1
|
Ordónez-Rubiano EG, Cómbita A, Baldoncini M, Payán-Gómez C, Gómez-Amarillo DF, Hakim F, Camargo J, Zorro-Sepúlveda V, Luzzi S, Zorro O, Parra-Medina R. Cellular Senescence in Diffuse Gliomas: From Physiopathology to Possible Treatments. World Neurosurg 2024; 191:138-148. [PMID: 39233309 DOI: 10.1016/j.wneu.2024.08.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
Cellular senescence in gliomas is a complex process that is induced by aging and replication, ionizing radiation, oncogenic stress, and the use of temozolomide. However, the escape routes that gliomas must evade senescence and achieve cellular immortality are much more complex, in which the expression of telomerase and the alternative lengthening of telomeres, as well as the mutation of some proto-oncogenes or tumor suppressor genes, are involved. In gliomas, these molecular mechanisms related to cellular senescence can have a tumor-suppressing or promoting effect and are directly involved in tumor recurrence and progression. From these cellular mechanisms related to cellular senescence, it is possible to generate targeted senostatic and senolytic therapies that improve the response to currently available treatments and improve survival rates. This review aims to summarize the mechanisms of induction and evasion of cellular senescence in gliomas, as well as review possible treatments with therapies targeting pathways related to cellular senescence.
Collapse
Affiliation(s)
- Edgar G Ordónez-Rubiano
- Department of Neurological Surgery, Fundación Universitaria de Ciencias de la Salud (FUCS), Hospital de San José - Sociedad de Cirugía de Bogotá, Bogotá, Colombia; School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia; Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia.
| | - Alba Cómbita
- Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia; Department of Microbiology, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Matías Baldoncini
- School of Medicine, Laboratory of Microsurgical Neuroanatomy, Second Chair of Gross Anatomy, University of Buenos Aires, Buenos Aires, Argentina; Department of Neurological Surgery, Hospital San Fernando, Buenos Aires, Argentina
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz, Colombia
| | - Diego F Gómez-Amarillo
- Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Fernando Hakim
- Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Julián Camargo
- Department of Neurosurgery, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | | | - Sabino Luzzi
- Neurosurgery Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Oscar Zorro
- Department of Neurological Surgery, Fundación Universitaria de Ciencias de la Salud (FUCS), Hospital de San José - Sociedad de Cirugía de Bogotá, Bogotá, Colombia
| | - Rafael Parra-Medina
- Department of Pathology, Instituto Nacional de Cancerología, Bogotá, Colombia; Research Institute, Fundación Universitaria de Ciencias de la Salud (FUCS), Hospital de San José - Sociedad de Cirugía de Bogotá, Bogotá, Colombia
| |
Collapse
|
2
|
Xiong J, Dong L, Lv Q, Yin Y, Zhao J, Ke Y, Wang S, Zhang W, Wu M. Targeting senescence-associated secretory phenotypes to remodel the tumour microenvironment and modulate tumour outcomes. Clin Transl Med 2024; 14:e1772. [PMID: 39270064 PMCID: PMC11398298 DOI: 10.1002/ctm2.1772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/17/2024] [Accepted: 07/08/2024] [Indexed: 09/15/2024] Open
Abstract
Tumour cell senescence can be induced by various factors, including DNA damage, inflammatory signals, genetic toxins, ionising radiation and nutrient metabolism. The senescence-associated secretory phenotype (SASP), secreted by senescent tumour cells, possesses the capacity to modulate various immune cells, including macrophages, T cells, natural killer cells and myeloid-derived suppressor cells, as well as vascular endothelial cells and fibroblasts within the tumour microenvironment (TME), and this modulation can result in either the promotion or suppression of tumorigenesis and progression. Exploring the impact of SASP on the TME could identify potential therapeutic targets, yet limited studies have dissected its functions. In this review, we delve into the causes and mechanisms of tumour cell senescence. We then concentrate on the influence of SASP on the tumour immune microenvironment, angiogenesis, extracellular matrix and the reprogramming of cancer stem cells, along with their associated tumour outcomes. Last, we present a comprehensive overview of the diverse array of senotherapeutics, highlighting their prospective advantages and challenge for the treatment of cancer patients. KEY POINTS: Senescence-associated secretory phenotype (SASP) secretion from senescent tumour cells significantly impacts cancer progression and biology. SASP is involved in regulating the remodelling of the tumour microenvironment, including immune microenvironment, vascular, extracellular matrix and cancer stem cells. Senotherapeutics, such as senolytic, senomorphic, nanotherapy and senolytic vaccines, hold promise for enhancing cancer treatment efficacy.
Collapse
Affiliation(s)
- Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lu Dong
- The Second Clinical College of Wuhan University, Wuhan, China
| | - Qiongying Lv
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yutong Yin
- The First Clinical College of Wuhan University, Wuhan, China
| | - Jiahui Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Youning Ke
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Saleh T. Therapy-induced senescence is finally escapable, what is next? Cell Cycle 2024; 23:713-721. [PMID: 38879812 PMCID: PMC11229739 DOI: 10.1080/15384101.2024.2364579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024] Open
Abstract
Several breakthrough articles have recently confirmed the ability of tumor cells to escape the stable cell cycle arrest imposed by Therapy-Induced Senescence (TIS). Subsequently, accepting the hypothesis that TIS is escapable should encourage serious reassessments of the fundamental roles of senescence in cancer treatment. The potential for escape from TIS undermines the well-established tumor suppressor function of senescence, proposes it as a mechanism of tumor dormancy leading to disease recurrence and invites for further investigation of its unfavorable contribution to cancer therapy outcomes. Moreover, escaping TIS strongly indicates that the elimination of senescent tumor cells, primarily through pharmacological means, is a suitable approach for increasing the efficacy of cancer treatment, one that still requires further exploration. This commentary provides an overview of the recent evidence that unequivocally demonstrated the ability of therapy-induced senescent tumor cells in overcoming the terminal growth arrest fate and provides future perspectives on the roles of TIS in tumor biology.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
4
|
Song KX, Wang JX, Huang D. Therapy-induced senescent tumor cells in cancer relapse. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:273-278. [PMID: 39036667 PMCID: PMC11256611 DOI: 10.1016/j.jncc.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 07/23/2024] Open
Abstract
Cellular senescence is characterized by a generally irreversible cell cycle arrest and the secretion of bioactive factors known as the senescence-associated secretory phenotype (SASP). In an oncogenic context, senescence is considered a tumor suppressive mechanism as it prevents cell proliferation and inhibits the progression from pre-malignant to malignant disease. However, recent studies have demonstrated that senescent tumor cells, which could spontaneously exist within cancer tissues or arise in response to various cancer interventions (the so-called therapy-induced senescence, TIS), can acquire pro-tumorigenic properties and are capable of driving local and metastatic relapse. This highlights the complex and multifaceted nature of cellular senescence in cancer biology. Here, we summarize the current knowledge of the pathological function of therapy-induced senescent tumor cells and discuss possible mechanisms by which tumor cell senescence contributes to cancer relapse. We also discuss implications for future studies toward targeting these less appreciated cells.
Collapse
Affiliation(s)
- Ke-Xin Song
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jun-Xian Wang
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - De Huang
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
5
|
Bui I, Baritaki S, Libra M, Zaravinos A, Bonavida B. Cancer Resistance Is Mediated by the Upregulation of Several Anti-Apoptotic Gene Products via the Inducible Nitric Oxide Synthase/Nitric Oxide Pathway: Therapeutic Implications. Antioxid Redox Signal 2023; 39:853-889. [PMID: 37466477 DOI: 10.1089/ars.2023.0250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Significance: Several therapeutic strategies for cancer treatments have been developed with time, and significant milestones have been achieved recently. However, with these novel therapies, not all cancer types respond and in the responding cancer types only a subset is affected. The failure to respond is principally the result that these cancers develop several mechanisms of resistance. Thus, a focus of current research investigations is to unravel the various mechanisms that regulate resistance and identify suitable targets for new therapeutics. Recent Advances: Hence, many human cancer types have been reported to overexpress the inducible nitric oxide synthase (iNOS) and it has been suggested that iNOS/nitric oxide (NO) plays a pivotal role in the regulation of resistance. We have postulated that iNOS overexpression or NO regulates the overexpression of pivotal anti-apoptotic gene products such as B-cell lymphoma 2 (Bcl-2), B-cell lymphoma extra large (Bcl-xL), myeloid cell leukemia-1 (Mcl-1), and survivin. In this report, we describe the various mechanisms, transcriptional, post-transcriptional, and post-translational, by which iNOS/NO regulates the expression of the above anti-apoptotic gene products. Critical Issues: The iNOS/NO-mediated regulation of the four gene products is not the same with both specific and overlapping pathways. Our findings are, in large part, validated by bioinformatic analyses demonstrating, in several cancers, several direct correlations between the expression of iNOS and each of the four examined anti-apoptotic gene products. Future Directions: We have proposed that targeting iNOS may be highly efficient since it will result in the underexpression of multiple anti-apoptotic proteins and shifting the balance toward the proapoptotic gene products and reversal of resistance. Antioxid. Redox Signal. 39, 853-889.
Collapse
Affiliation(s)
- Indy Bui
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
| | - Stavroula Baritaki
- Laboratory of Experimental Oncology, Department of Surgery, School of Medicine, University of Crete, Heraklion, Greece
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Italian League Against Cancer, Catania, Italy
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
6
|
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: 4.0] [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
|
7
|
Le NT. Metabolic regulation of endothelial senescence. Front Cardiovasc Med 2023; 10:1232681. [PMID: 37649668 PMCID: PMC10464912 DOI: 10.3389/fcvm.2023.1232681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/18/2023] [Indexed: 09/01/2023] Open
Abstract
Endothelial cell (EC) senescence is increasingly recognized as a significant contributor to the development of vascular dysfunction and age-related disorders and diseases, including cancer and cardiovascular diseases (CVD). The regulation of cellular senescence is known to be influenced by cellular metabolism. While extensive research has been conducted on the metabolic regulation of senescence in other cells such as cancer cells and fibroblasts, our understanding of the metabolic regulation of EC senescence remains limited. The specific metabolic changes that drive EC senescence are yet to be fully elucidated. The objective of this review is to provide an overview of the intricate interplay between cellular metabolism and senescence, with a particular emphasis on recent advancements in understanding the metabolic changes preceding cellular senescence. I will summarize the current knowledge on the metabolic regulation of EC senescence, aiming to offer insights into the underlying mechanisms and future research directions.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
8
|
Chojak R, Fares J, Petrosyan E, Lesniak MS. Cellular senescence in glioma. J Neurooncol 2023; 164:11-29. [PMID: 37458855 DOI: 10.1007/s11060-023-04387-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/01/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Glioma is the most common primary brain tumor and is often associated with treatment resistance and poor prognosis. Standard treatment typically involves radiotherapy and temozolomide-based chemotherapy, both of which induce cellular senescence-a tumor suppression mechanism. DISCUSSION Gliomas employ various mechanisms to bypass or escape senescence and remain in a proliferative state. Importantly, senescent cells remain viable and secrete a large number of factors collectively known as the senescence-associated secretory phenotype (SASP) that, paradoxically, also have pro-tumorigenic effects. Furthermore, senescent cells may represent one form of tumor dormancy and play a role in glioma recurrence and progression. CONCLUSION In this article, we delineate an overview of senescence in the context of gliomas, including the mechanisms that lead to senescence induction, bypass, and escape. Furthermore, we examine the role of senescent cells in the tumor microenvironment and their role in tumor progression and recurrence. Additionally, we highlight potential therapeutic opportunities for targeting senescence in glioma.
