1
|
Zeng Y, Wang X, Hu J, Tian C, Liu R, Chen X, Huang L, Liang X, Wang X, Fan H, Zhang H, Lu Y. LZTS3 represses tumorigenesis and radioresistance via CK1δ and β-TrCP-mediated ubiquitination pathway in lung cancer. Cell Signal 2025; 129:111655. [PMID: 39956246 DOI: 10.1016/j.cellsig.2025.111655] [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: 11/17/2024] [Revised: 01/29/2025] [Accepted: 02/09/2025] [Indexed: 02/18/2025]
Abstract
Radioresistance is one of the main causes for local treatment failure in lung cancer. Nevertheless, the potential mechanisms of radioresistance in lung cancer have not been elucidated completely. Here, we discover a carcinoma-inhibiting protein called leucine zipper tumor suppressor 3 (LZTS3), which is low-expressed and related to adverse outcome in lung cancer. Moreover, our studies demonstrate that LZTS3 restrains cell proliferation and radioresistance in vitro and in vivo. Mechanistically, protein kinase CK1δ interacts with LZTS3, resulting in E3 ubiquitin ligase β-TrCP recognizes and binds to LZTS3. Thus, LZTS3 is degraded by the ubiquitin-proteasome pathway. We also identify two conserved degrons (DSGRNS and DSGRAS) are essential for the ubiquitinated degradation of LZTS3 by CK1δ and β-TrCP. More importantly, we detect that the CK1δ and β-TrCP-mediated degradation of LZTS3 facilitate the cell growth, proliferation and radioresistance in lung cancer. Collectivelly, our results suggest that LZTS3 regulates tumorigenesis and radioresistance in lung cancer depend on a CK1δ and β-TrCP-mediated ubiquitin-proteasome pathway. LZTS3 may be a new molecular target for lung cancer treatment.
Collapse
Affiliation(s)
- Yulan Zeng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology,Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiao Wang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Department of medical oncology, Sir run run shaw hospital, School of medicine, Zhejiang university, China
| | - Ji Hu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Tian
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruiqi Liu
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Department of Pathology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiaoyan Chen
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luanluan Huang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaodong Liang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xian Wang
- Department of medical oncology, Sir run run shaw hospital, School of medicine, Zhejiang university, China
| | - Hongjie Fan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hu Bei 430022, China.
| | - Haibo Zhang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Yanwei Lu
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Hu H, Zhang G, Chen T, Liu Y, Meng L, Holmdahl R, Dai L, Zhao Y. Immunosenescence in autoimmune diseases. Autoimmun Rev 2025; 24:103805. [PMID: 40132774 DOI: 10.1016/j.autrev.2025.103805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Autoimmune diseases (AIDs) are a group of disorders in which the immune system mistakenly attacks the body's own tissues, characterized by the loss of tolerance to self-antigens and destruction of tissues. Aging is a natural process of physiological decline that also alters the immune system, a condition known as immunosenescence. During immunosenescence, the immune system undergoes various changes, including modifications and antigenicity of self-antigens, abnormalities in the quantity, phenotype, and function of lymphocytes and antibodies, as well as a narrowing of the B and T cell receptor repertoire, changes that may increase susceptibility to AIDs. Additionally, senescent immune cells and the senescence-associated secretory phenotype (SASP) contribute to target organ involvement in AIDs, exacerbating chronic inflammation and tissue damage. Mitochondrial dysfunction and metabolic imbalances in AIDs lead to the accumulation of senescent cells, which act as upstream drivers of immunosenescence. In this review, we summarize the bidirectional relationship between AIDs and immunosenescence, as well as its potential mechanisms. Therapeutic approaches targeting immunosenescence in AIDs remain at an early stage. Strategies aimed at resetting or reversing the aging immune system are expected to become a novel direction in the future.
Collapse
Affiliation(s)
- Huifang Hu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Guangyue Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Tao Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Liesu Meng
- Department of Rheumatology, and National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Rikard Holmdahl
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China.
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Gai S, Cao P, Zhong X, Lin Y, Lin B, Jiang M. Designing an anticancer Pd(II) complex as poly(ADP-ribose) polymerase 1 inhibitor. Int J Biol Macromol 2025; 297:139885. [PMID: 39818402 DOI: 10.1016/j.ijbiomac.2025.139885] [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: 09/23/2024] [Revised: 01/01/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Targeting DNA repair mechanisms, particularly PARP-1 inhibition, has emerged as a promising strategy for developing anticancer therapies. we designed and synthesized two 2-thiazolecarboxaldehyde thiosemicarbazone palladium(II) complexes (C1 and C2), and evaluated their anti-cancer activities. These Pd(II) complexes exhibited potent PARP-1 enzyme inhibition and demonstrated considerable antiproliferative activity against various cancer cell lines. In vivo studies using the A549 tumor xenograft model revealed that C2 effectively suppressed tumor growth and exhibited minimal systemic toxicity. Mechanistically, C2 induced A549 cell death through multiple pathways: cell cycle arrest, elevated intracellular reactive oxygen species (ROS) levels, DNA damage induction, exacerbated DNA double-strand breakage via PARP-1 inhibition, mitochondrial membrane potential reduction, and ultimately apoptosis. These findings provide a new design strategy for developing safe and highly effective PARP-1 inhibitors.
Collapse
Affiliation(s)
- Shuangshuang Gai
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Peng Cao
- School of Food and Health, Guilin Tourism University, Guilin, Guangxi 541006, China
| | - Xuwei Zhong
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - YiCan Lin
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Benxin Lin
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Ming Jiang
- School of Biological and Food Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China.
| |
Collapse
|
4
|
González I, Maldonado-Agurto R. The role of cellular senescence in endothelial dysfunction and vascular remodelling in arteriovenous fistula maturation. J Physiol 2025. [PMID: 39977444 DOI: 10.1113/jp287387] [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: 08/06/2024] [Accepted: 01/29/2025] [Indexed: 02/22/2025] Open
Abstract
Haemodialysis (HD) is often required for patients with end-stage renal disease. Arteriovenous fistulas (AVFs), a surgical procedure connecting an artery to a vein, are the preferred vascular access for HD due to their durability and lower complication rates. The aim of AVFs is to promote vein remodelling to accommodate increased blood flow needed for dialysis. However, many AVFs fail to mature properly, making them unsuitable for dialysis. Successful maturation requires remodelling, resulting in an increased luminal diameter and thickened walls to support the increased blood flow. After AVF creation, haemodynamic changes due to increased blood flow on the venous side of the AVF initiate a cascade of events that, when successful, lead to the proper maturation of the AVF, making it suitable for cannulation. In this process, endothelial cells play a crucial role since they are in direct contact with the frictional forces exerted by the blood, known as shear stress. Patients requiring HD often have other conditions that increase the burden of senescent cells, such as ageing, diabetes and hypertension. These senescent cells are characterized by irreversible growth arrest and the secretion of pro-inflammatory and pro-thrombotic factors, collectively known as the senescence-associated secretory phenotype (SASP). This accumulation can impair vascular function by promoting inflammation, reducing vasodilatation, and increasing thrombosis risk, thus hindering proper AVF maturation and function. This review explores the contribution of senescent endothelial cells to AVF maturation and explores potential therapeutic strategies to alleviate the effects of senescent cell accumulation, aiming to improve AVF maturation rates.
