1
|
He Y, Qiu Y, Yang X, Lu G, Zhao SS. Remodeling of tumor microenvironment by cellular senescence and immunosenescence in cervical cancer. Semin Cancer Biol 2025; 108:17-32. [PMID: 39586414 DOI: 10.1016/j.semcancer.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
Cellular senescence is a response to various stress signals, which is characterized by stable cell cycle arrest, alterations in cellular morphology, metabolic reprogramming and production of senescence-associated secretory phenotype (SASP). When it occurs in the immune system, it is called immunosenescence. Cervical cancer is a common gynecological malignancy, and cervical cancer screening is generally recommended before the age of 65. Elderly women (≥65 years) are more often diagnosed with advanced disease and have poorer prognosis compared to younger patients. Despite extensive research, the tumor microenvironment requires more in-depth exploration, particularly in elderly patients. In cervical cancer, senescent cells have a double-edged sword effect on tumor progression. Induction of preneoplastic cell senescence prevents tumor initiation, and several treatment approaches of cervical cancer act in part by inducing cancer cell senescence. However, senescent immune cell populations within the tumor microenvironment facilitate tumor development, recurrence, treatment resistance, etc. Amplification of beneficial effects and inhibition of aging-related pro-tumorigenic pathways contribute to improving antitumor effects. This review discusses senescent cancer and immune cells present in the tumor microenvironment of cervical cancer and how these senescent cells and their SASP remodel the tumor microenvironment, influence antitumor immunity and tumor initiation and development. Moreover, we discuss the significance of senotherapeutics that enable to eliminate senescent cells and prevent tumor progression and development through improving antitumor immunity and affecting the tumor microenvironment.
Collapse
Affiliation(s)
- Yijiang He
- Abdominal Radiation Oncology Ward II, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yue Qiu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Xiansong Yang
- Department of Day Chemotherapy Ward, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, Shandong 266042, China
| | - Guimei Lu
- Department of Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Shan-Shan Zhao
- Department of Gynecology Surgery 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
2
|
Ibrahim Fouad G, Rizk MZ. Neurotoxicity of the antineoplastic drugs: "Doxorubicin" as an example. J Mol Histol 2024; 55:1023-1050. [PMID: 39352546 DOI: 10.1007/s10735-024-10247-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/11/2024] [Indexed: 11/16/2024]
Abstract
There is an increased prevalence of cancer, and chemotherapy is widely and routinely utilized to manage the majority of cancers; however, administration of chemotherapeutic drugs has faced limitations concerning the "off-target" cytotoxicity. Chemobrain and impairment of neurocognitive functions have been observed in a significant fraction of cancer patients or survivors and reduce their life quality; this could be ascribed to the ability of chemotherapeutic drugs to alter the structure and function of the brain. Doxorubicin (DOX), an FDA-approved chemotherapeutic drug with therapeutic effectiveness, is commonly used to treat several carcinomas clinically. DOX-triggered neurotoxicity is the most serious adverse reaction after DOX-induced cardiotoxicity which greatly limits its clinical application. DOX-induced neurotoxicity is a net of multiple mechanisms that have been verified in pre-clinical and clinical studies, such as oxidative stress, neuroinflammation, mitochondrial disruption, apoptosis, autophagy, disruption of neurotransmitters, and impairment of neurogenesis. There is a massive need for developing novel therapeutics for both cancer and DOX-associated neurotoxicity; therefore investigating the implicated mechanisms of DOX-induced chemobrain will reveal multi-targets for novel curative strategies. Recently, various neuroprotective mechanisms were employed to mitigate DOX-mediated neurotoxicity. For this purpose, therapeutic interventions using pharmacological compounds were developed to protect healthy "off-target" tissues from DOX-induced toxicity. In addition, nanoplatforms were used to enable target delivery of DOX; to prevent its deposition in non-cancerous tissues. The aim of the current review is to provide some reference value for the future management of DOX-induced neurotoxicity and to summarize the underlying mechanisms of DOX-mediated neurotoxicity and the potential therapeutic interventions.
Collapse
Affiliation(s)
- Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt.
| | - Maha Z Rizk
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt
| |
Collapse
|
3
|
Pundlik SS, Barik A, Venkateshvaran A, Sahoo SS, Jaysingh MA, Math RGH, Lal H, Hashmi MA, Ramanathan A. Senescent cells inhibit mouse myoblast differentiation via the SASP-lipid 15d-PGJ 2 mediated modification and control of HRas. eLife 2024; 13:RP95229. [PMID: 39196610 PMCID: PMC11357351 DOI: 10.7554/elife.95229] [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] [Indexed: 08/29/2024] Open
Abstract
Senescent cells are characterized by multiple features such as increased expression of senescence-associated β-galactosidase activity (SA β-gal) and cell cycle inhibitors such as p21 or p16. They accumulate with tissue damage and dysregulate tissue homeostasis. In the context of skeletal muscle, it is known that agents used for chemotherapy such as Doxorubicin (Doxo) cause buildup of senescent cells, leading to the inhibition of tissue regeneration. Senescent cells influence the neighboring cells via numerous secreted factors which form the senescence-associated secreted phenotype (SASP). Lipids are emerging as a key component of SASP that can control tissue homeostasis. Arachidonic acid-derived lipids have been shown to accumulate within senescent cells, specifically 15d-PGJ2, which is an electrophilic lipid produced by the non-enzymatic dehydration of the prostaglandin PGD2. This study shows that 15d-PGJ2 is also released by Doxo-induced senescent cells as an SASP factor. Treatment of skeletal muscle myoblasts with the conditioned medium from these senescent cells inhibits myoblast fusion during differentiation. Inhibition of L-PTGDS, the enzyme that synthesizes PGD2, diminishes the release of 15d-PGJ2 by senescent cells and restores muscle differentiation. We further show that this lipid post-translationally modifies Cys184 of HRas in C2C12 mouse skeletal myoblasts, causing a reduction in the localization of HRas to the Golgi, increased HRas binding to Ras Binding Domain (RBD) of RAF Kinase (RAF-RBD), and activation of cellular Mitogen Activated Protein (MAP) kinase-Extracellular Signal Regulated Kinase (Erk) signaling (but not the Akt signaling). Mutating C184 of HRas prevents the ability of 15d-PGJ2 to inhibit the differentiation of muscle cells and control the activity of HRas. This work shows that 15d-PGJ2 released from senescent cells could be targeted to restore muscle homeostasis after chemotherapy.
