697 DOI: 10.1007/s10555-023-10085-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023]
Abstract
Tumor recurrence is a paradoxical function of a machinery, whereby a small proportion of the cancer cell population enters a resistant, dormant state, persists long-term in this condition, and then transitions to proliferation. The dormant phenotype is typical of cancer stem cells, tumor-initiating cells, disseminated tumor cells, and drug-tolerant persisters, which all demonstrate similar or even equivalent properties. Cancer cell dormancy and its conversion to repopulation are regulated by several protein signaling systems that inhibit or induce cell proliferation and provide optimal interrelations between cancer cells and their special niche; these systems act in close connection with tumor microenvironment and immune response mechanisms. During dormancy and reawakening periods, cell proteostasis machineries, autophagy, molecular chaperones, and the unfolded protein response are recruited to protect refractory tumor cells from a wide variety of stressors and therapeutic insults. Proteostasis mechanisms functionally or even physically interfere with the main regulators of tumor relapse, and the significance of these interactions and implications in the tumor recurrence phases are discussed in this review.
Collapse
Affiliation(s)
- Bashar Alhasan
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia.
| | - Marina Mikeladze
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Irina Guzhova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Boris Margulis
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| |
Collapse
|
27
|
Zhang X, Zhang H, Zhang J, Yang M, Zhu M, Yin Y, Fan X, Yu F. The paradoxical role of radiation-induced cGAS-STING signalling network in tumour immunity. Immunology 2023; 168:375-388. [PMID: 36217274 DOI: 10.1111/imm.13592] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 10/06/2022] [Indexed: 11/27/2022] Open
Abstract
The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is an essential component of the innate immune system and is central to the identification of abnormal DNA leakage caused by ionising radiation (IR) damage. Cell-intrinsic cGAS-STING initiation has been revealed to have tremendous potential for facilitating interferon synthesis and T-cell priming. Targeting the cGAS-STING axis has been proposed as a strategy to improve radiosensitivity or enhance immunosurveillance. However, due to the complex biology of the irradiated tumour microenvironment and the extensive involvement of the cGAS-STING pathway in various physiological and pathological processes, many defects in this strategy limit the therapeutic effect. Here, we outline the molecular mechanisms by which IR activates the cGAS-STING pathway and analyse the dichotomous roles of the cGAS-STING pathway in modulating cancer immunity after radiotherapy (RT). Then, based on the crosstalk between the cGAS-STING pathway and other signalling events induced by IR, such as necroptosis, autophagy and other cellular effects, we discuss the immunomodulatory actions of the broad cGAS-STING signalling network in RT and their potential therapeutic applications. Finally, recent advances in combination therapeutic strategies targeting cGAS-STING in RT are explored.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Han Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Jiajia Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Mengdie Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Mengqin Zhu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Yuzhen Yin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Xin Fan
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Liu Y, Pagacz J, Wolfgeher DJ, Bromerg KD, Gorman JV, Kron SJ. Senescent cancer cell vaccines induce cytotoxic T cell responses targeting primary tumors and disseminated tumor cells. J Immunother Cancer 2023; 11:e005862. [PMID: 36792123 PMCID: PMC9933761 DOI: 10.1136/jitc-2022-005862] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Immune tolerance contributes to resistance to conventional cancer therapies such as radiation. Radiotherapy induces immunogenic cell death, releasing a burst of tumor antigens, but this appears insufficient to stimulate an effective antitumor immune response. Radiation also increases infiltration of cytotoxic T lymphocytes (CTLs), but their effector function is short lived. Although CTL exhaustion may be at fault, combining immune checkpoint blockade with radiation is insufficient to restore CTL function in most patients. An alternative model is that antigen presentation is the limiting factor, suggesting a defect in dendritic cell (DC) function. METHODS Building on our prior work showing that cancer cells treated with radiation in the presence of the poly(ADP-ribose) polymerase-1 inhibitor veliparib undergo immunogenic senescence, we reexamined senescent cells (SnCs) as preventative or therapeutic cancer vaccines. SnCs formed in vitro were cocultured with splenocytes and evaluated by scRNA-seq to examine immunogenicity. Immature bone-marrow-derived DCs cocultured with SnCs were examined for maturation and activation by flow cytometry and T cell proliferation assays. Viable SnCs or SnC-activated DCs were injected subcutaneously, and vaccine effects were evaluated by analysis of immune response, prevention of tumor engraftment, regression of established tumors and/or potentiation of immunotherapy or radiotherapy. RESULTS Murine CT26 colon carcinoma or 4T1 mammary carcinoma cells treated with radiation and veliparib form SnCs that promote DC maturation and activation in vitro, leading to efficient, STING-dependent CTL priming. Injecting mice with SnCs induces antigen-specific CTLs and confers protection from tumor engraftment. Injecting immunogenic SnCs into tumor-bearing mice increases inflammation with activated CTLs, suppresses tumor growth, potentiates checkpoint blockade, enhances radiotherapy and blocks colonization by disseminated tumor cells. Addressing the concern that reinjecting tumor cells into patients may be impractical, DCs activated with SnCs in vitro were similarly effective to SnCs in suppressing established tumors and blocking metastases. CONCLUSIONS Therapeutic vaccines based on senescent tumor cells and/or SnC-activated DCs have the potential to improve genotoxic and immune therapies and limit recurrence or metastasis.