Collapse
Affiliation(s)
- Ignacia González
- Center for Biomedical Research (CIBMED), Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Rodrigo Maldonado-Agurto
- Center for Biomedical Research (CIBMED), Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| |
Collapse
|
5
|
Zhai Z, Li H. Identification of CircRNAs that promote cancer and their potential contribution to hepatocellular carcinoma (HCC) pathogenesis. Clin Exp Med 2025; 25:60. [PMID: 39961942 PMCID: PMC11832687 DOI: 10.1007/s10238-025-01585-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/02/2025] [Indexed: 02/20/2025]
Abstract
The critical involvement of circRNAs in tumour progression and development is becoming increasingly evident. This study aimed to identify novel cancer-promoting circRNAs and explore their potential contribution to the pathogenesis of hepatocellular carcinoma (HCC). Expression profiles of circRNAs, miRNAs, and mRNAs associated with HCC were predicted through interaction analysis using data from the GEO and TCGA databases. A circRNA-miRNA-mRNA network was constructed, and the biological functions of the target mRNAs were predicted via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. A protein-protein interaction (PPI) network was generated to identify important hub genes. Weighted Gene Co-expression Network Analysis (WGCNA) was applied to determine the key modules related to cancer-promoting circRNAs. OncomiR and GEPIA were used to investigate the correlation between miRNAs, mRNAs, and clinicopathological features, while TIMER was utilized to explore the relationship between gene expression and immune cell infiltration. A network of 18 cancer-promoting circRNAs in HCC was identified, which enhanced the expression of 141 downstream mRNAs through competitive binding with 10 miRNAs. GO, KEGG, and PPI network analyses revealed that E2F1, H2AFX, TOP2A, and RAD51 are key hub genes within the competitive endogenous RNA (ceRNA) network, primarily involved in cell cycle regulation, cancer-related pathways, and angiogenesis. WGCNA identified the "HCC DUcircRNA Module". Moreover, these core genes and key modules were closely associated with pathological stage, patient survival, and B-cell immune infiltration. We constructed a ceRNA network related to cancer-promoting circRNAs. The genes and key modules involved in this network may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Zhensheng Zhai
- Department of General Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, NO.99 Longcheng Road, Xiaodian District, Taiyuan, 030032, Shanxi, People's Republic of China
| | - Huiyu Li
- Department of General Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, NO.99 Longcheng Road, Xiaodian District, Taiyuan, 030032, Shanxi, People's Republic of China.
| |
Collapse
|
6
|
Qin Y, Liu H, Wu H. Cellular Senescence in Health, Disease, and Lens Aging. Pharmaceuticals (Basel) 2025; 18:244. [PMID: 40006057 PMCID: PMC11859104 DOI: 10.3390/ph18020244] [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: 01/13/2025] [Revised: 02/04/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Cellular senescence is a state of irreversible cell cycle arrest that serves as a critical regulator of tissue homeostasis, aging, and disease. While transient senescence contributes to development, wound healing, and tumor suppression, chronic senescence drives inflammation, tissue dysfunction, and age-related pathologies, including cataracts. Lens epithelial cells (LECs), essential for maintaining lens transparency, are particularly vulnerable to oxidative stress-induced senescence, which accelerates lens aging and cataract formation. This review examines the dual role of senescence in LEC function and its implications for age-related cataractogenesis, alongside emerging senotherapeutic interventions. Methods: This review synthesizes findings on the molecular mechanisms of senescence, focusing on oxidative stress, mitochondrial dysfunction, and the senescence-associated secretory phenotype (SASP). It explores evidence linking LEC senescence to cataract formation, highlighting key studies on stress responses, DNA damage, and antioxidant defense. Recent advances in senotherapeutics, including senolytics and senomorphics, are analyzed for their potential to mitigate LEC senescence and delay cataract progression. Conclusions: LEC senescence is driven by oxidative damage, mitochondrial dysfunction, and impaired redox homeostasis. These factors activate senescence path-ways, including p53/p21 and p16/Rb, resulting in cell cycle arrest and SASP-mediated inflammation. The accumulation of senescent LECs reduces regenerative capacity, disrupts lens homeostasis, and contributes to cataractogenesis. Emerging senotherapeutics, such as dasatinib, quercetin, and metformin, show promise in reducing the senescent cell burden and modulating the SASP to preserve lens transparency.
Collapse
Affiliation(s)
- Ying Qin
- Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (Y.Q.); (H.L.)
| | - Haoxin Liu
- Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (Y.Q.); (H.L.)
| | - Hongli Wu
- Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (Y.Q.); (H.L.)
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
7
|
Wang PH, Wang Y, Guo YY, Ma ZH, Wu C, Xing L. Ibuprofen modulates macrophage polarization by downregulating poly (ADP-ribose) polymerase 1. Int Immunopharmacol 2024; 143:113502. [PMID: 39488918 DOI: 10.1016/j.intimp.2024.113502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Ibuprofen, a non-steroidal drug, is well known for its anti-inflammatory activity. The effects of ibuprofen on the polarization of macrophages are still not clear. Herein, we used THP-1 monocyte-derived macrophages to find that ibuprofen has inhibitory effects on the polarization of both classically activated M1 macrophages and alternatively activated M2 macrophages by downregulating NF-κB and JAK/STAT signaling pathways. During M1 or M2 polarization, ibuprofen also downregulated the expression of poly (ADP-ribose) polymerase 1 (PARP1). Furthermore, knockdown of PARP1 by either small interfering RNA or PARP1 inhibitor PJ34 can exert inhibitory effects on the polarization of M1 and M2, and alter the immune response of macrophages to the infection of Mycobacterium tuberculosis H37Ra. The results demonstrate that PARP1 plays a regulatory role in the ibuprofen-modulated polarization of macrophage, revealing the interplay between the DNA repair response process and macrophage polarization.