Collapse
Affiliation(s)
- Swarang Sachin Pundlik
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Alok Barik
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| | - Ashwin Venkateshvaran
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| | - Snehasudha Subhadarshini Sahoo
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- University of North Carolina at Chapel HillChapel HillUnited States
| | - Mahapatra Anshuman Jaysingh
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K)MohanpurIndia
- Division of Biology and Biomedical Sciences, Washington University in St LouisSt LouisUnited States
| | | | - Heera Lal
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Maroof Athar Hashmi
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Arvind Ramanathan
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| |
Collapse
|
4
|
McClelland S, Maxwell PJ, Branco C, Barry ST, Eberlein C, LaBonte MJ. Targeting IL-8 and Its Receptors in Prostate Cancer: Inflammation, Stress Response, and Treatment Resistance. Cancers (Basel) 2024; 16:2797. [PMID: 39199570 PMCID: PMC11352248 DOI: 10.3390/cancers16162797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
This review delves into the intricate roles of interleukin-8 (IL-8) and its receptors, CXCR1 and CXCR2, in prostate cancer (PCa), particularly in castration-resistant (CRPC) and metastatic CRPC (mCRPC). This review emphasizes the crucial role of the tumour microenvironment (TME) and inflammatory cytokines in promoting tumour progression and response to tumour cell targeting agents. IL-8, acting through C-X-C chemokine receptor type 1 (CXCR1) and type 2 (CXCR2), modulates multiple signalling pathways, enhancing the angiogenesis, proliferation, and migration of cancer cells. This review highlights the shift in PCa research focus from solely tumour cells to the non-cancer-cell components, including vascular endothelial cells, the extracellular matrix, immune cells, and the dynamic interactions within the TME. The immunosuppressive nature of the PCa TME significantly influences tumour progression and resistance to emerging therapies. Current treatment modalities, including androgen deprivation therapy and chemotherapeutics, encounter persistent resistance and are complicated by prostate cancer's notably "immune-cold" nature, which limits immune system response to the tumour. These challenges underscore the critical need for novel approaches that both overcome resistance and enhance immune engagement within the TME. The therapeutic potential of inhibiting IL-8 signalling is explored, with studies showing enhanced sensitivity of PCa cells to treatments, including radiation and androgen receptor inhibitors. Clinical trials, such as the ACE trial, demonstrate the efficacy of combining CXCR2 inhibitors with existing treatments, offering significant benefits, especially for patients with resistant PCa. This review also addresses the challenges in targeting cytokines and chemokines, noting the complexity of the TME and the need for precision in therapeutic targeting to avoid side effects and optimize outcomes.
Collapse
Affiliation(s)
- Shauna McClelland
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (S.M.); (P.J.M.); (C.B.)
| | - Pamela J. Maxwell
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (S.M.); (P.J.M.); (C.B.)
| | - Cristina Branco
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (S.M.); (P.J.M.); (C.B.)
| | - Simon T. Barry
- Bioscience Early Oncology, AstraZeneca, Cambridge CB2 0AA, UK; (S.T.B.); (C.E.)
| | - Cath Eberlein
- Bioscience Early Oncology, AstraZeneca, Cambridge CB2 0AA, UK; (S.T.B.); (C.E.)
| | - Melissa J. LaBonte
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (S.M.); (P.J.M.); (C.B.)
| |
Collapse
|
5
|
Oyama K, Iwagami Y, Kobayashi S, Sasaki K, Yamada D, Tomimaru Y, Noda T, Asaoka T, Takahashi H, Tanemura M, Doki Y, Eguchi H. Removal of gemcitabine-induced senescent cancer cells by targeting glutaminase1 improves the therapeutic effect in pancreatic ductal adenocarcinoma. Int J Cancer 2024; 154:912-925. [PMID: 37699232 DOI: 10.1002/ijc.34725] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023]
Abstract
Insufficient cancer treatment can induce senescent cancer cell formation and treatment resistance. The characteristics of induced senescent cancer (iSnCa) cells remain unclear. Pancreatic ductal adenocarcinoma (PDAC) has a low and nondurable response rate to current treatments. Our study aimed to analyze the properties of iSnCa cells and the relationship between cellular senescence and prognosis in PDAC. We evaluated the characteristics of gemcitabine-induced senescent cancer cells and the effect of senescence-associated secretory phenotype (SASP) factors released by iSnCa cells on surrounding PDAC cells. The relationship between cellular senescence and the prognosis was investigated in 50 patients with PDAC treated with gemcitabine-based neoadjuvant chemotherapy. Exposure to 5 ng/mL gemcitabine-induced senescence, decreased proliferation and increased senescence-associated β-galactosidase-cell staining without cell death in PDAC cells; the expression of glutaminase1 (GLS1) and SASP factors also increased and caused epithelial-mesenchymal transition in surrounding PDAC cells. iSnCa cells were selectively removed by the GLS1 inhibitor bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) through apoptosis induction. Cellular senescence was induced in PDAC cells via insufficient gemcitabine in subcutaneous tumor model mice. GLS1 expression was an independent prognostic factor in patients with PDAC who received gemcitabine-based neoadjuvant chemotherapy. This is the first study to identify the relationship between senescence and GLS1 in PDAC. Low-dose gemcitabine-induced senescence and increased GLS1 expression were observed in PDAC cells. Cellular senescence may contribute to treatment resistance of PDAC, hence targeting GLS1 in iSnCa cells may improve the therapeutic effect.