Collapse
Affiliation(s)
- Rafał Chojak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jawad Fares
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Edgar Petrosyan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA.
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
9
|
Evangelou K, Belogiannis K, Papaspyropoulos A, Petty R, Gorgoulis VG. Escape from senescence: molecular basis and therapeutic ramifications. J Pathol 2023; 260:649-665. [PMID: 37550877 DOI: 10.1002/path.6164] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 08/09/2023]
Abstract
Cellular senescence constitutes a stress response mechanism in reaction to a plethora of stimuli. Senescent cells exhibit cell-cycle arrest and altered function. While cell-cycle withdrawal has been perceived as permanent, recent evidence in cancer research introduced the so-called escape-from-senescence concept. In particular, under certain conditions, senescent cells may resume proliferation, acquiring highly aggressive features. As such, they have been associated with tumour relapse, rendering senescence less effective in inhibiting cancer progression. Thus, conventional cancer treatments, incapable of eliminating senescence, may benefit if revisited to include senolytic agents. To this end, it is anticipated that the assessment of the senescence burden in everyday clinical material by pathologists will play a crucial role in the near future, laying the foundation for more personalised approaches. Here, we provide an overview of the investigations that introduced the escape-from-senescence phenomenon, the identified mechanisms, as well as the major implications for pathology and therapy. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Belogiannis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelos Papaspyropoulos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Russell Petty
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
10
|
DeLuca VJ, Saleh T. Insights into the role of senescence in tumor dormancy: mechanisms and applications. Cancer Metastasis Rev 2023; 42:19-35. [PMID: 36681750 DOI: 10.1007/s10555-023-10082-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/12/2023] [Indexed: 01/23/2023]
Abstract
One of the most formidable challenges in oncology and tumor biology research is to provide an accurate understanding of tumor dormancy mechanisms. Dormancy refers to the ability of tumor cells to go undetected in the body for a prolonged period, followed by "spontaneous" escape. Various models of dormancy have been postulated, including angiogenic, immune-mediated, and cellular dormancy. While the former two propose mechanisms by which tumor growth may remain static at a population level, cellular dormancy refers to molecular processes that restrict proliferation at the cell level. Senescence is a form of growth arrest, during which cells undergo distinct phenotypic, epigenetic, and metabolic changes. Senescence is also associated with the development of a robust secretome, comprised of various chemokines and cytokines that interact with the surrounding microenvironment, including other tumor cells, stromal cells, endothelial cells, and immune cells. Both tumor and non-tumor cells can undergo senescence following various stressors, many of which are present during tumorigenesis and therapy. As such, senescent cells are present within forming tumors and in residual tumors post-treatment and therefore play a major role in tumor biology. However, the contributions of senescence to dormancy are largely understudied. Here, we provide an overview of multiple processes that have been well established as being involved in tumor dormancy, and we speculate on how senescence may contribute to these mechanisms.
Collapse
Affiliation(s)
- Valerie J DeLuca
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan.
| |
Collapse
|
11
|
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
|
12
|
The Molecular and Cellular Strategies of Glioblastoma and Non-Small-Cell Lung Cancer Cells Conferring Radioresistance. Int J Mol Sci 2022; 23:ijms232113577. [PMID: 36362359 PMCID: PMC9656305 DOI: 10.3390/ijms232113577] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Ionizing radiation (IR) has been shown to play a crucial role in the treatment of glioblastoma (GBM; grade IV) and non-small-cell lung cancer (NSCLC). Nevertheless, recent studies have indicated that radiotherapy can offer only palliation owing to the radioresistance of GBM and NSCLC. Therefore, delineating the major radioresistance mechanisms may provide novel therapeutic approaches to sensitize these diseases to IR and improve patient outcomes. This review provides insights into the molecular and cellular mechanisms underlying GBM and NSCLC radioresistance, where it sheds light on the role played by cancer stem cells (CSCs), as well as discusses comprehensively how the cellular dormancy/non-proliferating state and polyploidy impact on their survival and relapse post-IR exposure.
Collapse
|
13
|
Tumor cell dormancy: Molecular mechanisms, and pharmacological approaches to target dormant cells for countering tumor. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
14
|
Zanotti S, Decaesteker B, Vanhauwaert S, De Wilde B, De Vos WH, Speleman F. Cellular senescence in neuroblastoma. Br J Cancer 2022; 126:1529-1538. [PMID: 35197583 PMCID: PMC9130206 DOI: 10.1038/s41416-022-01755-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/14/2022] [Accepted: 02/10/2022] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is a tumour that arises from the sympathoadrenal lineage occurring predominantly in children younger than five years. About half of the patients are diagnosed with high-risk tumours and undergo intensive multi-modal therapy. The success rate of current treatments for high-risk neuroblastoma is disappointingly low and survivors suffer from multiple therapy-related long-term side effects. Most chemotherapeutics drive cancer cells towards cell death or senescence. Senescence has long been considered to represent a terminal non-proliferative state and therefore an effective barrier against tumorigenesis. This dogma, however, has been challenged by recent observations that infer a much more dynamic and reversible nature for this process, which may have implications for the efficacy of therapy-induced senescence-oriented treatment strategies. Neuroblastoma cells in a dormant, senescent-like state may escape therapy, whilst their senescence-associated secretome may promote inflammation and invasiveness, potentially fostering relapse. Conversely, due to its distinct molecular identity, senescence may also represent an opportunity for the development of novel (combination) therapies. However, the limited knowledge on the molecular dynamics and diversity of senescence signatures demands appropriate models to study this process in detail. This review summarises the molecular knowledge about cellular senescence in neuroblastoma and investigates current and future options towards therapeutic exploration.
Collapse
Affiliation(s)
- Sofia Zanotti
- grid.5284.b0000 0001 0790 3681Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Antwerp, 2610 Belgium ,grid.5342.00000 0001 2069 7798Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.510942.bCancer Research Institute Ghent (CRIG), Ghent, 9000 Belgium
| | - Bieke Decaesteker
- grid.5342.00000 0001 2069 7798Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.510942.bCancer Research Institute Ghent (CRIG), Ghent, 9000 Belgium
| | - Suzanne Vanhauwaert
- grid.5342.00000 0001 2069 7798Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.510942.bCancer Research Institute Ghent (CRIG), Ghent, 9000 Belgium
| | - Bram De Wilde
- grid.5342.00000 0001 2069 7798Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.5342.00000 0001 2069 7798Department of Internal Medicine and Pediatrics, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.410566.00000 0004 0626 3303Department of Pediatric Hematology Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, 9000 Belgium
| | - Winnok H. De Vos
- grid.5284.b0000 0001 0790 3681Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Antwerp, 2610 Belgium
| | - Frank Speleman
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000, Belgium. .,Cancer Research Institute Ghent (CRIG), Ghent, 9000, Belgium.
| |
Collapse
|
15
|
Saini G, Joshi S, Garlapati C, Li H, Kong J, Krishnamurthy J, Reid MD, Aneja R. Polyploid giant cancer cell characterization: New frontiers in predicting response to chemotherapy in breast cancer. Semin Cancer Biol 2022; 81:220-231. [PMID: 33766651 PMCID: PMC8672208 DOI: 10.1016/j.semcancer.2021.03.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023]
Abstract
Although polyploid cells were first described nearly two centuries ago, their ability to proliferate has only recently been demonstrated. It also becomes increasingly evident that a subset of tumor cells, polyploid giant cancer cells (PGCCs), play a critical role in the pathophysiology of breast cancer (BC), among other cancer types. In BC, PGCCs can arise in response to therapy-induced stress. Their progeny possess cancer stem cell (CSC) properties and can repopulate the tumor. By modulating the tumor microenvironment (TME), PGCCs promote BC progression, chemoresistance, metastasis, and relapse and ultimately impact the survival of BC patients. Given their pro- tumorigenic roles, PGCCs have been proposed to possess the ability to predict treatment response and patient prognosis in BC. Traditionally, DNA cytometry has been used to detect PGCCs.. The field will further derive benefit from the development of approaches to accurately detect PGCCs and their progeny using robust PGCC biomarkers. In this review, we present the current state of knowledge about the clinical relevance of PGCCs in BC. We also propose to use an artificial intelligence-assisted image analysis pipeline to identify PGCC and map their interactions with other TME components, thereby facilitating the clinical implementation of PGCCs as biomarkers to predict treatment response and survival outcomes in BC patients. Finally, we summarize efforts to therapeutically target PGCCs to prevent chemoresistance and improve clinical outcomes in patients with BC.
Collapse
Affiliation(s)
- Geetanjali Saini
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Shriya Joshi
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | | | - Hongxiao Li
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA; Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA; Department of Computer Science, Georgia State University, Atlanta, GA, USA; Department of Computer Science, Emory University, Atlanta, GA, USA
| | | | - Michelle D Reid
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
16
|
Sartorius K, Antwi SO, Chuturgoon A, Roberts LR, Kramvis A. RNA Therapeutic Options to Manage Aberrant Signaling Pathways in Hepatocellular Carcinoma: Dream or Reality? Front Oncol 2022; 12:891812. [PMID: 35600358 PMCID: PMC9115561 DOI: 10.3389/fonc.2022.891812] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/04/2022] [Indexed: 11/24/2022] Open
Abstract
Despite the early promise of RNA therapeutics as a magic bullet to modulate aberrant signaling in cancer, this field remains a work-in-progress. Nevertheless, RNA therapeutics is now a reality for the treatment of viral diseases (COVID-19) and offers great promise for cancer. This review paper specifically investigates RNAi as a therapeutic option for HCC and discusses a range of RNAi technology including anti-sense oligonucleotides (ASOs), Aptamers, small interfering RNA (siRNA), ribozymes, riboswitches and CRISPR/Cas9 technology. The use of these RNAi based interventions is specifically outlined in three primary strategies, namely, repressing angiogenesis, the suppression of cell proliferation and the promotion of apoptosis. We also discuss some of the inherent chemical and delivery problems, as well as targeting issues and immunogenic reaction to RNAi interventions.
Collapse
Affiliation(s)
- Kurt Sartorius
- Hepatitis Virus Diversity Research Unit, School of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa.,The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL, United States.,Department of Surgery, KZN Kwazulu-Natal (UKZN) Gastrointestinal Cancer Research Centre, Durban, South Africa
| | - Samuel O Antwi
- The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL, United States.,Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, United States
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, South Africa
| | - Lewis R Roberts
- The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL, United States.,Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Anna Kramvis
- Hepatitis Virus Diversity Research Unit, School of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
17
|
Stoczynska-Fidelus E, Węgierska M, Kierasińska A, Ciunowicz D, Rieske P. Role of Senescence in Tumorigenesis and Anticancer Therapy. JOURNAL OF ONCOLOGY 2022; 2022:5969536. [PMID: 35342397 PMCID: PMC8956409 DOI: 10.1155/2022/5969536] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/18/2022] [Accepted: 02/05/2022] [Indexed: 12/20/2022]
Abstract
Although the role of senescence in many physiological and pathological processes is becoming more identifiable, many aspects of senescence are still enigmatic. A special attention is paid to the role of this phenomenon in tumor development and therapy. This review mainly deals with a large spectrum of oncological issues, beginning with therapy-induced senescence and ending with oncogene-induced senescence. Moreover, the role of senescence in experimental approaches, such as primary cancer cell culture or reprogramming into stem cells, is also beginning to receive further consideration. Additional focus is made on senescence resulting from mitotic catastrophe processes triggered by events occurring during mitosis and jeopardizing chromosomal stability. It has to be also realized that based on recent findings, the basics of senescent cell property interpretation, such as irreversibility of proliferation blockade, can be undermined. It shows that the definition of senescence probably requires updating. Finally, the role of senescence is lately more understandable in the immune system, especially since senescence can diminish the effectiveness of the chimeric antigen receptor T-cell (CAR-T) therapy. In this review, we summarize the current knowledge regarding all these issues.