Collapse
Affiliation(s)
- Pei-Hua Wang
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China
| | - Yan Wang
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China
| | - Yan-Yan Guo
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China
| | - Zi-Hui Ma
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China; Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China
| | - Li Xing
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan 030006, Shanxi Province, China; Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China.
| |
Collapse
|
8
|
Ben Diouf O, Gilbert A, Bernay B, Syljuåsen RG, Tudor M, Temelie M, Savu DI, Soumboundou M, Sall C, Chevalier F. Phospho-Proteomics Analysis of Early Response to X-Ray Irradiation Reveals Molecular Mechanism Potentially Related to U251 Cell Radioresistance. Proteomes 2024; 13:1. [PMID: 39846632 PMCID: PMC11755531 DOI: 10.3390/proteomes13010001] [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: 08/31/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Glioblastoma (GBM) is a devastating malignant brain tumor with a poor prognosis. GBM is associated with radioresistance. Post-translational modifications (PTMs) such as protein phosphorylation can play an important role in the cellular response to radiation. To better understand the early cellular activities after radiation in GBM, we carried out a phospho-proteomic study on the U251 cell line 3 h after X-ray irradiation (6Gy) and on non-irradiated cells. Our study showed a strong modification of proteoform phosphorylation in response to radiation. We found 453 differentially expressed phosphopeptides (DEPs), with 211 being upregulated and 242 being downregulated. A GO enrichment analysis of DEPs showed a strong enrichment of the signaling pathways involved in DNA damage response after irradiation and categorized them into biological processes (BPs), cellular components (CCs) and molecular functions (MFs). Certain accessions such as BRCA1, MDC1, H2AX, MDC1, TP53BP1 were dynamically altered in our fraction and are highly associated with the signaling pathways enriched after radiation.
Collapse
Affiliation(s)
- Ousseynou Ben Diouf
- Mixed Research Exploration and Diagnosis (UMRED), UFR-Healthy, Iba Der THIAM University of Thies, Thies BP A967, Senegal; (O.B.D.)
| | - Antoine Gilbert
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN, Université de Caen Normandie, 14000 Caen, France
| | - Benoit Bernay
- Proteogen Platform, US EMerode, CAEN Normandie University, 14032 Caen, France
| | - Randi G. Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Mihaela Tudor
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania (D.I.S.)
| | - Mihaela Temelie
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania (D.I.S.)
| | - Diana I. Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania (D.I.S.)
| | - Mamadou Soumboundou
- Mixed Research Exploration and Diagnosis (UMRED), UFR-Healthy, Iba Der THIAM University of Thies, Thies BP A967, Senegal; (O.B.D.)
| | - Cheikh Sall
- Mixed Research Exploration and Diagnosis (UMRED), UFR-Healthy, Iba Der THIAM University of Thies, Thies BP A967, Senegal; (O.B.D.)
| | - François Chevalier
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN, Université de Caen Normandie, 14000 Caen, France
| |
Collapse
|
9
|
Kim YS, Lupatov AY, Burunova VV, Bagmet NN, Chardarov NK, Malov SL, Kholodenko RV, Shatverian GA, Manukyan GV, Yarygin KN, Kholodenko IV. Human Liver MSCs Retain Their Basic Cellular Properties in Chronically Inflamed Liver Tissue. Int J Mol Sci 2024; 25:13374. [PMID: 39769138 PMCID: PMC11676302 DOI: 10.3390/ijms252413374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Every 25th death worldwide is associated with liver pathology. The development of novel approaches to liver diseases therapy and protocols for maintaining the vital functions of patients on the liver transplant waiting list are urgently needed. Resident mesenchymal stem cells (MSCs) play a significant role in supporting liver tissue integrity and improve the liver condition after infusion. However, it remains unclear whether MSCs isolated from chronically inflamed livers are similar in their basic cellular properties to MSCs obtained from healthy livers. We applied a large array of tests to compare resident MSCs isolated from apparently normal liver tissue and from chronically inflamed livers of patients with fibrosis, cirrhosis, and viral hepatitis. Chronic inflammatory environment did not alter the major cellular characteristics of MSCs, including the expression of MSC markers, stem cell markers, adhesion molecules, and the hallmarks of senescence, as well as cell proliferation, migration, and secretome. Only the expression of some immune checkpoints and toll-like receptors was different. Evidently, MSCs with unchanged cellular properties are present in human liver even at late stages of inflammatory diseases. These cells can be isolated and used as starting material in the development of cell therapies of liver diseases.
Collapse
Affiliation(s)
- Yan S. Kim
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Alexey Yu. Lupatov
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Veronika V. Burunova
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Nikolay N. Bagmet
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Nikita K. Chardarov
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Svyatoslav L. Malov
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Roman V. Kholodenko
- Laboratory of Molecular Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Garnik A. Shatverian
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Garik V. Manukyan
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
- Department of General Pathology and Pathophysiology, Russian Medical Academy of Continuous Professional Education, 125284 Moscow, Russia
| | - Irina V. Kholodenko
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| |
Collapse
|
10
|
Nam D, Park J, Lee J, Son J, Kim JE. mTOR potentiates senescent phenotypes and primary cilia formation after cisplatin-induced G2 arrest in retinal pigment epithelial cells. Cell Signal 2024; 124:111402. [PMID: 39251051 DOI: 10.1016/j.cellsig.2024.111402] [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: 02/27/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Cisplatin, a platinum-based anticancer drug, is used to treat several types of cancer. Despite its effectiveness, cisplatin-induced side effects have often been reported. Although cisplatin-induced toxicities, such as apoptosis and/or necrosis, have been well studied, the fate of cells after exposure to sublethal doses of cisplatin needs further elucidation. Treatment with a sublethal dose of cisplatin induced cell cycle arrest at the G2 phase in retinal pigment epithelial cells. Following cisplatin withdrawal, the cells irreversibly exited the cell cycle and became senescent. Notably, the progression from the G2 to the G1 phase occurred without mitotic entry, a phenomenon referred to as mitotic bypass, resulting in the accumulation of cells containing 4N DNA content. Cisplatin-exposed cells exhibited morphological changes associated with senescence, including an enlarged size of cell and nucleus and increased granularity. In addition, the senescent cells possessed primary cilia and persistent DNA lesions. Senescence induced by transient exposure to cisplatin involves mTOR activation. Although transient co-exposure with an mTORC1 inhibitor rapamycin did not prevent mitotic bypass and entry into senescence, it delayed the progression of senescence and attenuated senescent phenotypes, resulting in shorter primary cilia formation. Conclusively, cisplatin induces senescence in retinal pigment epithelial cells by promoting mTOR activation.
Collapse
Affiliation(s)
- Dajeong Nam
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaejung Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaehong Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juyoung Son
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
11
|
Pu C, Liu Y, Lan S, Fan H, Liu L, Liu J, Guo Y. Enhancing therapeutic efficacy in homologous recombination-proficient pancreatic cancer via the combination of PARP1-PROTAC and a BRD4 inhibitor. Bioorg Med Chem 2024; 115:117970. [PMID: 39476572 DOI: 10.1016/j.bmc.2024.117970] [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: 09/10/2024] [Revised: 10/10/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Currently, poly (ADP-ribose) polymerase inhibitors (PARPi) have been approved by U.S. Food and Drug Administration for BRCA-mutated pancreatic cancer therapy. However, limited indications hinder their further application. Repression of bromodomain-containing protein 4 (BRD4) can block the homologous recombination (HR) repair pathway and has the potential to enhance the response to PARPi in HR-proficient pancreatic cancer therapy. In addition, proteolysis targeting chimeras (PROTACs) can hijack E3 ligase within the cell to ubiquitinate degradation-targeted proteins effectively and quickly, thus enhancing the therapeutic effect on tumors. In the present study, the LB23 compound, which induces PARP1 degradation, was employed in combination with the BRD4 inhibitor JQ1, confirming their synergistic effect in HR-proficient pancreatic cancer through various methods. Moreover, compared to the JQ1 and PARPi olaparib combination, PARP1-PROTAC and JQ1 had more notable synergistic effects. Further research into the synergistic mechanism demonstrated that combination therapy enhanced DNA damage and suppressed DNA repair by inducing cell cycle arrest and cell apoptosis. The present study therefore provides the experimental data for this type of combination therapy, which is expected to be an innovative approach for the treatment of HR-proficient pancreatic cancer.