Collapse
Affiliation(s)
- Keisuke Oyama
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tadafumi Asaoka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Surgery, Osaka Police Hospital, Osaka, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masahiro Tanemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Surgery, Rinku General Medical Center, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
6
|
Smoots SG, Schreiber AR, Jackson MM, Bagby SM, Dominguez ATA, Dus ED, Binns CA, MacBeth M, Whitty PA, Diamond JR, Pitts TM. Overcoming doxorubicin resistance in triple-negative breast cancer using the class I-targeting HDAC inhibitor bocodepsin/OKI-179 to promote apoptosis. Breast Cancer Res 2024; 26:35. [PMID: 38429789 PMCID: PMC10908182 DOI: 10.1186/s13058-024-01799-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with a poor prognosis. Doxorubicin is part of standard curative therapy for TNBC, but chemotherapy resistance remains an important clinical challenge. Bocodepsin (OKI-179) is a small molecule class I histone deacetylase (HDAC) inhibitor that promotes apoptosis in TNBC preclinical models. The purpose of this study was to investigate the combination of bocodepsin and doxorubicin in preclinical TNBC models and evaluate the impact on terminal cell fate, including apoptosis and senescence. METHODS TNBC cell lines were treated with doxorubicin and CellTiter-Glo was used to assess proliferation and determine doxorubicin sensitivity. Select cell lines were treated with OKI-005 (in vitro version of bocodepsin) and doxorubicin and assessed for proliferation, apoptosis as measured by Annexin V/PI, and cell cycle by flow cytometry. Immunoblotting was used to assess changes in mediators of apoptosis, cell cycle arrest, and senescence. Senescence was measured by the senescence-associated β-galactosidase assay. An MDA-MB-231 xenograft in vivo model was treated with bocodepsin, doxorubicin, or the combination and assessed for inhibition of tumor growth. shRNA knockdown of p53 was performed in the CAL-51 cell line and proliferation, apoptosis and senescence were assessed in response to combination treatment. RESULTS OKI-005 and doxorubicin resulted in synergistic antiproliferative activity in TNBC cells lines regardless of p53 mutation status. The combination led to increased apoptosis and decreased senescence. In vivo, the combination resulted in increased tumor growth inhibition compared to either single agent. shRNA knock-down of p53 led to increased doxorubicin-induced senescence that was decreased with the addition of OKI-005 in vitro. CONCLUSION The addition of bocodepsin to doxorubicin resulted in synergistic antiproliferative activity in vitro, improved tumor growth inhibition in vivo, and promotion of apoptosis which makes this a promising combination to overcome doxorubicin resistance in TNBC. Bocodepsin is currently in clinical development and has a favorable toxicity profile compared to other HDAC inhibitors supporting the feasibility of evaluating this combination in patients with TNBC.
Collapse
Affiliation(s)
- Stephen G Smoots
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Anna R Schreiber
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Marilyn M Jackson
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Stacey M Bagby
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Adrian T A Dominguez
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Evan D Dus
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Cameron A Binns
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Morgan MacBeth
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Phaedra A Whitty
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Jennifer R Diamond
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA
| | - Todd M Pitts
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue MS8117, Aurora, CO, 80045, USA.
| |
Collapse
|
7
|
Thapa BV, Banerjee M, Glimm T, Saini DK, Bhat R. The senescent mesothelial matrix accentuates colonization by ovarian cancer cells. Cell Mol Life Sci 2023; 81:2. [PMID: 38043093 PMCID: PMC10694112 DOI: 10.1007/s00018-023-05017-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 12/05/2023]
Abstract
Ovarian cancer is amongst the most morbid of gynecological malignancies due to its diagnosis at an advanced stage, a transcoelomic mode of metastasis, and rapid transition to chemotherapeutic resistance. Like all other malignancies, the progression of ovarian cancer may be interpreted as an emergent outcome of the conflict between metastasizing cancer cells and the natural defense mounted by microenvironmental barriers to such migration. Here, we asked whether senescence in coelom-lining mesothelia, brought about by drug exposure, affects their interaction with disseminated ovarian cancer cells. We observed that cancer cells adhered faster on senescent human and murine mesothelial monolayers than on non-senescent controls. Time-lapse epifluorescence microscopy showed that mesothelial cells were cleared by a host of cancer cells that surrounded the former, even under sub-confluent conditions. A multiscale computational model predicted that such colocalized mesothelial clearance under sub-confluence requires greater adhesion between cancer cells and senescent mesothelia. Consistent with the prediction, we observed that senescent mesothelia expressed an extracellular matrix with higher levels of fibronectin, laminins and hyaluronan than non-senescent controls. On senescent matrix, cancer cells adhered more efficiently, spread better, and moved faster and persistently, aiding the spread of cancer. Inhibition assays using RGD cyclopeptides suggested the adhesion was predominantly contributed by fibronectin and laminin. These findings led us to propose that the senescence-associated matrisomal phenotype of peritoneal barriers enhances the colonization of invading ovarian cancer cells contributing to the metastatic burden associated with the disease.
Collapse
Affiliation(s)
- Bharat Vivan Thapa
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
- Undergraduate Program, Indian Institute of Science, Bangalore, 560012, India
| | - Mallar Banerjee
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Tilmann Glimm
- Department of Mathematics, Western Washington University, Bellingham, WA, 98229, USA
| | - Deepak K Saini
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India.
| | - Ramray Bhat
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
8
|
Gureghian V, Herbst H, Kozar I, Mihajlovic K, Malod-Dognin N, Ceddia G, Angeli C, Margue C, Randic T, Philippidou D, Nomigni MT, Hemedan A, Tranchevent LC, Longworth J, Bauer M, Badkas A, Gaigneaux A, Muller A, Ostaszewski M, Tolle F, Pržulj N, Kreis S. A multi-omics integrative approach unravels novel genes and pathways associated with senescence escape after targeted therapy in NRAS mutant melanoma. Cancer Gene Ther 2023; 30:1330-1345. [PMID: 37420093 PMCID: PMC10581906 DOI: 10.1038/s41417-023-00640-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/19/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023]
Abstract
Therapy Induced Senescence (TIS) leads to sustained growth arrest of cancer cells. The associated cytostasis has been shown to be reversible and cells escaping senescence further enhance the aggressiveness of cancers. Chemicals specifically targeting senescent cells, so-called senolytics, constitute a promising avenue for improved cancer treatment in combination with targeted therapies. Understanding how cancer cells evade senescence is needed to optimise the clinical benefits of this therapeutic approach. Here we characterised the response of three different NRAS mutant melanoma cell lines to a combination of CDK4/6 and MEK inhibitors over 33 days. Transcriptomic data show that all cell lines trigger a senescence programme coupled with strong induction of interferons. Kinome profiling revealed the activation of Receptor Tyrosine Kinases (RTKs) and enriched downstream signaling of neurotrophin, ErbB and insulin pathways. Characterisation of the miRNA interactome associates miR-211-5p with resistant phenotypes. Finally, iCell-based integration of bulk and single-cell RNA-seq data identifies biological processes perturbed during senescence and predicts 90 new genes involved in its escape. Overall, our data associate insulin signaling with persistence of a senescent phenotype and suggest a new role for interferon gamma in senescence escape through the induction of EMT and the activation of ERK5 signaling.