Collapse
Affiliation(s)
- Yue Liu
- Department of Molecular Genetics and Cell Biology and Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
| | - Joanna Pagacz
- Department of Molecular Genetics and Cell Biology and Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
| | - Donald J Wolfgeher
- Department of Molecular Genetics and Cell Biology and Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
| | | | - Jacob V Gorman
- Oncology Discovery, AbbVie, North Chicago, Illinois, USA
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology and Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
29
|
Ex Vivo Functional Assay for Evaluating Treatment Response in Tumor Tissue of Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15020478. [PMID: 36672427 PMCID: PMC9856585 DOI: 10.3390/cancers15020478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) displays a large heterogeneity in treatment response, and consequently in patient prognosis. Despite extensive efforts, no clinically validated model is available to predict tumor response. Here we describe a functional test for predicting tumor response to radiation and chemotherapy on the level of the individual patient. METHODS Resection material of 17 primary HNSCC patients was cultured ex vivo, irradiated or cisplatin-treated, after which the effect on tumor cell vitality was analyzed several days after treatment. RESULTS Ionizing radiation (IR) affected tumor cell growth and viability with a clear dose-response relationship, and marked heterogeneity between tumors was observed. After a single dose of 5Gy, proliferation in IR-sensitive tumors dropped below 30% of the untreated level, while IR-resistant tumors maintained at least 60% of proliferation. IR-sensitive tumors showed on average a twofold increase in apoptosis, as well as an increased number and size of DNA damage foci after treatment. No differences in the homologous recombination (HR) proficiency between IR-sensitive and -resistant tumors were detected. Cisplatin caused a decrease in proliferation, as well as induction of apoptosis, again with marked variation between the samples. CONCLUSIONS Our functional ex vivo assay discriminated between IR-sensitive and IR-resistant HNSCC tumors, and may also be suitable for predicting response to cisplatin. Its predictive value is currently under investigation in a prospective clinical study.
Collapse
|
30
|
Zamkova MA, Persiyantseva NA, Tatarskiy VV, Shtil AA. Therapy-Induced Tumor Cell Senescence: Mechanisms and Circumvention. BIOCHEMISTRY (MOSCOW) 2023; 88:86-104. [PMID: 37068872 DOI: 10.1134/s000629792301008x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Plasticity of tumor cells (multitude of molecular regulation pathways) allows them to evade cytocidal effects of chemo- and/or radiation therapy. Metabolic adaptation of the surviving cells is based on transcriptional reprogramming. Similarly to the process of natural cell aging, specific features of the survived tumor cells comprise the therapy-induced senescence phenotype. Tumor cells with this phenotype differ from the parental cells since they become less responsive to drugs and form aggressive progeny. Importance of the problem is explained by the general biological significance of transcriptional reprogramming as a mechanism of adaptation to stress, and by the emerging potential of its pharmacological targeting. In this review we analyze the mechanisms of regulation of the therapy-induced tumor cell senescence, as well as new drug combinations aimed to prevent this clinically unfavorable phenomenon.
Collapse
Affiliation(s)
- Maria A Zamkova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia.
- Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
| | - Nadezhda A Persiyantseva
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
- Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Alexander A Shtil
- Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
- Institute of Cyber Intelligence Systems, National Research Nuclear University MEPHI, Moscow, 115409, Russia
| |
Collapse
|
31
|
Papadaki S, Magklara A. Regulation of Metabolic Plasticity in Cancer Stem Cells and Implications in Cancer Therapy. Cancers (Basel) 2022; 14:5912. [PMID: 36497394 PMCID: PMC9741285 DOI: 10.3390/cancers14235912] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer stem cells (CSCs), a subpopulation of tumor cells with self-renewal capacity, have been associated with tumor initiation, progression, and therapy resistance. While the bulk of tumor cells mainly use glycolysis for energy production, CSCs have gained attention for their ability to switch between glycolysis and oxidative phosphorylation, depending on their energy needs and stimuli from their microenvironment. This metabolic plasticity is mediated by signaling pathways that are also implicated in the regulation of CSC properties, such as the Wnt/β-catenin, Notch, and Hippo networks. Two other stemness-associated processes, autophagy and hypoxia, seem to play a role in the metabolic switching of CSCs as well. Importantly, accumulating evidence has linked the metabolic plasticity of CSCs to their increased resistance to treatment. In this review, we summarize the metabolic signatures of CSCs and the pathways that regulate them; we especially highlight research data that demonstrate the metabolic adaptability of these cells and their role in stemness and therapy resistance. As the development of drug resistance is a major challenge for successful cancer treatment, the potential of specific elimination of CSCs through targeting their metabolism is of great interest and it is particularly examined.