Collapse
Affiliation(s)
- Ewelina Stoczynska-Fidelus
- Department of Molecular Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| | - Marta Węgierska
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| | - Amelia Kierasińska
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| | - Damian Ciunowicz
- Department of Molecular Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| |
Collapse
|
18
|
Bai Z, Zhou Y, Ye X, Li Y, Peng Y, Guan Q, Zhang W, Ma L. Survivin suppression heightens BZML-induced mitotic catastrophe to overcome multidrug resistance by removing therapy-induced senescent A549/Taxol cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119174. [PMID: 34808206 DOI: 10.1016/j.bbamcr.2021.119174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
Mitotic catastrophe (MC) is a newly identified type of anticancer mechanism for multidrug resistance (MDR) prevention. However, the long cellular death process resulting from MC is not beneficial for anticancer treatment. BZML is a novel colchicine-binding site inhibitor which can overcome MDR by inducing MC; however, BZML-induced MC cells underwent a long cellular death process. Thus, to improve anticancer therapies based on drug-induced MC, BZML-induced MC was served as a model to further study the underlying molecular mechanisms in the process of MC. Here, BZML could induce p53-dependent senescence in A549/Taxol cells, a MDR cell line. This senescence was a secondary effect of MC in overcoming MDR. During MC, BZML-induced destruction of protein-degradation system contributed not only to an increase of p53 protein but also to the accumulation of survivin in nucleus of A549/Taxol cells. Importantly, the nuclear accumulation of survivin was not the inducer but the result of BZML-induced MC, and it promoted the survival of senescent cells. Moreover, it provided additional vulnerability and critical opportunities for sequentially applied therapies. Further, targeting survivin with YM155 accelerated the death of MC cells by timely eliminating therapy-induced senescent cells and strengthening the efficiency of BZML in overcoming MDR in A549/Taxol cells. Collectively, nuclear accumulation of survivin delayed cellular death during MC by promoting the survival of BZML-induced senescent A549/Taxol cells. Moreover, "one-two punch" approach to cancer treatment based on combination therapy with YM155 for survivin suppression might be a new strategy for potentiating MC to overcome MDR.
Collapse
Affiliation(s)
- Zhaoshi Bai
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China.
| | - Yiran Zhou
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Xinyue Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yupeng Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yaling Peng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
19
|
Olszewska A, Borkowska A, Granica M, Karolczak J, Zglinicki B, Kieda C, Was H. Escape From Cisplatin-Induced Senescence of Hypoxic Lung Cancer Cells Can Be Overcome by Hydroxychloroquine. Front Oncol 2022; 11:738385. [PMID: 35127467 PMCID: PMC8813758 DOI: 10.3389/fonc.2021.738385] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/23/2021] [Indexed: 12/23/2022] Open
Abstract
Chemotherapy is the commonly used treatment for advanced lung cancer. However, it produces side effects such as the development of chemoresistance. A possible responsible mechanism may be therapy-induced senescence (TIS). TIS cells display increased senescence-associated β-galactosidase (SA-β-gal) activity and irreversible growth arrest. However, recent data suggest that TIS cells can reactivate their proliferative potential and lead to cancer recurrence. Our previous study indicated that reactivation of proliferation by TIS cells might be related with autophagy modulation. However, exact relationship between both processes required further studies. Therefore, the aim of our study was to investigate the role of autophagy in the senescence-related chemoresistance of lung cancer cells. For this purpose, human and murine lung cancer cells were treated with two commonly used chemotherapeutics: cisplatin (CIS), which forms DNA adducts or docetaxel (DOC), a microtubule poison. Hypoxia, often overlooked in experimental settings, has been implicated as a mechanism responsible for a significant change in the response to treatment. Thus, cells were cultured under normoxic (~19% O2) or hypoxic (1% O2) conditions. Herein, we show that hypoxia increases resistance to CIS. Lung cancer cells cultured under hypoxic conditions escaped from CIS-induced senescence, displayed reduced SA-β-gal activity and a decreased percentage of cells in the G2/M phase of the cell cycle. In turn, hypoxia increased the proliferation of lung cancer cells and the proportion of cells proceeding to the G0/G1 phase. Further molecular analyses demonstrated that hypoxia inhibited the prosenescent p53/p21 signaling pathway and induced epithelial to mesenchymal transition in CIS-treated cancer cells. In cells treated with DOC, such effects were not observed. Of importance, pharmacological autophagy inhibitor, hydroxychloroquine (HCQ) was capable of overcoming short-term CIS-induced resistance of lung cancer cells in hypoxic conditions. Altogether, our data demonstrated that hypoxia favors cancer cell escape from CIS-induced senescence, what could be overcome by inhibition of autophagy with HCQ. Therefore, we propose that HCQ might be used to interfere with the ability of senescent cancer cells to repopulate following exposure to DNA-damaging agents. This effect, however, needs to be tested in a long-term perspective for preclinical and clinical applications.
Collapse
Affiliation(s)
- Aleksandra Olszewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Agata Borkowska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Monika Granica
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- Doctoral School of Translational Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Justyna Karolczak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Bartosz Zglinicki
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Halina Was
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
- *Correspondence: Halina Was,
| |
Collapse
|
20
|
Cell death mechanisms in head and neck cancer cells in response to low and high-LET radiation. Expert Rev Mol Med 2022. [DOI: 10.1017/erm.2021.31] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AbstractHead and neck squamous cell carcinoma (HNSCC) is a common malignancy that develops in or around the throat, larynx, nose, sinuses and mouth, and is mostly treated with a combination of chemo- and radiotherapy (RT). The main goal of RT is to kill enough of the cancer cell population, whilst preserving the surrounding normal and healthy tissue. The mechanisms by which conventional photon RT achieves this have been extensively studied over several decades, but little is known about the cell death pathways that are activated in response to RT of increasing linear energy transfer (LET), including proton beam therapy and heavy ions. Here, we provide an up-to-date review on the observed radiobiological effects of low- versus high-LET RT in HNSCC cell models, particularly in the context of specific cell death mechanisms, including apoptosis, necrosis, autophagy, senescence and mitotic death. We also detail some of the current therapeutic strategies targeting cell death pathways that have been investigated to enhance the radiosensitivity of HNSCC cells in response to RT, including those that may present with clinical opportunities for eventual patient benefit.
Collapse
|
21
|
Liao Z, Yeo HL, Wong SW, Zhao Y. Cellular Senescence: Mechanisms and Therapeutic Potential. Biomedicines 2021; 9:1769. [PMID: 34944585 PMCID: PMC8698401 DOI: 10.3390/biomedicines9121769] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Cellular senescence is a complex and multistep biological process which cells can undergo in response to different stresses. Referring to a highly stable cell cycle arrest, cellular senescence can influence a multitude of biological processes-both physiologically and pathologically. While phenotypically diverse, characteristics of senescence include the expression of the senescence-associated secretory phenotype, cell cycle arrest factors, senescence-associated β-galactosidase, morphogenesis, and chromatin remodelling. Persistent senescence is associated with pathologies such as aging, while transient senescence is associated with beneficial programmes, such as limb patterning. With these implications, senescence-based translational studies, namely senotherapy and pro-senescence therapy, are well underway to find the cure to complicated diseases such as cancer and atherosclerosis. Being a subject of major interest only in the recent decades, much remains to be studied, such as regarding the identification of unique biomarkers of senescent cells. This review attempts to provide a comprehensive understanding of the diverse literature on senescence, and discuss the knowledge we have on senescence thus far.
Collapse
Affiliation(s)
- Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden
| | - Han Lin Yeo
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| | - Siaw Wen Wong
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore;
| | - Yan Zhao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| |
Collapse
|
22
|
Carpenter V, Saleh T, Min Lee S, Murray G, Reed J, Souers A, Faber AC, Harada H, Gewirtz DA. Androgen-deprivation induced senescence in prostate cancer cells is permissive for the development of castration-resistance but susceptible to senolytic therapy. Biochem Pharmacol 2021; 193:114765. [PMID: 34536356 DOI: 10.1016/j.bcp.2021.114765] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/30/2021] [Accepted: 09/13/2021] [Indexed: 01/26/2023]
Abstract
Prostate cancer (PCa) is one of the leading causes of cancer-related deaths in men. Although androgen deprivation therapies (ADT) and antiandrogens confer increased survival rates, most patients eventually develop castration resistant disease (CRPC). Previous studies have shown that these treatments have limited cytotoxicity, and instead, promote tumor cell growth arrest. We show here that PCa cells grown in either charcoal-stripped serum or exposed to the antiandrogen, bicalutamide, undergo a senescent growth arrest marked by morphological changes, upregulated senescence-associated-β-galactosidase (SA-β-Gal), cathepsin D accumulation, and expression of the senescence-associated secretory phenotype (SASP). The senescent growth arrest is, however, transient, as cells can resume proliferation upon restoration of normo-androgenic conditions. Intriguingly, enrichment for senescent cells confirmed that ADT-induced senescent cells recover their proliferative capacity, even under prolonged androgen deprivation, and form androgen-independent outgrowths. Transplantation of the enriched senescent population into castrated, syngeneic mice confirmed that senescent cells escape the growth arrest and form castration-resistant tumors in vivo. Outgrowth from senescence was associated with increased expression of constitutively active androgen receptor splice variants, a common mechanism of resistance to ADT. Finally, the selective elimination of senescent PCa cells following ADT in vitro by the senolytic navitoclax (ABT-263) interfered with the development of androgen-independent outgrowth. Taken together, these data support the premise that ADT-induced senescence is a transient cell state from which CRPC populations can emerge, identifying senescence as a potential driver of disease progression. Furthermore, it is feasible that senolytic therapy to eliminate senescent PCa cells could delay disease recurrence and/or progression to androgen independence.
Collapse
Affiliation(s)
- Valerie Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - So Min Lee
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Graeme Murray
- Department of Physics, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason Reed
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Physics, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew Souers
- AbbVie, 1 North Waukegan Road, North Chicago, IL, USA
| | - Anthony C Faber
- Philips Institute for Oral Health Research, School of Dentistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Hisashi Harada
- Philips Institute for Oral Health Research, School of Dentistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
23
|
Almasabi S, Ahmed AU, Boyd R, Williams BRG. A Potential Role for Integrin-Linked Kinase in Colorectal Cancer Growth and Progression via Regulating Senescence and Immunity. Front Genet 2021; 12:638558. [PMID: 34163519 PMCID: PMC8216764 DOI: 10.3389/fgene.2021.638558] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/08/2021] [Indexed: 01/10/2023] Open
Abstract
Integrin-linked kinase (ILK) has been implicated as a molecular driver and mediator in both inflammation and tumorigenesis of the colon. ILK functions as an adaptor and mediator protein linking the extracellular matrix with downstream signaling pathways. ILK is broadly expressed in many human tissues and cells. It is also overexpressed in many cancers, including colorectal cancer (CRC). Inflammation, as evidenced by inflammatory bowel disease (IBD), is one of the highest risk factors for initiating CRC. This has led to the hypothesis that targeting ILK therapeutically could have potential in CRC, as it regulates different cellular processes associated with CRC development and progression as well as inflammation in the colon. A number of studies have indicated an ILK function in senescence, a cellular process that arrests the cell cycle while maintaining active metabolism and transcription. Senescent cells produce different secretions collectively known as the senescence-associated secretory phenotype (SASP). The SASP secretions influence infiltration of different immune cells, either positively for clearing senescent cells or negatively for promoting tumor growth, reflecting the dual role of senescence in cancer. However, a role for ILK in senescence and immunity in CRC remains to be determined. In this review, we discuss the possible role for ILK in senescence and immunity, paying particular attention to the relevance of ILK in CRC. We also examine how activating Toll-like receptors (TLRs) and their agonists in CRC could trigger immune responses against cancer, as a combination therapy with ILK inhibition.