Collapse
Affiliation(s)
- Vincent Gureghian
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Hailee Herbst
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Ines Kozar
- Laboratoire National de Santé, Dudelange, Luxembourg
| | | | | | - Gaia Ceddia
- Barcelona Supercomputing Center, 08034, Barcelona, Spain
| | - Cristian Angeli
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Christiane Margue
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Tijana Randic
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Demetra Philippidou
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Milène Tetsi Nomigni
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Ahmed Hemedan
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Leon-Charles Tranchevent
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Mark Bauer
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Apurva Badkas
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Anthoula Gaigneaux
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Arnaud Muller
- LuxGen, TMOH and Bioinformatics platform, Data Integration and Analysis unit, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Fabrice Tolle
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Nataša Pržulj
- Barcelona Supercomputing Center, 08034, Barcelona, Spain
- Department of Computer Science, University College London, London, WC1E 6BT, UK
- ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain
| | - Stephanie Kreis
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg.
| |
Collapse
|
9
|
Yang Y, Mihajlovic M, Masereeuw R. Protein-Bound Uremic Toxins in Senescence and Kidney Fibrosis. Biomedicines 2023; 11:2408. [PMID: 37760849 PMCID: PMC10525416 DOI: 10.3390/biomedicines11092408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic kidney disease (CKD) is a progressive condition of kidney dysfunction due to diverse causes of injury. In healthy kidneys, protein-bound uremic toxins (PBUTs) are cleared from the systemic circulation by proximal tubule cells through the concerted action of plasma membrane transporters that facilitate their urinary excretion, but the endogenous metabolites are hardly removed with kidney dysfunction and may contribute to CKD progression. Accumulating evidence suggests that senescence of kidney tubule cells influences kidney fibrosis, the common endpoint for CKD with an excessive accumulation of extracellular matrix (ECM). Senescence is a special state of cells characterized by permanent cell cycle arrest and limitation of proliferation, which promotes fibrosis by releasing senescence-associated secretory phenotype (SASP) factors. The accumulation of PBUTs in CKD causes oxidative stress and increases the production of inflammatory (SASP) factors that could trigger fibrosis. Recent studies gave some clues that PBUTs may also promote senescence in kidney tubular cells. This review provides an overview on how senescence contributes to CKD, the involvement of PBUTs in this process, and how kidney senescence can be studied. Finally, some suggestions for future therapeutic options for CKD while targeting senescence are given.
Collapse
Affiliation(s)
- Yi Yang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Milos Mihajlovic
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium;
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| |
Collapse
|
10
|
Knopp RC, Erickson MA, Rhea EM, Reed MJ, Banks WA. Cellular senescence and the blood-brain barrier: Implications for aging and age-related diseases. Exp Biol Med (Maywood) 2023; 248:399-411. [PMID: 37012666 PMCID: PMC10281623 DOI: 10.1177/15353702231157917] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
The blood-brain barrier (BBB) is a critical physiochemical interface that regulates communication between the brain and blood. It is comprised of brain endothelial cells which regulate the BBB's barrier and interface properties and is surrounded by supportive brain cell types including pericytes and astrocytes. Recent reports have suggested that the BBB undergoes dysfunction during normative aging and in disease. In this review, we consider the effect of cellular senescence, one of the nine hallmarks of aging, on the BBB. We first characterize known normative age-related changes at the BBB, and then evaluate changes in neurodegenerative diseases, with an emphasis on if/how cellular senescence is influencing these changes. We then discuss what insight has been gained from in vitro and in vivo studies of cellular senescence at the BBB. Finally, we evaluate mechanisms by which cellular senescence in peripheral pathologies can indirectly or directly affect BBB function.
Collapse
Affiliation(s)
- Rachel C Knopp
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Michelle A Erickson
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - May J Reed
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| |
Collapse
|
11
|
Han D, Gong H, Wei Y, Xu Y, Zhou X, Wang Z, Feng F. Hesperidin inhibits lung fibroblast senescence via IL-6/STAT3 signaling pathway to suppress pulmonary fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154680. [PMID: 36736168 DOI: 10.1016/j.phymed.2023.154680] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/19/2022] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and fatal lung disease with obscure pathogenesis. Increasing evidence suggests that cellular senescence is an important mechanism underlying in IPF. Clinical treatment with drugs, such as pirfenidone and nintedanib, reduces the risk of acute exacerbation and delays the decline of pulmonary function in patients with mild to moderate pulmonary fibrosis, and with adverse reactions. Hesperidin was previously shown to alleviate pulmonary fibrosis in rats by attenuating the inflammation response. Our previous research indicated that the Citrus alkaline extracts, hesperidin as the main active ingredient, could exert anti-pulmonary fibrosis effects by inhibiting the senescence of lung fibroblasts. However, whether hesperidin could ameliorate pulmonary fibrosis by inhibiting fibroblast senescence needed further study. PURPOSE This work aimed to investigate whether and how hesperidin can inhibit lung fibroblast senescence and thereby alleviate pulmonary fibrosis METHODS: Bleomycin was used to establish a mouse model of pulmonary fibrosis and doxorubicin was used to establish a model of cellular senescence in MRC-5 cells in vitro. The therapeutic effects of hesperidin on pulmonary fibrosis using haematoxylin-eosin staining, Masson staining, enzyme-linked immunosorbent assay, immunohistochemistry, western blotting and quantitative Real-Time PCR. The anti-senescent effect of hesperidin in vivo and in vitro was assessed by western blotting, quantitative Real-Time PCR and senescence-associated β-galactosidase RESULTS: We demonstrated that hesperidin could alleviate bleomycin-induced pulmonary fibrosis in mice. The expression level of senescence marker proteins p53, p21, and p16 was were downregulated, along with the myofibroblast marker α-SMA. The number of senescence-associated β-galactosidase-positive cells was significantly reduced by hesperidin intervention in vivo and in vitro. In addition, hesperidin could inhibit the IL6/STAT3 signaling pathway. Furthermore, suppression of the IL-6/STAT3 signaling pathway by pretreatment with the IL-6 inhibitor LMT-28 attenuating effect of hesperidin on fibroblast senescence in vitro. CONCLUSIONS These data illustrated that hesperidin may be potentially used in the treatment of IPF based on its ability to inhibit lung fibroblast senescence.