Collapse
Affiliation(s)
- Styliani Papadaki
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Angeliki Magklara
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
- Biomedical Research Institute–Foundation for Research and Technology, 45110 Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina (URCI), 45110 Ioannina, Greece
| |
Collapse
|
32
|
Liu H, Zhao H, Sun Y. Tumor microenvironment and cellular senescence: Understanding therapeutic resistance and harnessing strategies. Semin Cancer Biol 2022; 86:769-781. [PMID: 34799201 DOI: 10.1016/j.semcancer.2021.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 01/27/2023]
Abstract
The tumor microenvironment (TME) is a major contributor to cancer malignancy including development of therapeutic resistance, a process mediated in part through intercellular crosstalk. Besides diverse soluble factors responsible for pro-survival pathway activation, immune evasion and extracellular matrix (ECM) remodeling further promote cancer resistance. Importantly, therapy-induced senescence (TIS) of cells in the TME is frequently observed in anticancer regimens, an off-target effect that can generate profound impacts on disease progression. By conferring the resistance and fueling the repopulation of remaining cancerous cells, TIS is responsible for tumor relapse and distant metastasis in posttreatment stage. This pathological trajectory can be substantially driven by the pro-inflammatory feature of senescent cells, termed as the senescence-associated secretory phenotype (SASP). Targeting strategies to selectively and efficiently remove senescent cells before they exert non-autonomous but largely deleterious effects, are emerging as an effective solution to prevent drug resistance acquired from a treatment-remodeled TME. In this review, we summarize the TME composition and key activities that affect tissue homeostasis and support treatment resistance. Promising opportunities that allow TME-manipulation and senescent cell-targeting (senotherapy) are discussed, with translational pipelines to overcome therapeutic barriers in clinical oncology projected.
Collapse
Affiliation(s)
- Hanxin Liu
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Huifang Zhao
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Yu Sun
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China; Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
33
|
Targeting epiregulin in the treatment-damaged tumor microenvironment restrains therapeutic resistance. Oncogene 2022; 41:4941-4959. [PMID: 36202915 DOI: 10.1038/s41388-022-02476-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023]
Abstract
The tumor microenvironment (TME) represents a milieu enabling cancer cells to develop malignant properties, while concerted interactions between cancer and stromal cells frequently shape an "activated/reprogramed" niche to accelerate pathological progression. Here we report that a soluble factor epiregulin (EREG) is produced by senescent stromal cells, which non-cell-autonomously develop the senescence-associated secretory phenotype (SASP) upon DNA damage. Genotoxicity triggers EREG expression by engaging NF-κB and C/EBP, a process supported by elevated chromatin accessibility and increased histone acetylation. Stromal EREG reprograms the expression profile of recipient neoplastic cells in a paracrine manner, causing upregulation of MARCHF4, a membrane-bound E3 ubiquitin ligase involved in malignant progression, specifically drug resistance. A combinational strategy that empowers EREG-specific targeting in treatment-damaged TME significantly promotes cancer therapeutic efficacy in preclinical trials, achieving response indices superior to those of solely targeting cancer cells. In clinical oncology, EREG is expressed in tumor stroma and handily measurable in circulating blood of cancer patients post-chemotherapy. This study establishes EREG as both a targetable SASP factor and a new noninvasive biomarker of treatment-damaged TME, thus disclosing its substantial value in translational medicine.
Collapse
|
34
|
STING mediates nuclear PD-L1 targeting-induced senescence in cancer cells. Cell Death Dis 2022; 13:791. [PMID: 36109513 PMCID: PMC9477807 DOI: 10.1038/s41419-022-05217-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 01/21/2023]
Abstract
Immune checkpoint molecule programmed death-ligand 1 (PD-L1) is overexpressed in cancer cells and imparts resistance to cancer therapy. Although membrane PD-L1 has been targeted for cancer immune therapy, nuclear PD-L1 was reported to confer cancer resistance. Therefore, it is important to regulate the nuclear PD-L1. The mechanisms underlying the therapeutic efficacy of PD-L1 targeting have not been well-established. Cellular senescence has been considered a pivotal mechanism to prevent cancer progression, and recently, PD-L1 inhibition was shown to be involved in cancer cell senescence. However, the relevance of PD-L1 targeting-induced senescence and the role of stimulator of interferon genes (STING) has not been reported. Therefore, we aimed to identify the role of PD-L1 in cancer progression and how it regulates cancer prevention. In this study, we found that PD-L1 depletion-induced senescence via strong induction of STING expression in mouse melanoma B16-F10 and colon cancer CT26 cells, and in human melanoma A375 and lung cancer A549 cells. Interestingly, nuclear PD-L1 silencing increased STING promoter activity, implying that PD-L1 negatively regulates STING expression via transcriptional modulation. Furthermore, we showed that PD-L1 binds to the STING promoter region, indicating that PD-L1 directly controls STING expression to promote cancer growth. In addition, when we combined PD-L1 silencing with the senescence-inducing chemotherapeutic agent doxorubicin, the effect of PD-L1-targeting was even more powerful. Overall, our findings can contribute to the understanding of the role of PD-L1 in cancer therapy by elucidating a novel mechanism for PD-L1 targeting in cancer cells.