Collapse
Affiliation(s)
- Chunlan Pu
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610031, China
| | - Yuanyuan Liu
- Sichuan Technical Inspection Center for Medical Products, Sichuan Technical Inspection Center for Vaccine, Chengdu, Sichuan 610015, China
| | - Suke Lan
- College of Chemistry & Environment Protection Engineering, Southwest Minzu University, Chengdu, Sichuan 610041, China
| | - Hengrui Fan
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610031, China
| | - Lvye Liu
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610031, China
| | - Jianyu Liu
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610031, China.
| | - Yuanbiao Guo
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610031, China.
| |
Collapse
|
12
|
Wuri L, Zarutskie PW, Arosh JA, Banu SK. Employment of a Newly Defined In Vitro Fertilization Protocol to Determine the Cytoskeletal Machinery, DNA Damage, and Subsequent DNA Repair Resulting from Endocrine Disruption by Hexavalent Chromium in Rat Metaphase II Oocytes. Curr Protoc 2024; 4:e70060. [PMID: 39711520 DOI: 10.1002/cpz1.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
These protocols describe a detailed method to determine the DNA damage and F-actin and microtubule defects of metaphase II oocytes caused by hexavalent chromium, Cr(VI), an endocrine disrupting chemical (EDC). The protocol provides systematic steps to determine protein expression encoded by pluripotency proteins such as Oct4, Nanog, and Cdx2 during early embryonic development. Occupational or environmental exposure to EDCs has significantly increased infertility in both men and women. The urinary concentration of the EDC bisphenol A in patients undergoing in vitro fertilization (IVF) is directly related to decreased implantation rates and the number of metaphase II oocytes recovered. This protocol outlines crucial steps in assessing the structure of F-actin and microtubules, DNA damage, and repair mechanisms in metaphase II oocytes as well as pluripotency protein markers of early-stage embryos. IVF techniques to achieve fertility goals in both humans and animals are of paramount importance. The interplay between F-actin and microtubules is crucial for bipolar spindle assembly and correct partitioning of the nuclear genome in mammalian oocyte meiosis. EDCs induce DNA damage and impair DNA repair mechanisms, compromising oocyte quality. In human IVF, this results in failure to implant, early miscarriage, and live births with congenital disorders, thus decreasing success rates and increasing poor outcomes. The application of IVF protocols in rats to understand EDC-mediated defects in the cytoskeletal network of metaphase II oocytes is not well established. We present a newly defined rat IVF protocol and demonstrate outcomes using these protocols to determine the adverse effects of Cr(VI) on metaphase II oocytes. Basic Protocol 1 includes steps to superovulate rats, dissect ampullae, retrieve oocytes/eggs, perform immunofluorescence staining of cytoskeletal machinery (microtubules and F-actin), and assess expression of the DNA double-strand break marker γ-H2AX and the DNA repair protein RAD51 in control and Cr(VI)-exposed rats. Basic Protocol 2 describes methods for detecting the pluripotency proteins Oct4, Nanog, and Cdx2 during early embryonic development in control rats. © 2024 Wiley Periodicals LLC. Basic Protocol 1: In vivo EDC treatment of rats and immunostaining of treated oocytes Basic Protocol 2: In vitro fertilization and immunostaining of early-stage embryos.
Collapse
Affiliation(s)
- Liga Wuri
- Department of Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Paul W Zarutskie
- Department of Clinical Medicine & Primary Care, Sam Houston State University College of Osteopathic Medicine, Conroe, Texas
| | - Joe A Arosh
- Department of Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Sakhila K Banu
- Department of Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
13
|
Goutas A, Goutzourelas N, Kevrekidou A, Kevrekidis DP, Malea P, Virgiliou C, Assimopoulou AN, Trachana V, Kollatos N, Moustafa T, Liu M, Lin X, Komiotis D, Stagos D. Hypnea musciformis Seaweed Extract Protected Human Mesenchymal Stem Cells From Oxidative Stress Through NRF2 Activation. Food Sci Nutr 2024; 12:10816-10835. [PMID: 39723057 PMCID: PMC11666820 DOI: 10.1002/fsn3.4615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 10/06/2024] [Accepted: 11/02/2024] [Indexed: 12/28/2024] Open
Abstract
Previous studies have shown that Hypnea musciformis seaweed extracts (HMEs) possess antioxidant properties, but the molecular mechanisms accounting for this activity are not known. Thus, the present study investigated the molecular mechanisms through which HME exerted its antioxidant activity in human mesenchymal stem cells (WJ-MSCs). After the isolation of HME, its chemical composition was analyzed with gas chromatography mass spectrometry, indicating that it contained amino acids, organic acids, organic amides, sugar alcohols, saturated fatty acids, hydrogenated diterpene alcohols, and other organic compounds. Afterward, HME was shown in vitro to scavenge DPPH·, ABTS·+, ·OH, and O2 ·- radicals, possess reducing activity, and protect from ROO·-induced DNA strand breakage. Finally, the results showed that HME treatment of WJ-MSCs prevented H2O2-induced oxidative stress by decreasing lipid peroxidation, protein oxidation, reactive oxygen species levels, and DNA damage and by increasing glutathione levels. Moreover, our findings showed for the first time that HME's antioxidant activity in WJ-MSCs was mediated through the activation of NRF2, which upregulated the expression of the antioxidant proteins GCLC, GSR, HMOX1, SOD1, TXN, and GPX1. These results provide new insights into H. musciformis' antioxidant properties, which could help substantially its use as a food supplement or for developing biofunctional foods.