Collapse
Affiliation(s)
- Saleh Almasabi
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Cartherics, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Clinical Laboratory Sciences, Applied Medical Sciences, Najran University, Najran, Saudi Arabia.,Department of Molecular and Translational Sciences, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Afsar U Ahmed
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Richard Boyd
- Cartherics, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Bryan R G Williams
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
24
|
Fitsiou E, Soto-Gamez A, Demaria M. Biological functions of therapy-induced senescence in cancer. Semin Cancer Biol 2021; 81:5-13. [PMID: 33775830 DOI: 10.1016/j.semcancer.2021.03.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/03/2021] [Accepted: 03/22/2021] [Indexed: 01/10/2023]
Abstract
Therapy-induced cellular senescence is a state of stable growth arrest induced by common cancer treatments such as chemotherapy and radiation. In an oncogenic context, therapy-induced senescence can have different consequences. By blocking cellular proliferation and by facilitating immune cell infiltration, it functions as tumor suppressive mechanism. By fueling the proliferation of bystander cells and facilitating metastasis, it acts as a tumor promoting factor. This dual role is mainly attributed to the differential expression and secretion of a set of pro-inflammatory cytokines and tissue remodeling factors, collectively known as the Senescence-Associated Secretory Phenotype (SASP). Here, we describe cell-autonomous and non-cell-autonomous mechanisms that senescent cells activate in response to chemotherapy and radiation leading to tumor suppression and tumor promotion. We present the current state of knowledge on the stimuli that affect the activation of these opposing mechanisms and the effect of senescent cells on their micro-environment eg. by regulating the functions of immune cells in tumor clearance as well as strategies to eliminate senescent tumor cells before exerting their deleterious side-effects.
Collapse
Affiliation(s)
- Eleni Fitsiou
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, 9713AV, Groningen, The Netherlands
| | - Abel Soto-Gamez
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, 9713AV, Groningen, The Netherlands; University of Groningen, Groningen Research Institute of Pharmacy, Chemical and Pharmaceutical Biology, Groningen, The Netherlands
| | - Marco Demaria
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, 9713AV, Groningen, The Netherlands.
| |
Collapse
|
25
|
Carpenter VJ, Patel BB, Autorino R, Smith SC, Gewirtz DA, Saleh T. Senescence and castration resistance in prostate cancer: A review of experimental evidence and clinical implications. Biochim Biophys Acta Rev Cancer 2020; 1874:188424. [PMID: 32956765 DOI: 10.1016/j.bbcan.2020.188424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/10/2023]
Abstract
The development of Castration-Resistant Prostate Cancer (CRPC) remains a major challenge in the treatment of this disease. While Androgen Deprivation Therapy (ADT) can result in tumor shrinkage, a primary response of Prostate Cancer (PCa) cells to ADT is a senescent growth arrest. As a response to cancer therapies, senescence has often been considered as a beneficial outcome due to its association with stable growth abrogation, as well as the potential for immune system activation via the Senescence-Associated Secretory Phenotype (SASP). However, there is increasing evidence that not only can senescent cells regain proliferative capacity, but that senescence contributes to deleterious effects of cancer chemotherapy, including disease recurrence. Notably, the preponderance of work investigating the consequences of therapy-induced senescence on tumor progression has been performed in non-PCa models. Here, we summarize the evidence that ADT promotes a senescent response in PCa and postulate mechanisms by which senescence may contribute to the development of castration-resistance. Primarily, we suggest that ADT-induced senescence may support CRPC development via escape from senescence, by cell autonomous-reprogramming, and by the formation of a pro-tumorigenic SASP. However, due to the scarcity of direct evidence from PCa models, the consequences of ADT-induced senescence outlined here remain speculative until the relationship between senescence and CRPC can be experimentally defined.
Collapse
Affiliation(s)
- Valerie J Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Bhaumik B Patel
- Department of Internal Medicine, Division of Hematology, Oncology & Palliative Care, VCU Health, Richmond, VA, USA
| | - Riccardo Autorino
- Department of Surgery, Division of Urology, VCU Health, Richmond, VA, USA
| | | | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tareq Saleh
- The Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan.
| |
Collapse
|
26
|
Murray D, Mirzayans R. Cellular Responses to Platinum-Based Anticancer Drugs and UVC: Role of p53 and Implications for Cancer Therapy. Int J Mol Sci 2020; 21:ijms21165766. [PMID: 32796711 PMCID: PMC7461110 DOI: 10.3390/ijms21165766] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022] Open
Abstract
Chemotherapy is intended to induce cancer cell death through apoptosis and other avenues. Unfortunately, as discussed in this article, moderate doses of genotoxic drugs such as cisplatin typical of those achieved in the clinic often invoke a cytostatic/dormancy rather than cytotoxic/apoptosis response in solid tumour-derived cell lines. This is commonly manifested by an extended apoptotic threshold, with extensive apoptosis only being seen after very high/supralethal doses of such agents. The dormancy response can be associated with senescence-like features, polyploidy and/or multinucleation, depending in part on the p53 status of the cells. In most solid tumour-derived cells, dormancy represents a long-term survival mechanism, ultimately contributing to disease recurrence. This review highlights the nonlinearity of key aspects of the molecular and cellular responses to bulky DNA lesions in human cells treated with chemotherapeutic drugs (e.g., cisplatin) or ultraviolet light-C (a widely used tool for unraveling details of the DNA damage-response) as a function of the level of genotoxic stress. Such data highlight the growing realization that targeting dormant cancer cells, which frequently emerge following conventional anticancer treatments, may represent a novel strategy to prevent or, at least, significantly suppress cancer recurrence.
Collapse
|
27
|
Saleh T, Bloukh S, Carpenter VJ, Alwohoush E, Bakeer J, Darwish S, Azab B, Gewirtz DA. Therapy-Induced Senescence: An "Old" Friend Becomes the Enemy. Cancers (Basel) 2020; 12:cancers12040822. [PMID: 32235364 PMCID: PMC7226427 DOI: 10.3390/cancers12040822] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 01/10/2023] Open
Abstract
For the past two decades, cellular senescence has been recognized as a central component of the tumor cell response to chemotherapy and radiation. Traditionally, this form of senescence, termed Therapy-Induced Senescence (TIS), was linked to extensive nuclear damage precipitated by classical genotoxic chemotherapy. However, a number of other forms of therapy have also been shown to induce senescence in tumor cells independently of direct genomic damage. This review attempts to provide a comprehensive summary of both conventional and targeted anticancer therapeutics that have been shown to induce senescence in vitro and in vivo. Still, the utility of promoting senescence as a therapeutic endpoint remains under debate. Since senescence represents a durable form of growth arrest, it might be argued that senescence is a desirable outcome of cancer therapy. However, accumulating evidence suggesting that cells have the capacity to escape from TIS would support an alternative conclusion, that senescence provides an avenue whereby tumor cells can evade the potentially lethal action of anticancer drugs, allowing the cells to enter a temporary state of dormancy that eventually facilitates disease recurrence, often in a more aggressive state. Furthermore, TIS is now strongly connected to tumor cell remodeling, potentially to tumor dormancy, acquiring more ominous malignant phenotypes and accounts for several untoward adverse effects of cancer therapy. Here, we argue that senescence represents a barrier to effective anticancer treatment, and discuss the emerging efforts to identify and exploit agents with senolytic properties as a strategy for elimination of the persistent residual surviving tumor cell population, with the goal of mitigating the tumor-promoting influence of the senescent cells and to thereby reduce the likelihood of cancer relapse.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (T.S.); (S.D.)
| | - Sarah Bloukh
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Valerie J. Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Enas Alwohoush
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Jomana Bakeer
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Sarah Darwish
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (T.S.); (S.D.)
| | - Belal Azab
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Correspondence:
| |
Collapse
|
28
|
C 2-Ceramide-Induced Rb-Dominant Senescence-Like Phenotype Leads to Human Breast Cancer MCF-7 Escape from p53-Dependent Cell Death. Int J Mol Sci 2019; 20:ijms20174292. [PMID: 31480728 PMCID: PMC6747432 DOI: 10.3390/ijms20174292] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/12/2019] [Accepted: 08/22/2019] [Indexed: 12/22/2022] Open
Abstract
Ceramide is a sphingolipid which regulates a variety of signaling pathways in eukaryotic cells. Exogenous ceramide has been shown to induce cellular apoptosis. In this study, we observed that exogenous ceramide induced two distinct morphologies of cell fate following C2-ceramide treatment between the two breast cancer cell lines MCF-7 (wild type p53) and MDA-MB-231 (mutant p53) cells. The growth assessment showed that C2-ceramide caused significant growth inhibition and apoptosis in MDA-MB-231 cells through down-regulating the expression of mutant p53 whereas up-regulating the expression of pro-apoptotic Bad, and the proteolytic activation of caspase-3. However, senescence-associated (SA)-β-galactosidase (β-gal) was regulated in MCF-7 cells after C2-ceramide treatment. The results of proliferation and apoptosis assays showed that MCF-7 cells were more resistant to C2-ceramide treatment compared to MDA-MB-231 cells. Furthermore, C2-ceramide treatment induced a time-responsive increase in Rb protein, a key regulator of senescence accompanied with the upregulation of both mRNA level and protein level of SA-genes PAI-1 and TGaseII in MCF-7 but not in MDA-MB-231 cells, suggesting that some cancer cells escape apoptosis through modulating senescence-like phenotype. The results of our present study depicted the mechanism of C2-ceramide-resistant breast cancer cells, which might benefit the strategic development of ceramide-based chemotherapeutics against cancer in the future.
Collapse
|
29
|
Yu X, Zhang Y, Wu B, Kurie JM, Pertsemlidis A. The miR-195 Axis Regulates Chemoresistance through TUBB and Lung Cancer Progression through BIRC5. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:288-298. [PMID: 31508486 PMCID: PMC6727248 DOI: 10.1016/j.omto.2019.07.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
Chemoresistance and metastasis are the major reasons for non-small cell lung cancer (NSCLC) treatment failure and patient deaths. We and others have shown that miR-195 regulates the sensitivity of NSCLC to microtubule-targeting agents (MTAs) in vitro and in vivo and that miR-195 represses the migration and invasion of NSCLC cells in vitro. However, the relationship between miR-195 and microtubule structure and function and whether miR-195 represses NSCLC metastasis in vivo remain unknown. We assessed the correlation between tumor levels of TUBB and patient survival, the effect of TUBB on drug response, and the effect of miR-195 on migration, invasion, and metastasis in vitro and in vivo. We found that miR-195 directly targets TUBB; knockdown of TUBB sensitizes cells to MTAs, while overexpression confers resistance; high expression of TUBB is correlated with worse survival of lung adenocarcinoma; TUBB is also regulated by CHEK1, which has been shown to regulate chemoresistance; and miR-195 targets BIRC5 to repress migration and invasion in vitro and metastasis in vivo. Our findings highlight the relevance of the miR-195/TUBB axis in regulating the response of NSCLC to MTAs and the importance of the miR-195/BIRC5 axis in regulating NSCLC metastasis.