Collapse
Affiliation(s)
- Di Han
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Haiying Gong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China
| | - Yun Wei
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yong Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xianmei Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Zhichao Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Fanchao Feng
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
12
|
Xiong J, Cheung YK, Fong WP, Wong CTT, Ng DKP. Selective photodynamic eradication of senescent cells with a β-galactosidase-activated photosensitiser. Chem Commun (Camb) 2023; 59:3471-3474. [PMID: 36877479 DOI: 10.1039/d2cc06661k] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
A β-galactosidase-responsive photosensitiser has been designed and synthesised. It contains a galactosyl substrate, a boron dipyrromethene-based photosensitising unit and a black hole quencher 2 connected via an AB2-type self-immolative linker. This novel photosensitiser can be selectively activated by the senescence-associated β-galactosidase in senescent cells, leading to restoration in fluorescence emission and effective killing of the cells via photodynamic action.
Collapse
Affiliation(s)
- Junlong Xiong
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Ying-Kit Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Wing-Ping Fong
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Clarence T T Wong
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Dennis K P Ng
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| |
Collapse
|
13
|
Kciuk M, Gielecińska A, Mujwar S, Kołat D, Kałuzińska-Kołat Ż, Celik I, Kontek R. Doxorubicin-An Agent with Multiple Mechanisms of Anticancer Activity. Cells 2023; 12:659. [PMID: 36831326 PMCID: PMC9954613 DOI: 10.3390/cells12040659] [Citation(s) in RCA: 222] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Doxorubicin (DOX) constitutes the major constituent of anti-cancer treatment regimens currently in clinical use. However, the precise mechanisms of DOX's action are not fully understood. Emerging evidence points to the pleiotropic anticancer activity of DOX, including its contribution to DNA damage, reactive oxygen species (ROS) production, apoptosis, senescence, autophagy, ferroptosis, and pyroptosis induction, as well as its immunomodulatory role. This review aims to collect information on the anticancer mechanisms of DOX as well as its influence on anti-tumor immune response, providing a rationale behind the importance of DOX in modern cancer therapy.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Damian Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, 90-136 Lodz, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, 90-136 Lodz, Poland
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, 38039 Kayseri, Turkey
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| |
Collapse
|
14
|
Banerjee P, Gaddam N, Pandita TK, Chakraborty S. Cellular Senescence as a Brake or Accelerator for Oncogenic Transformation and Role in Lymphatic Metastasis. Int J Mol Sci 2023; 24:ijms24032877. [PMID: 36769195 PMCID: PMC9917379 DOI: 10.3390/ijms24032877] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Cellular senescence-the irreversible cell cycle arrest driven by a variety of mechanisms and, more specifically, the senescence-associated secretory phenotype (SASP)-is an important area of research in the context of different age-related diseases, such as cardiovascular disease and cancer. SASP factors play both beneficial and detrimental roles in age-related disease progression depending on the source of the SASPs, the target cells, and the microenvironment. The impact of senescence and the SASP on different cell types, the immune system, and the vascular system has been widely discussed. However, the impact of replicative or stress-induced senescence on lymphatic biology and pathological lymphangiogenesis remains underexplored. The lymphatic system plays a crucial role in the maintenance of body fluid homeostasis and immune surveillance. The perturbation of lymphatic function can hamper normal physiological function. Natural aging or stress-induced premature aging influences the lymphatic vessel structure and function, which significantly affect the role of lymphatics in tumor dissemination and metastasis. In this review, we focus on the role of senescence on lymphatic pathobiology, its impact on cancer, and potential therapeutic interventions to manipulate the aged or senescent lymphatic system for disease management.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Niyanshi Gaddam
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Tej K. Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX 77030, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, TX 77807, USA
- Correspondence: ; Tel.: +1-979-436-0697
| |
Collapse
|
15
|
Wahlmueller M, Narzt MS, Missfeldt K, Arminger V, Krasensky A, Lämmermann I, Schaedl B, Mairhofer M, Suessner S, Wolbank S, Priglinger E. Establishment of In Vitro Models by Stress-Induced Premature Senescence for Characterizing the Stromal Vascular Niche in Human Adipose Tissue. Life (Basel) 2022; 12:life12101459. [PMID: 36294893 PMCID: PMC9605485 DOI: 10.3390/life12101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
Acting as the largest energy reservoir in the body, adipose tissue is involved in longevity and progression of age-related metabolic dysfunction. Here, cellular senescence plays a central role in the generation of a pro-inflammatory environment and in the evolution of chronic diseases. Within the complexity of a tissue, identification and targeting of senescent cells is hampered by their heterogeneity. In this study, we generated stress-induced premature senescence 2D and 3D in vitro models for the stromal vascular niche of human adipose tissue. We established treatment conditions for senescence induction using Doxorubicin (Dox), starting from adipose-derived stromal/stem cells (ASCs), which we adapted to freshly isolated microtissue-stromal vascular fraction (MT-SVF), where cells are embedded within their native extracellular matrix. Senescence hallmarks for the established in vitro models were verified on different cellular levels, including morphology, cell cycle arrest, senescence-associated β-galactosidase activity (SA-βgal) and gene expression. Two subsequent exposures with 200 nM Dox for six days were suitable to induce senescence in our in vitro models. We demonstrated induction of senescence in the 2D in vitro models through SA-βgal activity, at the mRNA level (LMNB1, CDK1, p21) and additionally by G2/M phase cell cycle arrest in ASCs. Significant differences in Lamin B1 and p21 protein expression confirmed senescence in our MT-SVF 3D model. MT-SVF 3D cultures were composed of multiple cell types, including CD31, CD34 and CD68 positive cells, while cell death remained unaltered upon senescence induction. As heterogeneity and complexity of adipose tissue senescence is given by multiple cell types, our established senescence models that represent the perivascular niche embedded within its native extracellular matrix are highly relevant for future clinical studies.