Collapse
Affiliation(s)
- Andreas Goutas
- Department of Biochemistry and Biotechnology, School of Health SciencesUniversity of ThessalyLarissaGreece
- Department of Biology, Faculty of MedicineUniversity of ThessalyLarissaGreece
| | - Nikolaos Goutzourelas
- Department of Biochemistry and Biotechnology, School of Health SciencesUniversity of ThessalyLarissaGreece
| | - Alkistis Kevrekidou
- Laboratory of Organic Chemistry, School of Chemical EngineeringAristotle University of ThessalonikiThessalonikiGreece
- Environmental Engineering Laboratory, Department of Chemical EngineeringAristotle University of ThessalonikiThessalonikiGreece
| | - Dimitrios Phaedon Kevrekidis
- Laboratory of Forensic Medicine and Toxicology, Department of MedicineAristotle University of ThessalonikiThessalonikiGreece
| | - Paraskevi Malea
- Department of Botany, School of BiologyAristotle University of ThessalonikiThessalonikiGreece
| | - Christina Virgiliou
- Laboratory of Analytical Chemistry, School of Chemical EngineeringAristotle University of ThessalonikiThessalonikiGreece
| | - Andreana N. Assimopoulou
- Laboratory of Organic Chemistry, School of Chemical EngineeringAristotle University of ThessalonikiThessalonikiGreece
| | - Varvara Trachana
- Department of Biology, Faculty of MedicineUniversity of ThessalyLarissaGreece
| | - Nikolaos Kollatos
- Department of Biochemistry and Biotechnology, School of Health SciencesUniversity of ThessalyLarissaGreece
| | - Tafa Moustafa
- Department of Biochemistry and Biotechnology, School of Health SciencesUniversity of ThessalyLarissaGreece
| | - Ming Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and PharmacyOcean University of ChinaQingdaoChina
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| | - Xiukun Lin
- Department of Pharmacology, School of PharmacySouthwest Medical UniversityLuzhouChina
| | - Dimitrios Komiotis
- Department of Biochemistry and Biotechnology, School of Health SciencesUniversity of ThessalyLarissaGreece
| | - Dimitrios Stagos
- Department of Biochemistry and Biotechnology, School of Health SciencesUniversity of ThessalyLarissaGreece
| |
Collapse
|
14
|
Yu L, Liu P. cGAS/STING signalling pathway in senescence and oncogenesis. Semin Cancer Biol 2024; 106-107:87-102. [PMID: 39222763 PMCID: PMC11625615 DOI: 10.1016/j.semcancer.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The cGAS/STING signaling pathway is a crucial component of the innate immune system, playing significant roles in sensing cytosolic DNA, regulating cellular senescence, and contributing to oncogenesis. Recent advances have shed new lights into the molecular mechanisms governing pathway activation in multiple pathophysiological settings, the indispensable roles of cGAS/STING signaling in cellular senescence, and its context-dependent roles in cancer development and suppression. This review summarizes current knowledge related to the biology of cGAS/STING signaling pathway and its participations into senescence and oncogenesis. We further explore the clinical implications and therapeutic potential for cGAS/STING targeted therapies, and faced challenges in the field. With a focus on molecular mechanisms and emerging pharmacological targets, this review underscores the importance of future studies to harness the therapeutic potential of the cGAS/STING pathway in treating senescence-related disorders and cancer. Advanced understanding of the regulatory mechanisms of cGAS/STING signaling, along with the associated deregulations in diseases, combined with the development of new classes of cGAS/STING modulators, hold great promises for creating novel and effective therapeutic strategies. These advancements could address current treatment challenges and unlock the full potential of cGAS/STING in treating senescence-related disorders and oncogenesis.
Collapse
Affiliation(s)
- Le Yu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biochemistry and Biophysics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biochemistry and Biophysics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
15
|
Bombelli S, Grasselli C, Mazzola P, Veronesi V, Morabito I, Zucchini N, Scollo CM, Blanco SI, De Marco S, Torsello B, Vitarelli F, Antolini L, Bianchi C, Leoni V, Bellelli G, Perego RA. Impairment of Renal and Hematopoietic Stem/Progenitor Cell Compartments in Frailty Syndrome: Link With Oxidative Stress, Plasma Cytokine Profiles, and Nuclear DNA Damage. J Gerontol A Biol Sci Med Sci 2024; 79:glae188. [PMID: 39066510 PMCID: PMC11439494 DOI: 10.1093/gerona/glae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Indexed: 07/28/2024] Open
Abstract
Frailty is an age-related syndrome that drives multiple physiological system impairments in some older adults, and its pathophysiological mechanisms remain unclear. We evaluated whether frailty-related biological processes could impair stem cell compartments, specifically the renal stem compartment, given that kidney dysfunctions are frequent in frailty. A well-characterized in vitro nephrosphere model of human adult renal stem/progenitor cells has been instrumental to and was appropriate for verifying this hypothesis in our current research. Evaluating the effects of plasma from older individuals with frailty (frail plasma) on allogeneic renal stem/progenitor cells, we showed significant functional impairment and nuclear DNA damage in the treated cells of the renal stem compartment. The analysis of the frail plasma revealed mitochondrial functional impairment associated with the activation of oxidative stress and a unique inflammatory mediator profile in frail individuals. In addition, the plasma of frail subjects also contained the highest percentage of DNA-damaged autologous circulating hematopoietic progenitor/stem cells. The integration of both molecular and functional data obtained allowed us to discern patterns associated with frailty status, irrespective of the comorbidities present in the frail individuals. The data obtained converged toward biological conditions that in frailty caused renal and hematopoietic impairment of stem cells, highlighting the possibility of concomitant exhaustion of several stem compartments.
Collapse
Affiliation(s)
- Silvia Bombelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Chiara Grasselli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paolo Mazzola
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Acute Geriatric Unit, IRCCS San Gerardo, Monza, Italy
| | - Valentina Veronesi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Center - B4, University of Milano-Bicocca, Monza, Italy
| | - Ivana Morabito
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Chiara M Scollo
- Immunotransfusional Unit, Laboratory of Hematology, IRCCS San Gerardo, Monza, Italy
| | | | - Sofia De Marco
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Barbara Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Federica Vitarelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Laboratory of Clinical Pathology and Toxicology, Pio XI Hospital, ASST-Brianza, Desio, Italy
| | - Laura Antolini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Center - B4, University of Milano-Bicocca, Monza, Italy
| | - Cristina Bianchi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Valerio Leoni
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Laboratory of Clinical Pathology and Toxicology, Pio XI Hospital, ASST-Brianza, Desio, Italy
| | - Giuseppe Bellelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Acute Geriatric Unit, IRCCS San Gerardo, Monza, Italy
| | - Roberto A Perego
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
16
|
Rezaeian AH, Wei W. Molecular signaling and clinical implications in the human aging-cancer cycle. Semin Cancer Biol 2024; 106-107:28-42. [PMID: 39197809 PMCID: PMC11625621 DOI: 10.1016/j.semcancer.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024]
Abstract
It is well documented that aging is associated with cancer, and likewise, cancer survivors display accelerated aging. As the number of aging individuals and cancer survivors continues to grow, it raises additional concerns across society. Therefore, unraveling the molecular mechanisms of aging in tissues is essential to developing effective therapies to fight the aging and cancer diseases in cancer survivors and cancer patients. Indeed, cellular senescence is a critical response, or a natural barrier to suppress the transition of normal cells into cancer cells, however, hypoxia which is physiologically required to maintain the stem cell niche, is increased by aging and inhibits senescence in tissues. Interestingly, oxygen restriction or hypoxia increases longevity and slows the aging process in humans, but hypoxia can also drive angiogenesis to facilitate cancer progression. In addition, cancer treatment is considered as one of the major reasons that drive cellular senescence, subsequently followed by accelerated aging. Several clinical trials have recently evaluated inhibitors to eliminate senescent cells. However, some mechanisms of aging typically can also retard cancer cell growth and progression, which might require careful strategy for better clinical outcomes. Here we describe the molecular regulation of aging and cancer in crosstalk with DNA damage and hypoxia signaling pathways in cancer patients and cancer survivors. We also update several therapeutic strategies that might be critical in reversing the cancer treatment-associated aging process.