Collapse
Affiliation(s)
- Xiaojie Yu
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA.,Department of Cell Systems and Anatomy, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Yiqiang Zhang
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Binggen Wu
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA.,Xiangya School of Medicine, Central South University, Changsha, Hunan 410000, China
| | - Jonathan M Kurie
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Alexander Pertsemlidis
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA.,Department of Cell Systems and Anatomy, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA.,Department of Pediatrics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| |
Collapse
|
30
|
Luo Z, Chen W, Wu W, Luo W, Zhu T, Guo G, Zhang L, Wang C, Li M, Shi S. Metformin promotes survivin degradation through AMPK/PKA/GSK-3β-axis in non-small cell lung cancer. J Cell Biochem 2019; 120:11890-11899. [PMID: 30793366 DOI: 10.1002/jcb.28470] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/28/2018] [Accepted: 01/09/2019] [Indexed: 01/24/2023]
Abstract
Metformin, a first-line antidiabetic drug, has been reported with anticancer activities in many types of cancer. However, its molecular mechanisms remain largely unknown. As a member of inhibitor of apoptosis proteins, survivin plays an important role in the regulation of cell death. In the present study, we investigated the role of survivin in metformin-induced anticancer activity in non-small cell lung cancer in vitro. Metformin mainly induced apoptotic cell death in A549 and H460 cell lines. It remarkably suppressed the expression of survivin, decreased the stability of this protein, then promoted its proteasomal degradation. Moreover, metformin greatly suppressed protein kinase A (PKA) activity and induced its downstream glycogen synthase kinase 3β (GSK-3β) activation. PKA activators, both 8-Br-cAMP and forskolin, significantly increased the expression of survivin. Consistently both GSK-3β inhibitor LiCl and siRNA restored the expression of survivin in lung cancer cells. Furthermore, metformin induced adenosine 5'-monophosphate-activated protein kinase (AMPK) activation. Suppression of the activity of AMPK with Compound C reversed the degradation of survivin induced by metformin, and meanwhile, restored the activity of PKA and GSK-3β. These results suggest that metformin kills lung cancer cells through AMPK/PKA/GSK-3β-axis-mediated survivin degradation, providing novel insights into the anticancer effects of metformin.
Collapse
Affiliation(s)
- Zhuang Luo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Wei Chen
- Department of Pathophysiology in School of Basic Medical Science, North Sichuan Medical College, Nanchong, P.R. China
| | - Wenjuan Wu
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Wei Luo
- Department of Pulmonary and Critical Care Medicine, The People's Hospital of Leshan, Leshan, P.R. China
| | - Tingting Zhu
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Gang Guo
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Liyan Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Chu Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Min Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Shaoqing Shi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| |
Collapse
|
31
|
Franco MS, Roque MC, Oliveira MC. Short and Long-Term Effects of the Exposure of Breast Cancer Cell Lines to Different Ratios of Free or Co-Encapsulated Liposomal Paclitaxel and Doxorubicin. Pharmaceutics 2019; 11:pharmaceutics11040178. [PMID: 30979090 PMCID: PMC6523953 DOI: 10.3390/pharmaceutics11040178] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 03/29/2019] [Accepted: 03/31/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Associating paclitaxel (PTX) to doxorubicin (DXR) is one of the main chemotherapy strategies for breast cancer (BC) management. Protocols currently available consist in administering both drugs on their maximum tolerated dose, not taking into account the possible differences in efficacy due to their combination ratio. In the present study, the short and long-term cytotoxic effects as well as migratory effects of PTX, DXR, and its combinations at 10:1; 1:1 and 1:10 PTX:DXR molar ratios either free or co-encapsulated in liposomes were evaluated against three human BC cell lines (MDA-MB-231, MCF-7, and SKBR-3). Method: The MTT assay was used to screen for synergy or antagonism between PTX and DXR and the combination index value was calculated using the CalcuSyn software. Nuclear morphological alterations were evaluated by staining the cells with Hoescht 33342. The investigation of senescence and clonogenicity of BC cell lines exposed to different treatments was also studied. In addition, the ability of these cells to migrate was assessed. Results: Taken together, the results presented herein allow us to suggest that there is no benefit in enhancing the PTX concentration above that of DXR in the combination for any of the three cell lines tested. Conclusion: The developed liposomes co-encapsulating PTX and DXR in different molar ratios retained the biological properties of the mixture of free drugs and are valuable for planning new therapeutic strategies.
Collapse
Affiliation(s)
- Marina Santiago Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Marjorie Coimbra Roque
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Mônica Cristina Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
32
|
Recasens A, Munoz L. Targeting Cancer Cell Dormancy. Trends Pharmacol Sci 2019; 40:128-141. [PMID: 30612715 DOI: 10.1016/j.tips.2018.12.004] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/22/2018] [Accepted: 12/06/2018] [Indexed: 12/22/2022]
Abstract
Cancer cell dormancy is a process whereby cells enter reversible cell cycle arrest, termed quiescence. Quiescence is essential for cancer cells to acquire additional mutations, to survive in a new environment and initiate metastasis, to become resistant to cancer therapy, and to evade immune destruction. Thus, dormant cancer cells are considered to be responsible for cancer progression. As we start to understand the mechanisms that enable quiescence, we can begin to develop pharmacological strategies to target dormant cancer cells. Herein, we summarize the major molecular mechanisms underlying the dormancy of disseminated tumor cells and drug-tolerant persister cells. We then analyze the current pharmacological strategies aimed (i) to keep cancer cells in the harmless dormant state, (ii) to reactivate dormant cells to increase their susceptibility to anti-proliferative drugs, and (iii) to eradicate dormant cancer cells.
Collapse
Affiliation(s)
- Ariadna Recasens
- Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
| | - Lenka Munoz
- Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, NSW 2006, Australia.
| |
Collapse
|
33
|
Gayle SS, Sahni JM, Webb BM, Weber-Bonk KL, Shively MS, Spina R, Bar EE, Summers MK, Keri RA. Targeting BCL-xL improves the efficacy of bromodomain and extra-terminal protein inhibitors in triple-negative breast cancer by eliciting the death of senescent cells. J Biol Chem 2018; 294:875-886. [PMID: 30482844 DOI: 10.1074/jbc.ra118.004712] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/24/2018] [Indexed: 12/13/2022] Open
Abstract
Inhibitors of bromodomain and extra-terminal proteins (BETi) suppress oncogenic gene expression and have been shown to be efficacious in many in vitro and murine models of cancer, including triple-negative breast cancer (TNBC), a highly aggressive disease. However, in most cancer models, responses to BETi can be highly variable. We previously reported that TNBC cells either undergo senescence or apoptosis in response to BETi, but the specific mechanisms dictating these two cell fates remain unknown. Using six human TNBC cell lines, we show that the terminal response of TNBC cells to BETi is dictated by the intrinsic expression levels of the anti-apoptotic protein B-cell lymphoma-extra large (BCL-xL). BCL-xL levels were higher in cell lines that senesce in response to BETi compared with lines that primarily die in response to these drugs. Moreover, BCL-xL expression was further reduced in cells that undergo BETi-mediated apoptosis. Forced BCL-xL overexpression in cells that normally undergo apoptosis following BETi treatment shifted them to senescence without affecting the reported mechanism of action of BETi in TNBC, that is, mitotic catastrophe. Most importantly, pharmacological or genetic inhibition of BCL-xL induced apoptosis in response to BETi, and inhibiting BCL-xL, even after BETi-induced senescence had already occurred, still induced cell death. These results indicate that BCL-xL provides a senescent cell death-inducing or senolytic target that may be exploited to improve therapeutic outcomes of TNBC in response to BETi. They also suggest that the basal levels of BCL-xL should be predictive of tumor responses to BETi in current clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mathew K Summers
- Department of Radiation Oncology and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210
| | - Ruth A Keri
- From the Departments of Pharmacology, .,Genetics and Genome Sciences and Division of General Medical Sciences-Oncology, Case Western Reserve University, Cleveland, Ohio 44106 and
| |
Collapse
|
34
|
Niu LL, Cheng CL, Li MY, Yang SL, Hu BG, Chong CCN, Chan SL, Ren J, Chen GG, Lai PBS. ID1-induced p16/IL6 axis activation contributes to the resistant of hepatocellular carcinoma cells to sorafenib. Cell Death Dis 2018; 9:852. [PMID: 30154433 PMCID: PMC6113298 DOI: 10.1038/s41419-018-0926-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
Sorafenib is the only approved drug for the treatment of advanced hepatocellular carcinoma (HCC). However, its efficacy is limited by the emergence of primary and/or acquired resistance. Senescence-associated secretory phenotype (SASP)-mediated chemo-resistance, which depends on the secreted bioactive molecules, has attracted increasing attention but never revealed in HCC. In this study, we investigated the effect of SASP-related p16/IL6 axis on sorafenib resistance in HCC. Initially, we noticed that HCC cells with a high level of p16/IL6 axis exhibited a low sensitivity to sorafenib. Further in vivo and in vitro studies demonstrated that such a primary resistance resulted from ID1-mediated activation of p16/IL6 axis. Overexpression of ID1 or IL6 blocking in sorafenib-resistant HCC cells could increase the cytotoxicity of sorafenib. Moreover, SASP-related p16/IL6 axis contributed to the formation of acquired resistance in cells received long-term exposure to sorafenib. In acquired sorafenib-resistant cells, ID1 low expression, p16/IL6 axis up-regulation, and AKT phosphorylation activation were observed. A reduced cytotoxicity of sorafenib was detected when sorafenib-sensitive cells incubated with conditioned media from the resistant cells, accompanied by the stimulation of AKT phosphorylation. The reversal of sorafenib resistance could be achieved through ID1 overexpression, IL6 blocking, and AKT pathway inhibition. Our study reveals that SASP-related p16/IL6 axis activation is responsible for sorafenib resistance, which will be a novel strategy to prevent the drug resistance.