Collapse
|
35
|
Wattanathamsan O, Pongrakhananon V. Emerging role of microtubule-associated proteins on cancer metastasis. Front Pharmacol 2022; 13:935493. [PMID: 36188577 PMCID: PMC9515585 DOI: 10.3389/fphar.2022.935493] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/29/2022] [Indexed: 12/29/2022] Open
Abstract
The major cause of death in cancer patients is strongly associated with metastasis. While much remains to be understood, microtubule-associated proteins (MAPs) have shed light on metastatic progression’s molecular mechanisms. In this review article, we focus on the role of MAPs in cancer aggressiveness, particularly cancer metastasis activity. Increasing evidence has shown that a growing number of MAP member proteins might be fundamental regulators involved in altering microtubule dynamics, contributing to cancer migration, invasion, and epithelial-to-mesenchymal transition. MAP types have been established according to their microtubule-binding site and function in microtubule-dependent activities. We highlight that altered MAP expression was commonly found in many cancer types and related to cancer progression based on available evidence. Furthermore, we discuss and integrate the relevance of MAPs and related molecular signaling pathways in cancer metastasis. Our review provides a comprehensive understanding of MAP function on microtubules. It elucidates how MAPs regulate cancer progression, preferentially in metastasis, providing substantial scientific information on MAPs as potential therapeutic targets and prognostic markers for cancer management.
Collapse
Affiliation(s)
- Onsurang Wattanathamsan
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Varisa Pongrakhananon
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- *Correspondence: Varisa Pongrakhananon,
| |
Collapse
|
36
|
Llop-Hernández À, Verdura S, Cuyàs E, Menendez JA. Nutritional Niches of Cancer Therapy-Induced Senescent Cells. Nutrients 2022; 14:nu14173636. [PMID: 36079891 PMCID: PMC9460569 DOI: 10.3390/nu14173636] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Therapy-induced senescence (TIS) is a state of stable proliferative arrest of both normal and neoplastic cells that is triggered by exposure to anticancer treatments. TIS cells acquire a senescence-associated secretory phenotype (SASP), which is pro-inflammatory and actively promotes tumor relapse and adverse side-effects in patients. Here, we hypothesized that TIS cells adapt their scavenging and catabolic ability to overcome the nutritional constraints in their microenvironmental niches. We used a panel of mechanistically-diverse TIS triggers (i.e., bleomycin, doxorubicin, alisertib, and palbociclib) and Biolog Phenotype MicroArrays to identify (among 190 different carbon and nitrogen sources) candidate metabolites that support the survival of TIS cells in limiting nutrient conditions. We provide evidence of distinguishable TIS-associated nutrient consumption profiles involving a core set of shared (e.g., glutamine) and unique (e.g., glucose-1-phosphate, inosine, and uridine) nutritional sources after diverse senescence-inducing interventions. We also observed a trend for an inverse correlation between the intensity of the pro-inflammatory SASP provoked by different TIS agents and diversity of compensatory nutritional niches utilizable by senescent cells. These findings support the detailed exploration of the nutritional niche as a new metabolic dimension to understand and target TIS in cancer.
Collapse
Affiliation(s)
| | - Sara Verdura
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 17005 Girona, Spain
| | - Elisabet Cuyàs
- Girona Biomedical Research Institute, 17190 Girona, Spain
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 17005 Girona, Spain
- Correspondence: (E.C.); or (J.A.M.)
| | - Javier A. Menendez
- Girona Biomedical Research Institute, 17190 Girona, Spain
- Metabolism and Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 17005 Girona, Spain
- Correspondence: (E.C.); or (J.A.M.)
| |
Collapse
|
37
|
Beykou M, Arias-Garcia M, Roumeliotis TI, Choudhary JS, Moser N, Georgiou P, Bakal C. Proteomic characterisation of triple negative breast cancer cells following CDK4/6 inhibition. Sci Data 2022; 9:395. [PMID: 35817775 PMCID: PMC9273754 DOI: 10.1038/s41597-022-01512-1] [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: 02/02/2022] [Accepted: 06/28/2022] [Indexed: 01/10/2023] Open
Abstract
When used in combination with hormone treatment, Palbociclib prolongs progression-free survival of patients with hormone receptor positive breast cancer. Mechanistically, Palbociclib inhibits CDK4/6 activity but the basis for differing sensitivity of cancer to Palbociclib is poorly understood. A common observation in a subset of Triple Negative Breast Cancers (TNBCs) is that prolonged CDK4/6 inhibition can engage a senescence-like state where cells exit the cell cycle, whilst, remaining metabolically active. To better understand the senescence-like cell state which arises after Palbociclib treatment we used mass spectrometry to quantify the proteome, phosphoproteome, and secretome of Palbociclib-treated MDA-MB-231 TNBC cells. We observed altered levels of cell cycle regulators, immune response, and key senescence markers upon Palbociclib treatment. These datasets provide a starting point for the derivation of biomarkers which could inform the future use CDK4/6 inhibitors in TNBC subtypes and guide the development of potential combination therapies.