Collapse
Affiliation(s)
- Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| |
Collapse
|
17
|
Dasgupta N, Arnold R, Equey A, Gandhi A, Adams PD. The role of the dynamic epigenetic landscape in senescence: orchestrating SASP expression. NPJ AGING 2024; 10:48. [PMID: 39448585 PMCID: PMC11502686 DOI: 10.1038/s41514-024-00172-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
Senescence and epigenetic alterations stand out as two well-characterized hallmarks of aging. When cells become senescent, they cease proliferation and release inflammatory molecules collectively termed the Senescence-Associated Secretory Phenotype (SASP). Senescence and SASP are implicated in numerous age-related diseases. Senescent cell nuclei undergo epigenetic reprogramming, which intricately regulates SASP expression. This review outlines the current understanding of how senescent cells undergo epigenetic changes and how these alterations govern SASP expression.
Collapse
Affiliation(s)
- Nirmalya Dasgupta
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Rouven Arnold
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Anais Equey
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Armin Gandhi
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Peter D Adams
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
18
|
Lloyd E, Xia F, Moore K, Zertuche C, Rastogi A, Kozol R, Kenzior O, Warren W, Appelbaum L, Moran RL, Zhao C, Duboue E, Rohner N, Keene AC. Elevated DNA Damage without signs of aging in the short-sleeping Mexican Cavefish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.590174. [PMID: 38659770 PMCID: PMC11042282 DOI: 10.1101/2024.04.18.590174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Dysregulation of sleep has widespread health consequences and represents an enormous health burden. Short-sleeping individuals are predisposed to the effects of neurodegeneration, suggesting a critical role for sleep in the maintenance of neuronal health. While the effects of sleep on cellular function are not completely understood, growing evidence has identified an association between sleep loss and DNA damage, raising the possibility that sleep facilitates efficient DNA repair. The Mexican tetra fish, Astyanax mexicanus provides a model to investigate the evolutionary basis for changes in sleep and the consequences of sleep loss. Multiple cave-adapted populations of these fish have evolved to sleep for substantially less time compared to surface populations of the same species without identifiable impacts on healthspan or longevity. To investigate whether the evolved sleep loss is associated with DNA damage and cellular stress, we compared the DNA Damage Response (DDR) and oxidative stress levels between A. mexicanus populations. We measured markers of chronic sleep loss and discovered elevated levels of the DNA damage marker γH2AX in the brain, and increased oxidative stress in the gut of cavefish, consistent with chronic sleep deprivation. Notably, we found that acute UV-induced DNA damage elicited an increase in sleep in surface fish but not in cavefish. On a transcriptional level, only the surface fish activated the photoreactivation repair pathway following UV damage. These findings suggest a reduction of the DDR in cavefish compared to surface fish that coincides with elevated DNA damage in cavefish. To examine DDR pathways at a cellular level, we created an embryonic fibroblast cell line from the two populations of A. mexicanus. We observed that both the DDR and DNA repair were diminished in the cavefish cells, corroborating the in vivo findings and suggesting that the acute response to DNA damage is lost in cavefish. To investigate the long-term impact of these changes, we compared the transcriptome in the brain and gut of aged surface fish and cavefish. Strikingly, many genes that are differentially expressed between young and old surface fish do not transcriptionally vary by age in cavefish. Taken together, these findings suggest that cavefish have developed resilience to sleep loss, despite possessing cellular hallmarks of chronic sleep deprivation.
Collapse
Affiliation(s)
- Evan Lloyd
- Department of Biology, Texas A&M University, College Station, TX 77840
| | - Fanning Xia
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Kinsley Moore
- Department of Biology, Texas A&M University, College Station, TX 77840
| | - Carolina Zertuche
- Department of Biology, Texas A&M University, College Station, TX 77840
| | - Aakriti Rastogi
- Department of Biology, Texas A&M University, College Station, TX 77840
| | - Rob Kozol
- Harriet Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458
| | - Olga Kenzior
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Wesley Warren
- Department of Genomics, University of Missouri, Columbia, MO 65211
| | - Lior Appelbaum
- Faculty of Life Science and the Multidisciplinary Brain Research Center, Bar Illan University, Ramat Gan, Israel
| | - Rachel L Moran
- Department of Biology, Texas A&M University, College Station, TX 77840
| | - Chongbei Zhao
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Erik Duboue
- Harriet Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Alex C Keene
- Department of Biology, Texas A&M University, College Station, TX 77840
| |
Collapse
|
19
|
Zhang X, Wang T. YIPF2 regulates genome integrity. Cell Biosci 2024; 14:114. [PMID: 39238039 PMCID: PMC11376028 DOI: 10.1186/s13578-024-01300-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024] Open
Abstract
Understanding of the mechanisms for genome integrity maintenance can help in developing effective intervention strategies to combat aging. A whole-genome RNAi screen was conducted to identify novel factors involved in maintaining genome stability. The potential target genes identified in the screening are related to the cell cycle, proteasome, and spliceosomes. Unexpectedly, the Golgi protein YIPF2 has been found to play a critical role in maintaining genome stability. The depletion of YIPF2 hinders the process of homologous recombination (HR) repair, which then triggers DNA damage response mechanisms, ultimately leading to cellular senescence. The overexpression of YIPF2 facilitated cellular recovery from DNA damage induced by chemotherapy agents or replicative senescence-associated DNA damage. Our findings indicate that only the intact Golgi apparatus containing YIPF2 provides a protective effect on genome integrity.
Collapse
Affiliation(s)
- Xiao Zhang
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint, Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou, China
- GIBH-CUHK Joint Research Laboratory On Stem Cell and Regenerative Medicine, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tao Wang
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint, Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou, China.