Collapse
Affiliation(s)
- Lei-Lei Niu
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.,Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Chuan-le Cheng
- Department of Thoracic Surgery, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, China
| | - Ming-Yue Li
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Sheng-Li Yang
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.,Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bao-Guang Hu
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.,Department of Gastrointestinal Surgery, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Charing C N Chong
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Stephen L Chan
- Department of Clinical Oncology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Jianwei Ren
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - George G Chen
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| | - Paul B S Lai
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.
| |
Collapse
|
35
|
Liu X, Zhao Y, Zhang W, Gao Y, Huo M, Liu M, Xiao Z, Liang S, Xu N, Zhu H. Inhibition of survivin enhances radiosensitivity of esophageal cancer cells by switching radiation-induced senescence to apoptosis. Onco Targets Ther 2018; 11:3087-3100. [PMID: 29872320 PMCID: PMC5975611 DOI: 10.2147/ott.s166798] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose Strategies to increase radiosensitivity are urgently needed. Combining radiosensitizing reagents with radiotherapy could improve the outcome of cancer treatment. Some preclinical studies showed that sepantronium bromide (YM155) could sensitize cancer cells to radiation by inhibiting the survivin protein. In this study, we try to investigate the function of YM155 on radiosensitivity of esophageal squamous cell carcinoma (ESCC) cells. Materials and methods ESCC cell lines were treated with radiation and YM155, and the radiation efficacy was evaluated by cell counting kit-8 assay and clonogenic survival assay. Cell senescence was measured by senescence-associated β-galactosidase staining. Terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay, fluorescein isothiocyanate-labeled Annexin V/propidium iodide assay, and poly ADP-ribose polymerase cleavage were used to detect apoptosis. KYSE150 xenografts model was used to test the efficacy of radiation combined with YM155. Results YM155 could inhibit the upregulation of survivin induced by radiation in all ESCC cell lines, but the efficacy of radiosensitization varied in different cell lines. Radiation-induced senescence in KYSE150 and KYSE410 cells, and the combination with YM155 inhibited senescence and promoted apoptosis of ESCC cells, thereby enhancing radiosensitivity. Combination with YM155 and radiation delayed the growth of KYSE150 xenografts in nude mice by switching radiation-induced senescence to apoptosis. When p21 was inhibited in KYSE150 cells, radiation did not induce senescence, and the radiosensitization of YM155 was also attenuated. In KYSE510 and KYSE180 cells, radiation did not induce senescence, and YM155 could not enhance the radiosensitivity. Conclusion Our results suggest a new mechanism that YM155 might sensitize ESCC cells to radiation by switching radiation-induced senescence to apoptosis. The major determinant of radiosensitization by YM155 might be the induction of senescence by radiation.
Collapse
Affiliation(s)
- Xianghe Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yahui Zhao
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weina Zhang
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Gao
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miaomiao Huo
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zefen Xiao
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
36
|
Dexamethasone alleviates pemetrexed-induced senescence in Non-Small-Cell Lung Cancer. Food Chem Toxicol 2018; 119:86-97. [PMID: 29753869 DOI: 10.1016/j.fct.2018.05.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/05/2018] [Accepted: 05/09/2018] [Indexed: 01/01/2023]
Abstract
Pemetrexed (PEM) is a novel and multi-targeted antifolate used as an antineoplastic agent for non-small cell lung cancer (NSCLC) and pleural mesothelioma. Although glucocorticoid was often used with PEM to reduce toxicity during the chemotherapy, it is not clear yet whether glucocorticoid co-administration could affect PEM efficacy in NSCLC. Here we established NSCLC cell lines and examined the effects of dexamethasone (DEX) on PEM sensitivity in vitro and in xenograft models. DEX co-administration reduced chemotherapy sensitivity to PEM in xenograft models. DEX co-administration promoted cell growth and weakened senescence growth arrest, such as altered secretions of proinflammatory and mitogenic cytokines, reminiscent of a senescence associate secretory phenotype (SASP). CSCs in DEX co-administration group were subsequently found to be less sensitive towards PEM treatment as measured by cell proliferation and generation of tumor spheres in the presence of PEM. Survival molecule B-cell lymphoma-2 (Bcl-2) may involve in this process and blockage of Bcl-2 could reverse altered senescence and CSCs abilities, thus alleviated PEM insensitivity. As such, DEX might suppress the antitumor activity of PEM through altered SASP level that had induced traits similar to CSCs.
Collapse
|
37
|
Camorani S, Cerchia L, Fedele M, Erba E, D'Incalci M, Crescenzi E. Trabectedin modulates the senescence-associated secretory phenotype and promotes cell death in senescent tumor cells by targeting NF-κB. Oncotarget 2018; 9:19929-19944. [PMID: 29731994 PMCID: PMC5929437 DOI: 10.18632/oncotarget.24961] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 03/13/2018] [Indexed: 11/30/2022] Open
Abstract
Therapy-induced senescence is a major cellular response to chemotherapy in solid tumors. Senescent tumor cells acquire a secretory phenotype, or SASP, and produce pro-inflammatory factors, whose expression is largely under NF-κB transcriptional control. Secreted factors play a positive role in driving antitumor immunity, but also exert negative influences on the microenvironment, and promote tumor growth and metastasis. Moreover, subsets of cancer cells can escape the senescence arrest, driving tumor recurrence after treatments. Hence, removal the senescent tumor cells, or reprogramming of the senescent secretome, have become attractive therapeutic options. The marine drug trabectedin was shown to inhibit the production of pro-inflammatory mediators by tumor-infiltrating immune cells and by myxoid liposarcoma cells. Here, we demonstrate that trabectedin inhibits the SASP, thus limiting the pro-tumoral activities of senescent tumor cells in vitro. We show that trabectedin modulates NF-κB transcriptional activity in senescent tumor cells. This results in disruption of the balance between antiapoptotic and proapoptotic signals, and sensitization of cells to Fas-mediated apoptosis. Further, we found that trabectedin inhibits escape from therapy-induced senescence, at concentrations that do not affect the viability of bulk tumor population. Overall, our data demonstrate that trabectedin has the potential to inhibit multiple detrimental effects of therapy-induced senescence.
Collapse
Affiliation(s)
- Simona Camorani
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Laura Cerchia
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Monica Fedele
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Eugenio Erba
- Dipartimento di Oncologia, IRCCS Istituto Di Ricerche Farmacologiche Mario Negri, 20156 Milan, Italy
| | - Maurizio D'Incalci
- Dipartimento di Oncologia, IRCCS Istituto Di Ricerche Farmacologiche Mario Negri, 20156 Milan, Italy
| | - Elvira Crescenzi
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| |
Collapse
|
38
|
Bafilomycin A1 triggers proliferative potential of senescent cancer cells in vitro and in NOD/SCID mice. Oncotarget 2018; 8:9303-9322. [PMID: 28030837 PMCID: PMC5354733 DOI: 10.18632/oncotarget.14066] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 12/12/2016] [Indexed: 01/16/2023] Open
Abstract
Anticancer therapies that induce DNA damage tend to trigger senescence in cancer cells, a process known as therapy-induced senescence (TIS). Such cells may undergo atypical divisions, thus contributing to tumor re-growth. Accumulation of senescent cancer cells reduces survival of patients after chemotherapy. As senescence interplays with autophagy, a dynamic recycling process, we sought to study whether inhibition of autophagy interferes with divisions of TIS cells. We exposed human colon cancer HCT116 cells to repeated cycles of a chemotherapeutic agent – doxorubicin (doxo) and demonstrated induction of hallmarks of TIS (e.g. growth arrest, hypertrophy, poliploidization and secretory phenotype) and certain properties of cancer stem cells (increased NANOG expression, percentages of CD24+ cells and side population). Colonies of small and highly proliferative progeny appeared shortly after drug removal. Treatment with bafilomycin A1 (BAF A1), an autophagy inhibitor, postponed short term in vitro cell re-population. It was associated with reduction in the number of diploid and increase in the number of poliploid cells. In a long term, a pulse of BAF A1 resulted in reactivation of autophagy in a subpopulation of HCT116 cells and increased proliferation. Accordingly, the senescent HCT116 cells treated with BAF A1 when injected into NOD/SCID mice formed tumors, in contrast to the controls. Our results suggest that senescent cancer cells that appear during therapy, can be considered as dormant cells that contribute to cancer re-growth, when chemotherapeutic treatment is stopped. These data unveil new mechanisms of TIS-related cancer maintenance and re-population, triggered by a single pulse of BAF A1 treatment.
Collapse
|
39
|
Yu X, Zhang Y, Cavazos D, Ma X, Zhao Z, Du L, Pertsemlidis A. miR-195 targets cyclin D3 and survivin to modulate the tumorigenesis of non-small cell lung cancer. Cell Death Dis 2018; 9:193. [PMID: 29416000 PMCID: PMC5833354 DOI: 10.1038/s41419-017-0219-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/01/2017] [Accepted: 12/12/2017] [Indexed: 02/07/2023]
Abstract
miR-195 has recently been reported to function as a tumor suppressor in various cancers, including non-small cell lung cancer (NSCLC). However, the mechanisms by which miR-195 represses the tumorigenesis of NSCLC cells are not fully understood. We performed a high-throughput screen using an miRNA mimic library and confirmed the identification of miR-195 as a tumor suppressor in NSCLC. We demonstrated that overexpression or induced expression of miR-195 in lung tumors slows tumor growth and that repression of miR-195 accelerates tumor growth. In addition, we found that knockout of miR-195 promotes cancer cell growth. We demonstrated that miR-195 targets cyclin D3 to cause cell cycle arrest at the G1 phase and that miR-195 targets survivin to induce apoptosis and senescence in NSCLC cells. Overexpression of cyclin D3 or survivin reverses the effects of miR-195 in NSCLC cells. Through the analysis of data from The Cancer Genome Atlas, we confirmed that the expression of miR-195 is lower in tumors than in adjacent normal tissues and that low expression of miR-195 is associated with poor survival in both lung adenocarcinoma and squamous cell carcinoma patients. Specifically, we found that BIRC5, which codes for survivin, is upregulated in both adenocarcinoma and squamous cell carcinoma tissues and that high expression of BIRC5 is associated with poor survival in adenocarcinoma, but not squamous cell carcinoma. In addition, the ratio of miR-195 level to BIRC5 level is associated with both recurrence-free and overall survival in lung adenocarcinoma. Our results suggest that the miR-195/BIRC5 axis is a potential target for treatment of lung adenocarcinoma specifically, and NSCLC in general.
Collapse
Affiliation(s)
- Xiaojie Yu
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Cell Systems and Anatomy, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yiqiang Zhang
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - David Cavazos
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xiuye Ma
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Zhenze Zhao
- Department of Chemistry and Biochemistry, Texas State University at San Marcos, San Marcos, TX, USA
| | - Liqin Du
- Department of Chemistry and Biochemistry, Texas State University at San Marcos, San Marcos, TX, USA
| | - Alexander Pertsemlidis
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA. .,Department of Cell Systems and Anatomy, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA. .,Department of Pediatrics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
40
|
Xu Q, Liu M, Zhang J, Xue L, Zhang G, Hu C, Wang Z, He S, Chen L, Ma K, Liu X, Zhao Y, Lv N, Liang S, Zhu H, Xu N. Overexpression of KLF4 promotes cell senescence through microRNA-203-survivin-p21 pathway. Oncotarget 2018; 7:60290-60302. [PMID: 27531889 PMCID: PMC5312384 DOI: 10.18632/oncotarget.11200] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 07/26/2016] [Indexed: 02/05/2023] Open
Abstract
Krüppel-like factor 4 (KLF4) is a transcription factor and functions as a tumor suppressor or tumor promoter in different cancer types. KLF4 regulates many gene expression, thus affects the process of cell proliferation, differentiation, and apoptosis. Recently, KLF4 was reported to induce senescence during the generation of induced pluripotent stem (iPS) cells, but the exact mechanism is still unclear. In this study, we constructed two doxycycline-inducing KLF4 cell models, and demonstrated overexpression of KLF4 could promote cell senescence, detected by senescence-associated β-galactosidase activity assay. Then we confirmed that p21, a key effector of senescence, was directly induced by KLF4. KLF4 could also inhibit survivin, which could indirectly induce p21. By miRNA microarray, we found a series of miRNAs regulated by KLF4 and involved in senescence. We demonstrated that KLF4 could upregulate miR-203, and miR-203 contributed to senescence through miR-203-survivin-p21 pathway. Our results suggest that KLF4 could promote cell senescence through a complex network: miR-203, survivin, and p21, which were all regulated by overexpression of KLF4 and contributed to cell senescence.