Collapse
Affiliation(s)
- Melina Beykou
- Imperial College London, Department of Electrical and Electronic Engineering, Circuits and Systems Group, South Kensington Campus, London, SW7 2AZ, UK.
- Institute of Cancer Research, Division of Cancer Biology, Dynamical Cell Systems, London, SW3 6JB, UK.
- Cancer Research UK Convergence Science Centre, South Kensington Campus, London, SW7 2AZ, UK.
| | - Mar Arias-Garcia
- Institute of Cancer Research, Division of Cancer Biology, Dynamical Cell Systems, London, SW3 6JB, UK
| | - Theodoros I Roumeliotis
- Institute of Cancer Research, Division of Cancer Biology, Functional Proteomics, London, SW3 6JB, UK
| | - Jyoti S Choudhary
- Institute of Cancer Research, Division of Cancer Biology, Functional Proteomics, London, SW3 6JB, UK
| | - Nicolas Moser
- Imperial College London, Department of Electrical and Electronic Engineering, Circuits and Systems Group, South Kensington Campus, London, SW7 2AZ, UK.
- Cancer Research UK Convergence Science Centre, South Kensington Campus, London, SW7 2AZ, UK.
| | - Pantelis Georgiou
- Imperial College London, Department of Electrical and Electronic Engineering, Circuits and Systems Group, South Kensington Campus, London, SW7 2AZ, UK.
- Cancer Research UK Convergence Science Centre, South Kensington Campus, London, SW7 2AZ, UK.
| | - Chris Bakal
- Institute of Cancer Research, Division of Cancer Biology, Dynamical Cell Systems, London, SW3 6JB, UK.
- Cancer Research UK Convergence Science Centre, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
38
|
Effect of Autophagy Inhibitors on Radiosensitivity in DNA Repair-Proficient and -Deficient Glioma Cells. Medicina (B Aires) 2022; 58:medicina58070889. [PMID: 35888608 PMCID: PMC9317283 DOI: 10.3390/medicina58070889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/18/2023] Open
Abstract
Background and Objectives: The development of radioresistance is a fundamental barrier to successful glioblastoma therapy. Autophagy is thought to play a role in facilitating the DNA repair of DNA damage foci in radiation-exposed tumor cells, thus, potentially contributing to their restoration of proliferative capacity and development of resistance in vitro. However, the effect of autophagy inhibitors on DNA damage repair is not fully clear and requires further investigation. Materials and Methods: In this work, we utilized M059K (DNA-PKcs proficient) and M059J (DNA-PKcs deficient) glioma cell lines to investigate the role of autophagy inhibitors in the DNA repair of radiation-induced DNA damage. Cell viability following radiation was determined by trypan blue exclusion in both cell lines. Cell death and autophagy assays were performed to evaluate radiation-induced cell stress responses. DNA damage was measured as based on the intensity of phosphorylated γ-H2AX, a DNA double-stranded breaks (DSBs) marker, in the presence or absence of autophagy inhibitors. Results: The cell viability assay showed that M059J cells were more sensitive to the same dose of radiation (4 Gy) than M059K cells. This observation was accompanied by an elevation in γ-H2AX formation in M059J but not in M059K cells. In addition, the DAPI/TUNEL and Senescence-associated β-galactosidase (SA-β-gal) staining assays did not reveal significant differences in apoptosis and/or senescence induction in response to radiation, respectively, in either cell line. However, acridine orange staining demonstrated clear promotion of acidic vesicular organelles (AVOs) in both cell lines in response to 4 Gy radiation. Moreover, DNA damage marker levels were found to be elevated 72 h post-radiation when autophagy was inhibited by the lysosomotropic agent bafilomycin A1 (BafA1) or the PI3K inhibitor 3-methyl adenine (3-MA) in M059K cells. Conclusions: The extent of the DNA damage response remained high in the DNA-PKcs deficient cells following exposure to radiation, indicating their inability to repair the newly formed DNA-DSBs. On the other hand, radioresistant M059K cells showed more DNA damage response only when autophagy inhibitors were used with radiation, suggesting that the combination of autophagy inhibitors with radiation may interfere with DNA repair efficiency.