- GIBH-CUHK Joint Research Laboratory On Stem Cell and Regenerative Medicine, Guangzhou, China.
| |
Collapse
|
20
|
Youssef B, Feghaly C, Al Choboq J, Bou-Gharios J, Challita R, Azzi J, Bou Hadir H, Abi Antoun F, Araji T, Taddei PJ, Geara F, Sfeir P, Jurjus A, Abou-Kheir W, Bodgi L. Impaired DNA Double-Strand Break Repair in Irradiated Sheep Lung Fibroblasts: Late Effects of Previous Irradiation of the Spinal Thecal Sac. Cancers (Basel) 2024; 16:2968. [PMID: 39272826 PMCID: PMC11394103 DOI: 10.3390/cancers16172968] [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: 08/07/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Children with cancer previously treated with radiotherapy face the likelihood of side effects that can be debilitating or fatal. This study aimed to assess the long-term effect of medulloblastoma radiotherapy on the DNA double-strand break (DSB) repair capability of primary fibroblasts derived from lung biopsies of previously irradiated young sheep. This study included biopsies from three control and five irradiated sheep. The treated sheep had previously received spinal radiotherapy at a total dose of 28 Gy, which is equivalent to pediatric medulloblastoma treatment. Lung biopsies were taken 4 years post-irradiation from high-dose (HD, >18 Gy) and low-dose (LD, <2 Gy) regions. Fifteen cell lines were extracted (six control, four LD and five HD). The cells were irradiated, and DNA DSB repair was analyzed by immunofluorescence. Clonogenic, trypan blue and micronuclei assays were performed. Both the HD and LD cell lines had a significantly higher number of residual γH2AX foci 24 h and a significant decrease in pATM activity post-irradiation compared to the control. There was no statistically significant difference in the clonogenic assay, trypan blue and micronuclei results. Our study showed that a previous irradiation can impair the DNA DSB repair mechanism of ovine lung fibroblasts.
Collapse
Affiliation(s)
- Bassem Youssef
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Charbel Feghaly
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Joelle Al Choboq
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Jolie Bou-Gharios
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Rafka Challita
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Joyce Azzi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Hanine Bou Hadir
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Fabienne Abi Antoun
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Tarek Araji
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Phillip J Taddei
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
- Department of Radiation Oncology, Texas Oncology, Dallas, TX 75251, USA
| | - Fady Geara
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Pierre Sfeir
- Department of Surgery, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Larry Bodgi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
- U1296 Unit, "Radiation: Defense, Health and Environment", Centre Léon-Bérard, Inserm, 28 Rue Laennec, 69008 Lyon, France
| |
Collapse
|
21
|
Cohen A, Lubenski L, Mouzon A, Kupiec M, Weisman R. TORC2 is required for the accumulation of γH2A in response to DNA damage. J Biol Chem 2024; 300:107531. [PMID: 38971312 PMCID: PMC11321321 DOI: 10.1016/j.jbc.2024.107531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/08/2024] Open
Abstract
TOR protein kinases serve as the catalytic subunit of the TORC1 and TORC2 complexes, which regulate cellular growth, proliferation, and survival. In the fission yeast, Schizosaccharomyces pombe, cells lacking TORC2 or its downstream kinase Gad8 (AKT or SGK1 in human cells) exhibit sensitivity to a wide range of stress conditions, including DNA damage stress. One of the first responses to DNA damage is the phosphorylation of C-terminal serine residues within histone H2AX in human cells (γH2AX), or histone H2A in yeast cells (γH2A). The kinases responsible for γH2A in S. pombe are the two DNA damage checkpoint kinases Rad3 and Tel1 (ATR and ATM, respectively, in human cells). Here we report that TORC2-Gad8 signaling is required for accumulation of γH2A in response to DNA damage and during quiescence. Using the TOR-specific inhibitor, Torin1, we demonstrate that the effect of TORC2 on γH2A in response to DNA damage is immediate, rather than adaptive. The lack of γH2A is restored by deletion mutations of transcription and chromatin modification factors, including loss of components of Paf1C, SAGA, Mediator, and the bromo-domain proteins Bdf1/Bdf2. Thus, we suggest that TORC2-Gad8 may affect the accumulation of γH2A by regulating chromatin structure and function.
Collapse
Affiliation(s)
- Adiel Cohen
- Department of Natural Sciences, The Open University of Israel, Ranana, Israel
| | - Lea Lubenski
- The Shmunis School of Biomedicine & Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Ava Mouzon
- The Shmunis School of Biomedicine & Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Martin Kupiec
- The Shmunis School of Biomedicine & Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Weisman
- Department of Natural Sciences, The Open University of Israel, Ranana, Israel.
| |
Collapse
|
22
|
Alves-Silva JM, Pedreiro S, Zuzarte M, Cruz MT, Figueirinha A, Salgueiro L. Unlocking the Bioactive Potential and Exploring Novel Applications for Portuguese Endemic Santolina impressa. PLANTS (BASEL, SWITZERLAND) 2024; 13:1943. [PMID: 39065470 PMCID: PMC11280954 DOI: 10.3390/plants13141943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/26/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
The infusion of Santolina impressa, an endemic Portuguese plant, is traditionally used to treat various infections and disorders. This study aimed to assess its chemical profile by HPLC-DAD-ESI-MSn and validate its anti-inflammatory potential. In addition, the antioxidant capacity and effects on wound healing, lipogenesis, melanogenesis, and cellular senescence, all processes in which a dysregulated inflammatory response plays a pivotal role, were unveiled. The anti-inflammatory potential was assessed in lipopolysaccharide (LPS)-stimulated macrophages, cell migration was determined using a scratch wound assay, lipogenesis was assessed on T0901317-stimulated keratinocytes and melanogenesis on 3-isobutyl-1-methylxanthine (IBMX)-activated melanocytes. Etoposide was used to induce senescence in fibroblasts. Our results point out a chemical composition predominantly characterized by dicaffeoylquinic acids and low amounts of flavonols. Regarding the infusion's bioactive potential, an anti-inflammatory effect was evident through a decrease in nitric oxide production and inducible nitric oxide synthase and pro-interleukin-1β protein levels. Moreover, a decrease in fibroblast migration was observed, as well as an inhibition in both intracellular lipid accumulation and melanogenesis. Furthermore, the infusion decreased senescence-associated β-galactosidase activity, γH2AX nuclear accumulation and both p53 and p21 protein levels. Overall, this study confirms the traditional uses of S. impressa and ascribes additional properties of interest in the pharmaceutical and dermocosmetics industries.
Collapse
Affiliation(s)
- Jorge M. Alves-Silva
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (J.M.A.-S.); (M.Z.)
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
| | - Sónia Pedreiro
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (J.M.A.-S.); (M.Z.)