Collapse
Affiliation(s)
- Qing Xu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ju Zhang
- Division of Proteomics, Beijing Institute of Genomics, Chinese Academy of Science, Beijing, China
| | - Liyan Xue
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guo Zhang
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenfei Hu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zaozao Wang
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shun He
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lechuang Chen
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Ma
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianghe Liu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yahui Zhao
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Lv
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
Hai B, Zhao Q, Deveau MA, Liu F. Delivery of Sonic Hedgehog Gene Repressed Irradiation-induced Cellular Senescence in Salivary Glands by Promoting DNA Repair and Reducing Oxidative Stress. Theranostics 2018; 8:1159-1167. [PMID: 29464006 PMCID: PMC5817117 DOI: 10.7150/thno.23373] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/01/2017] [Indexed: 01/15/2023] Open
Abstract
Rationale: Irreversible hypofunction of salivary glands or xerostomia is common in head and neck cancer survivors treated with radiotherapy even when various new techniques are applied to minimize the irradiation (IR) damage. This condition severely impairs the quality of life of patients and can only be temporarily relieved with current treatments. We found recently that transient expression of Sonic Hedgehog (Shh) in salivary glands after IR rescued salivary function, but the underlying mechanisms are not totally clear. Methods: We generated a mouse model of IR-induced hyposalivation, and delivered adenoviral vectors carrying Shh or control GFP gene into submandibular glands (SMGs) via retrograde ductal instillation 3 days after IR. The cellular senescence was evaluated by senescence-associated beta-galactosidase assay and the expression of senescence markers. The underlying mechanisms were explored by examining DNA damage, oxidative stress, and the expression of related genes by qRT-PCR, Western blot and immunofluorescent staining. Results: Shh gene transfer repressed IR-induced cellular senescence by promoting DNA repair and decreasing oxidative stress, which is mediated through upregulating expression of genes related to DNA repair such as survivin and miR-21 and repressing expression of pro-senescence gene Gdf15 likely downstream of miR-21. Conclusion: Repressing cellular senescence contributes to the rescue of IR-induced hyposalivation by transient activation of Hh signaling, which is related to enhanced DNA repair and decreased oxidative stress in SMGs.
Collapse
|
42
|
Svobodova M, Raudenska M, Gumulec J, Balvan J, Fojtu M, Kratochvilova M, Polanska H, Horakova Z, Kostrica R, Babula P, Heger Z, Masarik M. Establishment of oral squamous cell carcinoma cell line and magnetic bead-based isolation and characterization of its CD90/CD44 subpopulations. Oncotarget 2017; 8:66254-66269. [PMID: 29029509 PMCID: PMC5630409 DOI: 10.18632/oncotarget.19914] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/28/2017] [Indexed: 12/26/2022] Open
Abstract
In this study, we describe the establishment of the human papillomavirus 18-positive, stage II, grade 1, T2N0M0 head and neck tumor primary cell line derived from oral squamous cell carcinoma of a non-smoking patient by using two different protocols. Furthermore, a preparation of subpopulations derived from this primary cell line according to the cluster of differentiation molecules CD44/CD90 status using magnetic bead-based separation and their characterization was performed. Impedance-based real-time cell analysis, enzyme-linked immunsorbant assay (ELISA), wound-healing assay, flow-cytometry, gene expression analysis, and MTT assay were used to characterize these four subpopulations (CD44+/CD90-, CD44-/CD90-, CD44+/CD90+, CD44-/CD90-). We optimised methodics for establishement of primary cell lines derived from oral squamous cell carcinoma tissue samples and subsequent separation of mesenchymal (CD90+) and epithelial (CD90-) types of tumorous cells. Primary cell line prepared by using trypsin proteolysis was more viable than the one prepared by using collagenase. According to our results, CD90 separation is a necessary step in preparation of permanent tumor-tissue derived cell lines. Based on the wound-healing assay, CD44+ cells exhibited stronger migratory capacity than CD44- subpopulations. CD44+ subpopulations had also significantly higher expression of BIRC5 and SOX2, lower expression of FLT1 and IL6, and higher levels of basal autophagy compared to CD44- subpopulations. Furthermore, co-cultivation experiments revealed that CD44-/CD90+ cells supported growth of epithelial tumor cells (CD44+/CD90-). On the contrary, factors released by CD44+/CD90+ type of cells seem to have rather inhibiting effect. The most cisplatin-resistant subpopulation with the shortest doubling time was CD44-/CD90+, but this subpopulation had a low migratory capacity.
Collapse
Affiliation(s)
- Marketa Svobodova
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Martina Raudenska
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Jaromir Gumulec
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Jan Balvan
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Michaela Fojtu
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Monika Kratochvilova
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Hana Polanska
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
| | - Zuzana Horakova
- Department of Otorhinolaryngology and Head and Neck Surgery, St. Anne’s Faculty Hospital, CZ-65691 Brno, Czech Republic
| | - Rom Kostrica
- Department of Otorhinolaryngology and Head and Neck Surgery, St. Anne’s Faculty Hospital, CZ-65691 Brno, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Zbynek Heger
- Central European Institute of Technology, Brno University of Technology, CZ-61600 Brno, Czech Republic
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
| | - Michal Masarik
- Department of Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic
| |
Collapse
|
43
|
Srdic-Rajic T, Santibañez JF, Kanjer K, Tisma-Miletic N, Cavic M, Galun D, Jevric M, Kardum N, Konic-Ristic A, Zoranovic T. Iscador Qu inhibits doxorubicin-induced senescence of MCF7 cells. Sci Rep 2017. [PMID: 28630419 PMCID: PMC5476621 DOI: 10.1038/s41598-017-03898-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chemotherapy in patients with inoperable or advanced breast cancer inevitably results in low-dose exposure of tumor-cell subset and senescence. Metabolically active senescent cells secrete multiple tumor promoting factors making their elimination a therapeutic priority. Viscum album is one of the most widely used alternative anti-cancer medicines facilitating chemotherapy tolerance of breast cancer patients. The aim of this study was to model and investigate how Viscum album extracts execute additive anti-tumor activity with low-dose Dox using ER + MCF7 breast cancer cells. We report that cotreatment of MCF7 with Viscum album and Dox abrogates G2/M cycle arrest replacing senescence with intrinsic apoptotic program. Mechanistically, this switch was associated with down-regulation of p21, p53/p73 as well as Erk1/2 and p38 activation. Our findings, therefore, identify a novel mechanistic axis of additive antitumor activity of Viscum album and low dose-Dox. In conclusion, ER + breast cancer patients may benefit from addition of Viscum album to low-dose Dox chemotherapy due to suppression of cancer cell senescence and induction of apoptosis.
Collapse
Affiliation(s)
- Tatjana Srdic-Rajic
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia
| | - Juan F Santibañez
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia.,Laboratorio de Bionanotecnologia, Universidad Bernardo O Higgins, General Gana 1780, 8370854, Santiago, Chile
| | - Ksenija Kanjer
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia
| | - Nevena Tisma-Miletic
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia
| | - Milena Cavic
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia
| | - Daniel Galun
- University Clinic for Digestive Surgery, Clinical center of Serbia, Belgrade, Serbia.,Medical School, University of Belgrade, Belgrade, Serbia
| | - Marko Jevric
- Department of Surgery, National Cancer Research Center, Belgrade, Serbia
| | - Nevena Kardum
- Institute for Medical Research, Center of Research Excellence in Nutrition and Metabolism, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Konic-Ristic
- Institute for Medical Research, Center of Research Excellence in Nutrition and Metabolism, University of Belgrade, Belgrade, Serbia
| | - Tamara Zoranovic
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia. .,Max Plank Institute for Infection Biology, Berlin Area, Germany.
| |
Collapse
|
44
|
Song Z, Pan Y, Ling G, Wang S, Huang M, Jiang X, Ke Y. Escape of U251 glioma cells from temozolomide-induced senescence was modulated by CDK1/survivin signaling. Am J Transl Res 2017; 9:2163-2180. [PMID: 28559969 PMCID: PMC5446501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 04/06/2017] [Indexed: 06/07/2023]
Abstract
Temozolomide (TMZ) has been widely used in conjunction with radiotherapy for treating various types of cancers. However, tumor cells arrested in senescence due to TMZ administration can sometimes escape and become drug resistant. In the current study, the possible role of survivin in the senescence escape of TMZ-treated glioma cells was comprehensively studied. The levels of survivin and CDK1 expression in a human glioma cell line (U251) were monitored, and cell apoptosis, cell cycle distribution, anchorage-independent growth, and senescence were studied in U251 cells in different degrees of senescence. To further investigate how survivin affects the TMZ-resistance of gliomas, we modulated the levels of survivin and CKD1 expression in TMZ-treated cells and then examined how the treated cells responded. The results showed that knockdown of the survivin gene increased the sensitivity of glioma cells to TMZ treatment by inducing senescent cells to become apoptotic. Moreover, after senescence was induced, expression of the survivin gene became suppressed, but survivin levels returned to normal after the cells had escaped from senescence. While down-regulation of the survivin gene in senescent and senescence-escaping U251 cells had no effect on cell apoptosis, cell cycle distribution, or senescence status, it dramatically reduced the anchorage-independent growth ability of the cells. Additionally, CDK1 was able to not only enhance the anchorage-independent growth ability of the cells, but also contribute to their further senescence escape by modulating the survivin and other pathways. In conclusion, the survivin gene was necessary for glioma cells to escape from and enter into senescence during treatment with TMZ.
Collapse
Affiliation(s)
- Zhenhua Song
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
- The Neurosurgery Institute of Guangdong, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
| | - Yunyun Pan
- Department of Pharmaceutical, The First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhou 510080, China
- Department of Clinical Pharmacy, Guangdong Pharmaceutical UniversityGuangzhou 510080, China
| | - Gengqiang Ling
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
- The Neurosurgery Institute of Guangdong, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
| | - Shiyong Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
- The Neurosurgery Institute of Guangdong, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
| | - Min Huang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
- The Neurosurgery Institute of Guangdong, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
| | - Xiaodan Jiang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
- The Neurosurgery Institute of Guangdong, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
| | - Yiquan Ke
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
- The Neurosurgery Institute of Guangdong, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical UniversityGuangzhou 510282, China
| |
Collapse
|
45
|
Stobiecka M, Chalupa A. DNA Strand Replacement Mechanism in Molecular Beacons Encoded for the Detection of Cancer Biomarkers. J Phys Chem B 2016; 120:4782-90. [PMID: 27187043 DOI: 10.1021/acs.jpcb.6b03475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signaling properties of a fluorescent hairpin oligonucleotide molecular beacon (MB) encoded to recognize protein survivin (Sur) mRNA have been investigated. The process of complementary target binding to SurMB with 20-mer loop sequence is spontaneous, as expected, and characterized by a high affinity constant (K = 2.51 × 10(16) M(-1)). However, the slow kinetics at room temperature makes it highly irreversible. To understand the intricacies of target binding to MB, a detailed kinetic study has been performed to determine the rate constants and activation energy Ea for the reaction at physiological temperature (37 °C). Special attention has been paid to assess the value of Ea in view of reports of negative activation enthalpy for some nucleic acid reactions that would make the target binding even slower at increasing temperatures in a non-Arrhenius process. The target-binding rate constant determined is k = 3.99 × 10(3) M(-1) s(-1) at 37 °C with Ea = 28.7 ± 2.3 kcal/mol (120.2 ± 9.6 kJ/mol) for the temperature range of 23 to 55 °C. The positive high value of Ea is consistent with a kinetically controlled classical Arrhenius process. We hypothesize that the likely contribution to the activation energy barrier comes from the SurMB stem melting (tm = 53.7 ± 0.2 °C), which is a necessary step in the completion of target strand hybridization with the SurMB loop. A low limit of detection (LOD = 2 nM) for target tDNA has been achieved. Small effects of conformational polymorphs of SurMB have been observed on melting curves. Although these polymorphs could potentially cause a negative Ea, their effect on kinetic transients for target binding is negligible. No toehold preceding steps in the mechanism of target binding were identified.