Collapse
|
39
|
"Derived Multiple Allogeneic Protein Paracrine Signaling (d-MAPPS)" Enhances T Cell-Driven Immune Response to Murine Mammary Carcinoma. Anal Cell Pathol (Amst) 2022; 2022:3655595. [PMID: 35757015 PMCID: PMC9217617 DOI: 10.1155/2022/3655595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/26/2022] [Indexed: 11/23/2022] Open
Abstract
Breast cancer is considered refractory to immunotherapy. Accordingly, there is an urgent need for the therapeutic use of new immunostimulatory agents which would enhance antitumor immune response against breast cancer cells. “Derived Multiple Allogeneic Protein Paracrine Signaling (d-MAPPS)” is a biological product whose activity is based on chemokines and cytokines that modulate homing and phenotype of immune cells. d-MAPPS contains high concentration of dendritic cell (DC) and T cell-attracting chemokine CXCL16 and potent T cell-activating cytokine IL-27 which enhance DC:T cell cross-talk in inflamed tissues. Herewith, we used 4T1 murine model of breast cancer to analyze d-MAPPS-dependent enhancement of T cell-driven antitumor immunity. 4T1+d-MAPPS-treated mice showed delayed mammary tumor appearance compared to 4T1+saline-treated animals. d-MAPPS significantly reduced tumor weight and volume and improved survival of 4T1-treated mice. Significantly increased concentration of CXCL16, IL-27, IFN-γ, and IL-17 and decreased concentration of immunosuppressive TGF-β and IL-10 were measured in serum samples and tumor tissues of 4T1+d-MAPPS-treated mice. d-MAPPS enhanced production of IL-12 and increased expression of MHC class II and costimulatory molecules on tumor-infiltrated DC, significantly improving their antigen-presenting properties. d-MAPPS in CXCL16-dependent manner promoted recruitment of antitumorigenic IFN-γ/IL-17-producing CD4+Th1/Th17 cells and in IL-27-dependent manner induced expansion of tumoricidal CD178+granzyme B-expressing CD8+CTLs and inhibited generation of tolerogenic DC, IL-10, and TGF-β-producing FoxP3-expressing T regulatory cells. In summing up, d-MAPPS, in CXL16- and IL-27-dependent manner, enhanced T cell-driven antitumor immune response and suppressed breast cancer growth in experimental mice.
Collapse
|
40
|
Wang Z, Gao J, Xu C. Tackling cellular senescence by targeting miRNAs. Biogerontology 2022; 23:387-400. [PMID: 35727469 DOI: 10.1007/s10522-022-09972-z] [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: 01/17/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022]
Abstract
Cellular senescence, which is characterized by permanent proliferation arrest, has become an important target for the amelioration of various human diseases. The activity of senescent cells is mainly related to the senescence-associated secretory phenotype (SASP). The SASP can cause chronic inflammation in local tissues and organs through autocrine and paracrine mechanisms, and a series of factors secreted by senescent cells can deteriorate the cellular microenvironment, promoting tumor formation and exacerbating aging-related diseases. Therefore, avoiding the promotion of cancer is an urgent problem. In recent years, increased attention has been given to the mechanistic study of microRNAs in senescence. As important posttranscriptional regulators, microRNAs possess unique tissue-specific expression in senescence. MicroRNAs can regulate the SASP by regulating proteins in the senescence signaling pathway, the reverse transcriptase activity of telomerase, the generation of reactive oxygen species and oxidative damage to mitochondria. Numerous studies have confirmed that removing senescent cells does not cause significant side effects, which also opens the door to the development of treatment modalities against senescent cells. Herein, this review discusses the double-edged sword of cellular senescence in tumors and aging-related diseases and emphasizes the roles of microRNAs in regulating the SASP, especially the potential of microRNAs to be used as therapeutic targets to inhibit senescence, giving rise to novel therapeutic approaches for the treatment of aging-associated diseases.
Collapse
Affiliation(s)
- Zehua Wang
- Obstetrics and Gynecology, Hospital of Fudan University, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Jianwen Gao
- School of Medical Engineering, Ma'anshan University, No. 8, Huangchi Road, Gushu Town, Dangtu County, Ma'anshan, 243100, Anhui, China. .,Major of Biotechnological Pharmaceutics, Shanghai Pharmaceutical School, Shanghai, 200135, China.
| | - Congjian Xu
- Obstetrics and Gynecology, Hospital of Fudan University, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China.,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, 200032, China
| |
Collapse
|
41
|
Homann L, Rentschler M, Brenner E, Böhm K, Röcken M, Wieder T. IFN-γ and TNF Induce Senescence and a Distinct Senescence-Associated Secretory Phenotype in Melanoma. Cells 2022; 11:cells11091514. [PMID: 35563820 PMCID: PMC9103004 DOI: 10.3390/cells11091514] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint blockade (ICB) therapy is a central pillar of melanoma treatment leading to durable response rates. Important mechanisms of action of ICB therapy include disinhibition of CD4+ and CD8+ T cells. Stimulated CD4+ T helper 1 cells secrete the effector cytokines interferon-gamma (IFN-γ) and tumor necrosis factor alpha (TNF), which induce senescence in tumor cells. Besides being growth-arrested, senescent cells are metabolically active and secrete a large spectrum of factors, which are summarized as senescence-associated secretory phenotype (SASP). This secretome affects the tumor growth. Here, we compared the SASP of cytokine-induced senescent (CIS) cells with the SASP of therapy-induced senescent (TIS) cells. Therefore, we established in vitro models for CIS and TIS in melanoma. The human melanoma cell lines SK-MEL-28 and WM115 were treated with the cytokines IFN-γ and TNF as CIS, the chemotherapeutic agent doxorubicin, and the cell cycle inhibitor palbociclib as TIS. Then, we determined several senescence markers, i.e., growth arrest, p21 expression, and senescence-associated β-galactosidase (SA-β-gal) activity. For SASP analyses, we measured the regulation and secretion of several common SASP factors using qPCR arrays, protein arrays, and ELISA. Each treatment initiated a stable growth arrest, enhanced SA-β-gal activity, and—except palbociclib—increased the expression of p21. mRNA and protein analyses revealed that gene expression and secretion of SASP factors were severalfold stronger in CIS than in TIS. Finally, we showed that treatment with the conditioned media (CM) derived from cytokine- and palbociclib-treated cells induced senescence characteristics in melanoma cells. Thus, we conclude that senescence induction via cytokines may lead to self-sustaining senescence surveillance of melanoma.