Collapse
Affiliation(s)
- Marlene Wahlmueller
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
| | - Marie-Sophie Narzt
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
| | - Karin Missfeldt
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Verena Arminger
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Anna Krasensky
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Ingo Lämmermann
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Department of Biotechnology, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
- Rockfish Bio AG, 1010 Vienna, Austria
| | - Barbara Schaedl
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Mario Mairhofer
- Department of Hematology and Internal Oncology, Johannes Kepler University, 4020 Linz, Austria
| | - Susanne Suessner
- Austrian Red Cross Blood Transfusion Service for Upper Austria, 4020 Linz, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Eleni Priglinger
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
- Correspondence:
| |
Collapse
|
16
|
STING mediates nuclear PD-L1 targeting-induced senescence in cancer cells. Cell Death Dis 2022; 13:791. [PMID: 36109513 PMCID: PMC9477807 DOI: 10.1038/s41419-022-05217-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 01/21/2023]
Abstract
Immune checkpoint molecule programmed death-ligand 1 (PD-L1) is overexpressed in cancer cells and imparts resistance to cancer therapy. Although membrane PD-L1 has been targeted for cancer immune therapy, nuclear PD-L1 was reported to confer cancer resistance. Therefore, it is important to regulate the nuclear PD-L1. The mechanisms underlying the therapeutic efficacy of PD-L1 targeting have not been well-established. Cellular senescence has been considered a pivotal mechanism to prevent cancer progression, and recently, PD-L1 inhibition was shown to be involved in cancer cell senescence. However, the relevance of PD-L1 targeting-induced senescence and the role of stimulator of interferon genes (STING) has not been reported. Therefore, we aimed to identify the role of PD-L1 in cancer progression and how it regulates cancer prevention. In this study, we found that PD-L1 depletion-induced senescence via strong induction of STING expression in mouse melanoma B16-F10 and colon cancer CT26 cells, and in human melanoma A375 and lung cancer A549 cells. Interestingly, nuclear PD-L1 silencing increased STING promoter activity, implying that PD-L1 negatively regulates STING expression via transcriptional modulation. Furthermore, we showed that PD-L1 binds to the STING promoter region, indicating that PD-L1 directly controls STING expression to promote cancer growth. In addition, when we combined PD-L1 silencing with the senescence-inducing chemotherapeutic agent doxorubicin, the effect of PD-L1-targeting was even more powerful. Overall, our findings can contribute to the understanding of the role of PD-L1 in cancer therapy by elucidating a novel mechanism for PD-L1 targeting in cancer cells.
Collapse
|
17
|
Response to the letter by Dr. Renba Liang. Cancer Lett 2022. [PMID: 35597480 DOI: 10.1016/j.canlet.2022.215650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Bhat S, Adiga D, Shukla V, Guruprasad KP, Kabekkodu SP, Satyamoorthy K. Metastatic suppression by DOC2B is mediated by inhibition of epithelial-mesenchymal transition and induction of senescence. Cell Biol Toxicol 2022; 38:237-258. [PMID: 33758996 PMCID: PMC8986756 DOI: 10.1007/s10565-021-09598-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/24/2021] [Indexed: 02/04/2023]
Abstract
Senescence induction and epithelial-mesenchymal transition (EMT) events are the opposite sides of the spectrum of cancer phenotypes. The key molecules involved in these processes may get influenced or altered by genetic and epigenetic changes during tumor progression. Double C2-like domain beta (DOC2B), an intracellular vesicle trafficking protein of the double C2 protein family, plays a critical role in exocytosis, neurotransmitter release, and intracellular vesicle trafficking. DOC2B is repressed by DNA promoter hypermethylation and functions as a tumor growth regulator in cervical cancer. To date, the molecular mechanisms of DOC2B in cervical cancer progression and metastasis is elusive. Herein, the biological functions and molecular mechanisms regulated by DOC2B and its impact on senescence and EMT are described. DOC2B inhibition promotes proliferation, growth, and migration by relieving G0/G1-S arrest, actin remodeling, and anoikis resistance in Cal27 cells. It enhanced tumor growth and liver metastasis in nude mice with the concomitant increase in metastasis-associated CD55 and CD61 expression. Inhibition of EMT and promotion of senescence by DOC2B is a calcium-dependent process and accompanied by calcium-mediated interaction between DOC2B and CDH1. In addition, we have identified several EMT and senescence regulators as targets of DOC2B. We show that DOC2B may act as a metastatic suppressor by inhibiting EMT through induction of senescence via DOC2B-calcium-EMT-senescence axis.
Collapse
Affiliation(s)
- Samatha Bhat
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, 576104, India
| | - Vaibhav Shukla
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, 576104, India
| | - Kanive Parashiva Guruprasad
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, 576104, India.
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, 576104, India.
| |
Collapse
|
19
|
In Vitro Characterization of Doxorubicin-Mediated Stress-Induced Premature Senescence in Human Chondrocytes. Cells 2022; 11:cells11071106. [PMID: 35406671 PMCID: PMC8998002 DOI: 10.3390/cells11071106] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023] Open
Abstract
Accumulation of senescent chondrocytes is thought to drive inflammatory processes and subsequent cartilage degeneration in age-related as well as posttraumatic osteoarthritis (OA). However, the underlying mechanisms of senescence and consequences on cartilage homeostasis are not completely understood so far. Therefore, suitable in vitro models are needed to study chondrocyte senescence. In this study, we established and evaluated a doxorubicin (Doxo)-based model of stress-induced premature senescence (SIPS) in human articular chondrocytes (hAC). Cellular senescence was determined by the investigation of various senescence associated (SA) hallmarks including β-galactosidase activity, expression of p16, p21, and SA secretory phenotype (SASP) markers (IL-6, IL-8, MMP-13), the presence of urokinase-type plasminogen activator receptor (uPAR), and cell cycle arrest. After seven days, Doxo-treated hAC displayed a SIPS-like phenotype, characterized by excessive secretion of SASP factors, enhanced uPAR-positivity, decreased proliferation rate, and increased β-galactosidase activity. This phenotype was proven to be stable seven days after the removal of Doxo. Moreover, Doxo-treated hAC exhibited increased granularity and flattened or fibroblast-like morphology. Further analysis implies that Doxo-mediated SIPS was driven by oxidative stress as demonstrated by increased ROS levels and NO release. Overall, we provide novel insights into chondrocyte senescence and present a suitable in vitro model for further studies.