Collapse
Affiliation(s)
- Magdalena Stobiecka
- Department of Biophysics, Warsaw University of Life Sciences (SGGW) , 02776 Warsaw, Poland
| | - Agata Chalupa
- Institute of Nanoparticle Nanocarriers , 11010 Barczewo, Poland
| |
Collapse
|
46
|
Unruhe B, Schröder E, Wünsch D, Knauer SK. An Old Flame Never Dies: Survivin in Cancer and Cellular Senescence. Gerontology 2015; 62:173-81. [PMID: 26159786 DOI: 10.1159/000432398] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/18/2015] [Indexed: 11/19/2022] Open
Abstract
Survivin (BIRC5) is highly expressed in the vast majority of human cancers and is associated with chemotherapy resistance, increased tumor recurrence and shortened patient survival, making it an attractive therapeutic target. Initially identified as an inhibitor of apoptosis protein, it also plays a major role in the regulation of cell division. As such, it acts as a subunit of the chromosomal passenger complex, composed of the mitotic kinase aurora B, borealin and inner centromere protein, and is essential for proper chromosome segregation and cytokinesis. For both biological functions, interaction of survivin's nuclear export signal with the nuclear export receptor chromosome region maintenance 1 is absolutely essential. The timely orchestration of survivin's wide protein interaction repertoire is further modulated by different posttranslational modifications occurring in a cell-cycle-dependent manner. Recent data furthermore indicate additional roles of survivin in the DNA damage response, contributing to therapy resistance, yet the underlying molecular details are still not completely resolved. This also holds true for a potential involvement of survivin in senescence regulation. An age-related accumulation of survivin probably contributes to the apoptosis resistance observed in aged as well as in senescent cells, while it might promote escape from therapy-induced senescence. This review seeks to integrate the current knowledge on survivin's diverse and complex biological functions. By linking the 'old' facts about survivin with recent findings in research areas such as DNA damage response and aging, we want to highlight survivin's crucial role in a variety of cellular processes.
Collapse
Affiliation(s)
- Britta Unruhe
- Institute for Molecular Biology II, Centre for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
| | | | | | | |
Collapse
|
47
|
Kaur E, Rajendra J, Jadhav S, Shridhar E, Goda JS, Moiyadi A, Dutt S. Radiation-induced homotypic cell fusions of innately resistant glioblastoma cells mediate their sustained survival and recurrence. Carcinogenesis 2015; 36:685-95. [PMID: 25863126 DOI: 10.1093/carcin/bgv050] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/23/2015] [Indexed: 12/13/2022] Open
Abstract
Understanding of molecular events underlying resistance and relapse in glioblastoma (GBM) is hampered due to lack of accessibility to resistant cells from patients undergone therapy. Therefore, we mimicked clinical scenario in an in vitro cellular model developed from five GBM grade IV primary patient samples and two cell lines. We show that upon exposure to lethal dose of radiation, a subpopulation of GBM cells, innately resistant to radiation, survive and transiently arrest in G2/M phase via inhibitory pCdk1(Y15). Although arrested, these cells show multinucleated and giant cell phenotype (MNGC). Significantly, we demonstrate that these MNGCs are not pre-existing giant cells from parent population but formed via radiation-induced homotypic cell fusions among resistant cells. Furthermore, cell fusions induce senescence, high expression of senescence-associated secretory proteins (SASPs) and activation of pro-survival signals (pAKT, BIRC3 and Bcl-xL) in MNGCs. Importantly, following transient non-proliferation, MNGCs escape senescence and despite having multiple spindle poles during mitosis, they overcome mitotic catastrophe to undergo normal cytokinesis forming mononucleated relapse population. This is the first report showing radiation-induced homotypic cell fusions as novel non-genetic mechanism in radiation-resistant cells to sustain survival. These data also underscore the importance of non-proliferative phase in resistant glioma cells. Accordingly, we show that pushing resistant cells into premature mitosis by Wee1 kinase inhibitor prevents pCdk1(Y15)-mediated cell cycle arrest and relapse. Taken together, our data provide novel molecular insights into a multistep process of radiation survival and relapse in GBM that can be exploited for therapeutic interventions.
Collapse
Affiliation(s)
- Ekjot Kaur
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Jacinth Rajendra
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Shailesh Jadhav
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Epari Shridhar
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Jayant Sastri Goda
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Aliasgar Moiyadi
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Shilpee Dutt
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| |
Collapse
|
48
|
Poleszczuk J, Krzywon A, Forys U, Widel M. Connecting radiation-induced bystander effects and senescence to improve radiation response prediction. Radiat Res 2015; 183:571-7. [PMID: 25844948 DOI: 10.1667/rr13907.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
For the last two decades radiation-induced bystander effects (RIBEs) have attracted significant attention due to their possible implications for radiotherapy. However, despite extensive research, the molecular pathways associated with RIBEs are still not completely known. In the current study we investigated the role of senescence in the bystander response. Irradiated (2, 4, 6 and 8 Gy) human colorectal carcinoma cells (HCT116) with p53(+/+) (wild-type) or p53(-/-) (knockout) gene were co-incubated with nonirradiated cells of the same type. Clonogenic and senescence assays were used for both irradiated and co-incubated bystander cell populations. We also performed additional measurements on the number of remaining cells after the whole co-incubation period. For radiation doses larger than 2 Gy we observed much larger fractions of senescent cells in p53-positive populations compared to their p53-negative counterparts (15.81% vs. 3.63% in the irradiated population; 2.89% vs. 1.05% in the bystander population; 8 Gy; P < 0.05). Statistically significant differences between cell lines in the clonogenic cell surviving fraction were observed for doses higher than 4 Gy (1.61% for p53(+/+) vs. 0.19% for p53(-/-) in irradiated population; 3.57% for +/+ vs. 50.39% for -/- in bystander population; 8 Gy; P < 0.05). Our main finding was that the number of senescent cells in the irradiated population correlated strongly with the clonogenic cell surviving fraction (R = -0.98, P < 0.001) and the number of senescent cells (R = 0.97, P < 0.001) in the bystander population. We also extended the standard linear-quadratic radiation response model by incorporating the influence of the signals released by the senescent cells, which accurately described the radiation response in the bystander population. Our findings suggest that radiation-induced senescence might be a key player in RIBE, i.e., the strength of RIBE depends on the amount of radiation-induced senescence.
Collapse
Affiliation(s)
- Jan Poleszczuk
- a College of Inter-faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, Warsaw, Poland
| | | | | | | |
Collapse
|
49
|
Artesi M, Kroonen J, Bredel M, Nguyen-Khac M, Deprez M, Schoysman L, Poulet C, Chakravarti A, Kim H, Scholtens D, Seute T, Rogister B, Bours V, Robe PA. Connexin 30 expression inhibits growth of human malignant gliomas but protects them against radiation therapy. Neuro Oncol 2014; 17:392-406. [PMID: 25155356 DOI: 10.1093/neuonc/nou215] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 07/28/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Glioblastomas remain ominous tumors that almost invariably escape treatment. Connexins are a family of transmembrane, gap junction-forming proteins, some members of which were reported to act as tumor suppressors and to modulate cellular metabolism in response to cytotoxic stress. METHODS We analyzed the copy number and expression of the connexin (Cx)30 gene gap junction beta-6 (GJB6), as well as of its protein immunoreactivity in several public and proprietary repositories of glioblastomas, and their influence on patient survival. We evaluated the effect of the expression of this gap junction protein on the growth, DNA repair and energy metabolism, and treatment resistance of these tumors. RESULTS The GJB6 gene was deleted in 25.8% of 751 analyzed tumors and mutated in 15.8% of 158 tumors. Cx30 immunoreactivity was absent in 28.9% of 145 tumors. Restoration of Cx30 expression in human glioblastoma cells reduced their growth in vitro and as xenografts in the striatum of immunodeficient mice. Cx30 immunoreactivity was, however, found to adversely affect survival in 2 independent retrospective cohorts of glioblastoma patients. Cx30 was found in clonogenic assays to protect glioblastoma cells against radiation-induced mortality and to decrease radiation-induced DNA damage. This radioprotection correlated with a heat shock protein 90-dependent mitochondrial translocation of Cx30 following radiation and an improved ATP production following this genotoxic stress. CONCLUSION These results underline the complex relationship between potential tumor suppressors and treatment resistance in glioblastomas and single out GJB6/Cx30 as a potential biomarker and target for therapeutic intervention in these tumors.
Collapse
Affiliation(s)
- Maria Artesi
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Jerome Kroonen
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Markus Bredel
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Minh Nguyen-Khac
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Manuel Deprez
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Laurent Schoysman
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Christophe Poulet
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Arnab Chakravarti
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Hyunsoo Kim
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Denise Scholtens
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Tatjana Seute
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Bernard Rogister
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Vincent Bours
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Pierre A Robe
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| |
Collapse
|
50
|
Zhang MZ, Li C, Fang BY, Yao MH, Ren QQ, Zhang L, Zhao YD. High transfection efficiency of quantum dot-antisense oligonucleotide nanoparticles in cancer cells through dual-receptor synergistic targeting. NANOTECHNOLOGY 2014; 25:255102. [PMID: 24896735 DOI: 10.1088/0957-4484/25/25/255102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Incorporating ligands with nanoparticle-based carriers for specific delivery of therapeutic nucleic acids (such as antisense oligonucleotides and siRNA) to tumor sites is a promising approach in anti-cancer strategies. However, nanoparticle-based carriers remain insufficient in terms of the selectivity and transfection efficiency. In this paper, we designed a dual receptor-targeted QDs gene carrier QD-(AS-ODN+GE11+c(RGDfK)) which could increase the cellular uptake efficiency and further enhance the transfection efficiency. Here, the targeting ligands used were peptides GE11 and c(RGDfK) which could recognize epidermal growth factor receptors (EGFR) and integrin ανβ3 receptors, respectively. Quantitative flow cytometry and ICP/MS showed that the synergistic effect between EGFR and integrin ανβ3 increased the cellular uptake of QDs carriers. The effects of inhibition agents showed the endocytosis pathway of QD-(AS-ODN+GE11+c(RGDfK)) probe was mainly clathrin-mediated. Western blot confirmed that QD-(AS-ODN+GE11+c(RGDfK)) could further enhance gene silencing efficiency compared to QD-(AS-ODN+GE11) and QD-(AS-ODN+c(RGDfK)), suggesting this dual receptor-targeted gene carrier achieved desired transfection efficiency. In this gene delivery system, QDs could not only be used as a gene vehicle but also as fluorescence probe, allowing for localization and tracking during the delivery process. This transport model is very well referenced for non-viral gene carriers to enhance the targeting ability and transfection efficiency.
Collapse
Affiliation(s)
- Ming-Zhen Zhang
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|