Collapse
Affiliation(s)
- Lorenzo Homann
- Department of Dermatology, University of Tuebingen, 72076 Tuebingen, Germany; (M.R.); (E.B.); (K.B.); (M.R.)
- Correspondence: (L.H.); (T.W.); Tel.: +49-7071-2986865 (L.H.); +49-7071-2978240 (T.W.)
| | - Maximilian Rentschler
- Department of Dermatology, University of Tuebingen, 72076 Tuebingen, Germany; (M.R.); (E.B.); (K.B.); (M.R.)
- Institute of Physiology I, Department of Vegetative and Clinical Physiology, University of Tuebingen, 72074 Tuebingen, Germany
| | - Ellen Brenner
- Department of Dermatology, University of Tuebingen, 72076 Tuebingen, Germany; (M.R.); (E.B.); (K.B.); (M.R.)
| | - Katharina Böhm
- Department of Dermatology, University of Tuebingen, 72076 Tuebingen, Germany; (M.R.); (E.B.); (K.B.); (M.R.)
| | - Martin Röcken
- Department of Dermatology, University of Tuebingen, 72076 Tuebingen, Germany; (M.R.); (E.B.); (K.B.); (M.R.)
| | - Thomas Wieder
- Institute of Physiology I, Department of Vegetative and Clinical Physiology, University of Tuebingen, 72074 Tuebingen, Germany
- Correspondence: (L.H.); (T.W.); Tel.: +49-7071-2986865 (L.H.); +49-7071-2978240 (T.W.)
| |
Collapse
|
42
|
Nutrition Interventions of Herbal Compounds on Cellular Senescence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1059257. [PMID: 35528514 PMCID: PMC9068308 DOI: 10.1155/2022/1059257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 11/01/2021] [Accepted: 04/02/2022] [Indexed: 01/10/2023]
Abstract
When cells undergo large-scale senescence, organ aging ensues, resulting in irreversible organ pathology and organismal aging. The study of senescence in cells provides an important avenue to understand the factors that influence aging and can be used as one of the useful tools for examining age-related human diseases. At present, many herbal compounds have shown effects on delaying cell senescence. This review summarizes the main characteristics and mechanisms of cell senescence, age-related diseases, and the recent progress on the natural products targeting cellular senescence, with the aim of providing insights to aid the clinical management of age-related diseases.
Collapse
|
43
|
Lee AH, Mejia Peña C, Dawson MR. Comparing the Secretomes of Chemorefractory and Chemoresistant Ovarian Cancer Cell Populations. Cancers (Basel) 2022; 14:1418. [PMID: 35326569 PMCID: PMC8946241 DOI: 10.3390/cancers14061418] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) constitutes the majority of all ovarian cancer cases and has staggering rates of both refractory and recurrent disease. While most patients respond to the initial treatment with paclitaxel and platinum-based drugs, up to 25% do not, and of the remaining that do, 75% experience disease recurrence within the subsequent two years. Intrinsic resistance in refractory cases is driven by environmental stressors like tumor hypoxia which alter the tumor microenvironment to promote cancer progression and resistance to anticancer drugs. Recurrent disease describes the acquisition of chemoresistance whereby cancer cells survive the initial exposure to chemotherapy and develop adaptations to enhance their chances of surviving subsequent treatments. Of the environmental stressors cancer cells endure, exposure to hypoxia has been identified as a potent trigger and priming agent for the development of chemoresistance. Both in the presence of the stress of hypoxia or the therapeutic stress of chemotherapy, cancer cells manage to cope and develop adaptations which prime populations to survive in future stress. One adaptation is the modification in the secretome. Chemoresistance is associated with translational reprogramming for increased protein synthesis, ribosome biogenesis, and vesicle trafficking. This leads to increased production of soluble proteins and extracellular vesicles (EVs) involved in autocrine and paracrine signaling processes. Numerous studies have demonstrated that these factors are largely altered between the secretomes of chemosensitive and chemoresistant patients. Such factors include cytokines, growth factors, EVs, and EV-encapsulated microRNAs (miRNAs), which serve to induce invasive molecular, biophysical, and chemoresistant phenotypes in neighboring normal and cancer cells. This review examines the modifications in the secretome of distinct chemoresistant ovarian cancer cell populations and specific secreted factors, which may serve as candidate biomarkers for aggressive and chemoresistant cancers.