Collapse
|
20
|
Niklander SE, Lambert DW, Hunter KD. Senescent Cells in Cancer: Wanted or Unwanted Citizens. Cells 2021; 10:cells10123315. [PMID: 34943822 PMCID: PMC8699088 DOI: 10.3390/cells10123315] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 01/10/2023] Open
Abstract
Over recent decades, the field of cellular senescence has attracted considerable attention due to its association with aging, the development of age-related diseases and cancer. Senescent cells are unable to proliferate, as the pathways responsible for initiating the cell cycle are irreversibly inhibited. Nevertheless, senescent cells accumulate in tissues and develop a pro-inflammatory secretome, known as the senescence-associated secretory phenotype (SASP), which can have serious deleterious effects if not properly regulated. There is increasing evidence suggesting senescent cells contribute to different stages of carcinogenesis in different anatomical sites, mainly due to the paracrine effects of the SASP. Thus, a new therapeutic field, known as senotherapeutics, has developed. In this review, we aim to discuss the molecular mechanisms underlying the senescence response and its relationship with cancer development, focusing on the link between senescence-related inflammation and cancer. We will also discuss different approaches to target senescent cells that might be of use for cancer treatment.
Collapse
Affiliation(s)
- Sven E. Niklander
- Unidad de Patologia y Medicina Oral, Facultad de Odontologia, Universidad Andres Bello, Viña del Mar 2520000, Chile
- Correspondence: ; Tel.: +56-(32)2845108
| | - Daniel W. Lambert
- Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK; (D.W.L.); (K.D.H.)
- Healthy Lifespan Institute, University of Sheffield, Sheffield S10 2TN, UK
| | - Keith D. Hunter
- Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK; (D.W.L.); (K.D.H.)
- Oral Biology and Pathology, University of Pretoria, Pretoria 0028, South Africa
| |
Collapse
|
21
|
Chemotherapy: a double-edged sword in cancer treatment. Cancer Immunol Immunother 2021; 71:507-526. [PMID: 34355266 DOI: 10.1007/s00262-021-03013-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
Chemotherapy is a well-known and effective treatment for different cancers; unfortunately, it has not been as efficient in the eradication of all cancer cells as been expected. The mechanism of this failure was not fully clarified, yet. Meanwhile, alterations in the physiologic conditions of the tumor microenvironment (TME) were suggested as one of the underlying possibilities. Chemotherapy drugs can activate multiple signaling pathways and augment the secretion of inflammatory mediators. Inflammation may show two opposite roles in the TME. On the one hand, inflammation, as an innate immune response, tries to suppress tumor growth but on the other hand, it might be not powerful enough to eradicate the cancer cells and even it can provide appropriate conditions for cancer promotion and relapse as well. Therefore, the administration of mild anti-inflammatory drugs during chemotherapy might result in more successful clinical results. Here, we will review and discuss this hypothesis. Most chemotherapy agents are triggers of inflammation in the tumor microenvironment through inducing the production of senescence-associated secretory phenotype (SASP) molecules. Some chemotherapy agents can induce systematic inflammation by provoking TLR4 signaling or triggering IL-1B secretion through the inflammasome pathway. NF-kB and MAPK are key signaling pathways of inflammation and could be activated by several chemotherapy drugs. Furthermore, inflammation can play a key role in cancer development, metastasis and exacerbation.
Collapse
|
22
|
Lee SY, Park KH, Lee G, Kim SJ, Song WH, Kwon SH, Koh JT, Huh YH, Ryu JH. Hypoxia-inducible factor-2α mediates senescence-associated intrinsic mechanisms of age-related bone loss. Exp Mol Med 2021; 53:591-604. [PMID: 33811248 PMCID: PMC8102580 DOI: 10.1038/s12276-021-00594-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 02/01/2023] Open
Abstract
Aging is associated with cellular senescence followed by bone loss leading to bone fragility in humans. However, the regulators associated with cellular senescence in aged bones need to be identified. Hypoxia-inducible factor (HIF)-2α regulates bone remodeling via the differentiation of osteoblasts and osteoclasts. Here, we report that HIF-2α expression was highly upregulated in aged bones. HIF-2α depletion in male mice reversed age-induced bone loss, as evidenced by an increase in the number of osteoblasts and a decrease in the number of osteoclasts. In an in vitro model of doxorubicin-mediated senescence, the expression of Hif-2α and p21, a senescence marker gene, was enhanced, and osteoblastic differentiation of primary mouse calvarial preosteoblast cells was inhibited. Inhibition of senescence-induced upregulation of HIF-2α expression during matrix maturation, but not during the proliferation stage of osteoblast differentiation, reversed the age-related decrease in Runx2 and Ocn expression. However, HIF-2α knockdown did not affect p21 expression or senescence progression, indicating that HIF-2α expression upregulation in senescent osteoblasts may be a result of aging rather than a cause of cellular senescence. Osteoclasts are known to induce a senescent phenotype during in vitro osteoclastogenesis. Consistent with increased HIF-2α expression, the expression of p16 and p21 was upregulated during osteoclastogenesis of bone marrow macrophages. ChIP following overexpression or knockdown of HIF-2α using adenovirus revealed that p16 and p21 are direct targets of HIF-2α in osteoclasts. Osteoblast-specific (Hif-2αfl/fl;Col1a1-Cre) or osteoclast-specific (Hif-2αfl/fl;Ctsk-Cre) conditional knockout of HIF-2α in male mice reversed age-related bone loss. Collectively, our results suggest that HIF-2α acts as a senescence-related intrinsic factor in age-related dysfunction of bone homeostasis.