Collapse
Affiliation(s)
- Amy H. Lee
- Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA;
| | - Carolina Mejia Peña
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA;
| | - Michelle R. Dawson
- Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA;
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA;
| |
Collapse
|
44
|
Lee SK, Shen Z, Han MS, Tung CH. Developing a far-red fluorogenic beta-galactosidase probe for senescent cell imaging and photoablation. RSC Adv 2022; 12:4543-4549. [PMID: 35425504 PMCID: PMC8981090 DOI: 10.1039/d2ra00377e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 01/31/2022] [Indexed: 01/10/2023] Open
Abstract
A methylene blue (MB)-based beta-galactosidase (β-gal) activatable molecule, Gal-MB, was developed for senescence imaging and light-triggered senolysis. When in contact with LacZ β-gal or senescence-associated β-gal (SA-β-gal), the photoinsensitive Gal-MB becomes fluorescent. Gal-MB also offered selective phototoxicity toward LacZ β-gal expressing cells and drug-induced senescent cells, which express SA-β-gal, after light illumination at 665 nm. A methylene blue (MB)-based beta-galactosidase (β-gal) activatable molecule, Gal-MB, was developed for senescence imaging and light-triggered senolysis.![]()
Collapse
Affiliation(s)
- Seung Koo Lee
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine New York NY 10021 USA
| | - Zhenhua Shen
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine New York NY 10021 USA
| | - Myung Shin Han
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine New York NY 10021 USA
| | - Ching-Hsuan Tung
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine New York NY 10021 USA
| |
Collapse
|
45
|
Mechanisms of Hydroxyurea-Induced Cellular Senescence: An Oxidative Stress Connection? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7753857. [PMID: 34707779 PMCID: PMC8545575 DOI: 10.1155/2021/7753857] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/09/2021] [Accepted: 09/25/2021] [Indexed: 01/10/2023]
Abstract
Hydroxyurea (HU) is a water-soluble antiproliferative agent used for decades in neoplastic and nonneoplastic conditions. HU is considered an essential medicine because of its cytoreduction functions. HU is an antimetabolite that inhibits ribonucleotide reductase, which causes a depletion of the deoxyribonucleotide pool and dramatically reduces cell proliferation. The proliferation arrest, depending on drug concentration and exposure, may promote a cellular senescence phenotype associated with cancer cell therapy resistance and inflammation, influencing neighboring cell functions, immunosuppression, and potential cancer relapse. HU can induce cellular senescence in both healthy and transformed cells in vitro, in part, because of increased reactive oxygen species (ROS). Here, we analyze the main molecular mechanisms involved in cytotoxic/genotoxic HU function, the potential to increase intracellular ROS levels, and the principal features of cellular senescence induction. Understanding the mechanisms involved in HU's ability to induce cellular senescence may help to improve current chemotherapy strategies and control undesirable treatment effects in cancer patients and other diseases.
Collapse
|
46
|
Targeting cellular senescence in cancer by plant secondary metabolites: A systematic review. Pharmacol Res 2021; 177:105961. [PMID: 34718135 DOI: 10.1016/j.phrs.2021.105961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/23/2021] [Accepted: 10/23/2021] [Indexed: 12/13/2022]
Abstract
Senescence suppresses tumor growth, while also developing a tumorigenic state in the nearby cells that is mediated by senescence-associated secretory phenotypes (SASPs). The dual function of cellular senescence stresses the need for identifying multi-targeted agents directed towards the promotion of cell senescence in cancer cells and suppression of the secretion of pro-tumorigenic signaling mediators in neighboring cells. Natural secondary metabolites have shown favorable anticancer responses in recent decades, as some have been found to target the senescence-associated mediators and pathways. Furthermore, phenolic compounds and polyphenols, terpenes and terpenoids, alkaloids, and sulfur-containing compounds have shown to be promising anticancer agents through the regulation of paracrine and autocrine pathways. Plant secondary metabolites are potential regulators of SASPs factors that suppress tumor growth through paracrine mediators, including growth factors, cytokines, extracellular matrix components/enzymes, and proteases. On the other hand, ataxia-telangiectasia mutated, ataxia-telangiectasia and Rad3-related, extracellular signal-regulated kinase/mitogen-activated protein kinase, phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin, nuclear factor-κB, Janus kinase/signal transducer and activator of transcription, and receptor tyrosine kinase-associated mediators are main targets of candidate phytochemicals in the autocrine senescence pathway. Such a regulatory role of phytochemicals on senescence-associated pathways are associated with cell cycle arrest and the attenuation of apoptotic/inflammatory/oxidative stress pathways. The current systematic review highlights the critical roles of natural secondary metabolites in the attenuation of autocrine and paracrine cellular senescence pathways, while also elucidating the chemopreventive and chemotherapeutic capabilities of these compounds. Additionally, we discuss current challenges, limitations, and future research indications.
Collapse
|
47
|
Liu J, Erenpreisa J, Sikora E. Polyploid giant cancer cells: An emerging new field of cancer biology. Semin Cancer Biol 2021; 81:1-4. [PMID: 34695579 DOI: 10.1016/j.semcancer.2021.10.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jinsong Liu
- Department of Anatomical Pathology and Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | - Ewa Sikora
- Laboratory of Moleuclar Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|