Collapse
Affiliation(s)
- Sun Young Lee
- grid.14005.300000 0001 0356 9399Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Ka Hyon Park
- grid.14005.300000 0001 0356 9399Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Gyuseok Lee
- grid.14005.300000 0001 0356 9399Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Su-Jin Kim
- grid.14005.300000 0001 0356 9399Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea ,grid.14005.300000 0001 0356 9399Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Won-Hyun Song
- grid.14005.300000 0001 0356 9399Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea ,grid.14005.300000 0001 0356 9399Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Seung-Hee Kwon
- grid.14005.300000 0001 0356 9399Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea ,grid.14005.300000 0001 0356 9399Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Jeong-Tae Koh
- grid.14005.300000 0001 0356 9399Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea ,grid.14005.300000 0001 0356 9399Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Yun Hyun Huh
- grid.61221.360000 0001 1033 9831School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Republic of Korea
| | - Je-Hwang Ryu
- grid.14005.300000 0001 0356 9399Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea ,grid.14005.300000 0001 0356 9399Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186 Republic of Korea
| |
Collapse
|
23
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Rahmani Moghadam E, Raei M, Kalantari M, Tavakol S, Mohammadinejad R, Najafi M, Tay FR, Makvandi P. Progress in Natural Compounds/siRNA Co-delivery Employing Nanovehicles for Cancer Therapy. ACS COMBINATORIAL SCIENCE 2020; 22:669-700. [PMID: 33095554 PMCID: PMC8015217 DOI: 10.1021/acscombsci.0c00099] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Chemotherapy using natural compounds, such as resveratrol, curcumin, paclitaxel, docetaxel, etoposide, doxorubicin, and camptothecin, is of importance in cancer therapy because of the outstanding therapeutic activity and multitargeting capability of these compounds. However, poor solubility and bioavailability of natural compounds have limited their efficacy in cancer therapy. To circumvent this hurdle, nanocarriers have been designed to improve the antitumor activity of the aforementioned compounds. Nevertheless, cancer treatment is still a challenge, demanding novel strategies. It is well-known that a combination of natural products and gene therapy is advantageous over monotherapy. Delivery of multiple therapeutic agents/small interfering RNA (siRNA) as a potent gene-editing tool in cancer therapy can maximize the synergistic effects against tumor cells. In the present review, co-delivery of natural compounds/siRNA using nanovehicles are highlighted to provide a backdrop for future research.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty
of Engineering and Natural Sciences, Sabanci
University, Orta Mahalle,
Üniversite Caddesi No. 27, Orhanlı,
Tuzla, 34956 Istanbul, Turkey
- Sabanci
University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul Turkey
| | - Ali Zarrabi
- Sabanci
University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul Turkey
| | - Kiavash Hushmandi
- Department
of Food Hygiene and Quality Control, Division of Epidemiology &
Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran
| | - Farid Hashemi
- Department
of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department
of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Mehdi Raei
- Health Research
Center, Life Style Institute, Baqiyatallah
University of Medical Sciences, Tehran 1435916471, Iran
| | - Mahshad Kalantari
- Department
of Genetics, Tehran Medical Sciences Branch, Azad University, Tehran 19168931813, Iran
| | - Shima Tavakol
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 1449614525, Iran
| | - Reza Mohammadinejad
- Pharmaceutics
Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Masoud Najafi
- Medical
Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Radiology
and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Franklin R. Tay
- College
of Graduate Studies, Augusta University, Augusta, Georgia 30912, United States
| | - Pooyan Makvandi
- Istituto
Italiano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa Italy
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, 14496-14535 Tehran, Iran
| |
Collapse
|
24
|
Therapy-induced polyploidization and senescence: Coincidence or interconnection? Semin Cancer Biol 2020; 81:83-95. [DOI: 10.1016/j.semcancer.2020.11.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
|
25
|
Hałas-Wiśniewska M, Izdebska M, Zielińska W, Grzanka A. The effect of low doses of doxorubicin on the rat glioma C6 cells in the context of the proteins involved in intercellular interactions. Acta Histochem 2020; 122:151625. [PMID: 33066845 DOI: 10.1016/j.acthis.2020.151625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/01/2020] [Accepted: 09/01/2020] [Indexed: 11/24/2022]
Abstract
The aim of this investigation was to determine the effect of doxorubicin on F-actin rearrangement and β-catenin and cofilin-1 in a rat glioma C6 cell line in combination with changes in their morphology and ultrastructure. The experimental material constituted rat glioma C6 cell line. The cells were incubated with sublethal doses of doxorubicin in the concentration of 50, 100 and 200 nM. The blue trypan dye method was used to determine the number of dead cells. Morphological and ultrastructural changes in the cells were evaluated using light and transmission electron microscope, respectively. In order to determine the rearrangements and level of expression of F-actin, β-catenin and cofilin-1 they were analyzed using a fluorecence microscope. In turn, cell death and cell cycle were evaluated by Guava 6HT-2 L Cytometer. The performed experiments showed a dose-dependent decrease in the survival of C6 cells after treatment with doxorubicin. The analysis of cell death showed a dose-dependent increase in the population of apoptotic and necrotic cells. These results were confirmed by microscopy observation. The changes in morphology, ultrastructure, and rearrangements of F-actin, β-catenin and cofilin-1 were also observed. The results obtained in the study showed that sublethal concentrations of doxorubicin influenced the structure of F-actin and other proteins involved in cell-cell interactions. Moreover, mitotic catastrophe may preceding apoptosis, what suggest the cytotoxic effect of low dose of doxorubicin. Furthermore, our results confirmed the multi-dimensional mechanism of DOX action in tumor cells.
Collapse
|
26
|
Mohamad Kamal NS, Safuan S, Shamsuddin S, Foroozandeh P. Aging of the cells: Insight into cellular senescence and detection Methods. Eur J Cell Biol 2020; 99:151108. [PMID: 32800277 DOI: 10.1016/j.ejcb.2020.151108] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/10/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular theory of aging states that human aging is the result of cellular aging, in which an increasing proportion of cells reach senescence. Senescence, from the Latin word senex, means "growing old," is an irreversible growth arrest which occurs in response to damaging stimuli, such as DNA damage, telomere shortening, telomere dysfunction and oncogenic stress leading to suppression of potentially dysfunctional, transformed, or aged cells. Cellular senescence is characterized by irreversible cell cycle arrest, flattened and enlarged morphology, resistance to apoptosis, alteration in gene expression and chromatin structure, expression of senescence associated- β-galactosidase (SA-β-gal) and acquisition of senescence associated secretory phenotype (SASP). In this review paper, different types of cellular senescence including replicative senescence (RS) which occurs due to telomere shortening and stress induced premature senescence (SIPS) which occurs in response to different types of stress in cells, are discussed. Biomarkers of cellular senescence and senescent assays including BrdU incorporation assay, senescence associated- β-galactosidase (SA-β-gal) and senescence-associated heterochromatin foci assays to detect senescent cells are also addressed.
Collapse
Affiliation(s)
- Nor Shaheera Mohamad Kamal
- School of Health Sciences, Universiti Sains Malaysia Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Sabreena Safuan
- School of Health Sciences, Universiti Sains Malaysia Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia; USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Georgetown, Penang, Malaysia
| | - Parisa Foroozandeh
- USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800 Georgetown, Penang, Malaysia.
| |
Collapse
|