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
| | - Maria Teresa Cruz
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Univ Coimbra Center for Neuroscience and Cell Biology (CNC-UC), Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal
| | - Artur Figueirinha
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Lígia Salgueiro
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Univ Coimbra, Chemical Engineering and Renewable Resources for Sustainability (CERES), Department of Chemical Engineering, 3030-790 Coimbra, Portugal
| |
Collapse
|
23
|
Duarte LF, Villalobos V, Farías MA, Rangel-Ramírez MA, González-Madrid E, Navarro AJ, Carbone-Schellman J, Domínguez A, Alvarez A, Riedel CA, Bueno SM, Kalergis AM, Cáceres M, González PA. Asymptomatic herpes simplex virus brain infection elicits cellular senescence phenotypes in the central nervous system of mice suffering multiple sclerosis-like disease. Commun Biol 2024; 7:811. [PMID: 38965360 PMCID: PMC11224417 DOI: 10.1038/s42003-024-06486-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a demyelinating disease affecting the central nervous system (CNS) in animals that parallels several clinical and molecular traits of multiple sclerosis in humans. Herpes simplex virus type 1 (HSV-1) infection mainly causes cold sores and eye diseases, yet eventually, it can also reach the CNS, leading to acute encephalitis. Notably, a significant proportion of healthy individuals are likely to have asymptomatic HSV-1 brain infection with chronic brain inflammation due to persistent latent infection in neurons. Because cellular senescence is suggested as a potential factor contributing to the development of various neurodegenerative disorders, including multiple sclerosis, and viral infections may induce a premature senescence state in the CNS, potentially increasing susceptibility to such disorders, here we examine the presence of senescence-related markers in the brains and spinal cords of mice with asymptomatic HSV-1 brain infection, EAE, and both conditions. Across all scenarios, we find a significant increases of senescence biomarkers in the CNS with some differences depending on the analyzed group. Notably, some senescence biomarkers are exclusively observed in mice with the combined conditions. These results indicate that asymptomatic HSV-1 brain infection and EAE associate with a significant expression of senescence biomarkers in the CNS.
Collapse
MESH Headings
- Animals
- Cellular Senescence
- Mice
- Brain/virology
- Brain/pathology
- Brain/metabolism
- Multiple Sclerosis/virology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/metabolism
- Herpesvirus 1, Human/physiology
- Herpesvirus 1, Human/pathogenicity
- Herpes Simplex/virology
- Herpes Simplex/pathology
- Female
- Mice, Inbred C57BL
- Encephalomyelitis, Autoimmune, Experimental/virology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Phenotype
- Central Nervous System/virology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Spinal Cord/virology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Biomarkers/metabolism
- Encephalitis, Herpes Simplex/virology
- Encephalitis, Herpes Simplex/pathology
- Encephalitis, Herpes Simplex/metabolism
Collapse
Affiliation(s)
- Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Verónica Villalobos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ma Andreina Rangel-Ramírez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Enrique González-Madrid
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Areli J Navarro
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javier Carbone-Schellman
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angélica Domínguez
- Departamento de Salud Pública, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Alvarez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica Cáceres
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
24
|
Liu X, Mao X, Zhu C, Liu H, Fang Y, Fu T, Fan L, Liu M, Xiong Z, Tang H, Hu P, Le A. COMMD10 inhibited DNA damage to promote the progression of gastric cancer. J Cancer Res Clin Oncol 2024; 150:305. [PMID: 38871970 PMCID: PMC11176250 DOI: 10.1007/s00432-024-05817-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE The copper metabolism MURR1 domain 10 (COMMD10) plays a role in a variety of tumors. Here, we investigated its role in gastric cancer (GC). METHODS Online prediction tools, quantitative real-time PCR, western blotting and immunohistochemistry were used to evaluate the expression of COMMD10 in GC. The effect of COMMD10 knockdown was investigated in the GC cell lines and in in vivo xenograft tumor experiments. Western blotting and immunofluorescence were used to explore the relationships between COMMD10 and DNA damage. RESULTS The expression of COMMD10 was upregulated in GC compared to that in para-cancerous tissue and correlated with a higher clinical TNM stage (P = 0.044) and tumor size (P = 0.0366). High COMMD10 expression predicted poor prognosis in GC. Knockdown of COMMD10 resulted in the suppression of cell proliferation, migration, and invasion, accompanied by cell cycle arrest and an elevation in apoptosis rate. Moreover, the protein expression of COMMD10 was decreased in cisplatin-induced DNA-damaged GC cells. Suppression of COMMD10 impeded DNA damage repair, intensified DNA damage, and activated ATM-p53 signaling pathway in GC. Conversely, restoration of COMMD10 levels suppressed DNA damage and activation of the ATM-p53 signaling cascade. Additionally, knockdown of COMMD10 significantly restrained the growth of GC xenograft tumors while inhibiting DNA repair, augmenting DNA damage, and activating the ATM-p53 signaling pathway in xenograft tumor tissue. CONCLUSION COMMD10 is involved in DNA damage repair and maintains genomic stability in GC; knockdown of COMMD10 impedes the development of GC by exacerbating DNA damage, suggesting that COMMD10 may be new target for GC therapy.
Collapse
Affiliation(s)
- Xiaohua Liu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Xiaocheng Mao
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Chao Zhu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Hongfei Liu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Yangyang Fang
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Tianmei Fu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Linwei Fan
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Mengwei Liu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Ziqing Xiong
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Hong Tang
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Piaoping Hu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China.
| | - Aiping Le
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China.
| |
Collapse
|
25
|
Kücük P, Abbey L, Schmitt J, Henninger C, Fritz G. Cardiomyocytes, cardiac endothelial cells and fibroblasts contribute to anthracycline-induced cardiac injury through RAS-homologous small GTPases RAC1 and CDC42. Pharmacol Res 2024; 203:107165. [PMID: 38561112 DOI: 10.1016/j.phrs.2024.107165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/01/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
The clinical use of the DNA damaging anticancer drug doxorubicin (DOX) is limited by irreversible cardiotoxicity, which depends on the cumulative dose. The RAS-homologous (RHO) small GTPase RAC1 contributes to DOX-induced DNA damage formation and cardiotoxicity. However, the pathophysiological relevance of other RHO GTPases than RAC1 and different cardiac cell types (i.e., cardiomyocytes, non-cardiomyocytes) for DOX-triggered cardiac damage is unclear. Employing diverse in vitro and in vivo models, we comparatively investigated the level of DOX-induced DNA damage in cardiomyocytes versus non-cardiomyocytes (endothelial cells and fibroblasts), in the presence or absence of selected RHO GTPase inhibitors. Non-cardiomyocytes exhibited the highest number of DOX-induced DNA double-strand breaks (DSB), which were efficiently repaired in vitro. By contrast, rather low levels of DSB were formed in cardiomyocytes, which however remained largely unrepaired. Moreover, DOX-induced apoptosis was detected only in non-cardiomyocytes but not in cardiomyocytes. Pharmacological inhibitors of RAC1 and CDC42 most efficiently attenuated DOX-induced DNA damage in all cell types examined in vitro. Consistently, immunohistochemical analyses revealed that the RAC1 inhibitor NSC23766 and the pan-RHO GTPase inhibitor lovastatin reduced the level of DOX-induced residual DNA damage in both cardiomyocytes and non-cardiomyocytes in vivo. Overall, we conclude that endothelial cells, fibroblasts and cardiomyocytes contribute to the pathophysiology of DOX-induced cardiotoxicity, with RAC1- and CDC42-regulated signaling pathways being especially relevant for DOX-stimulated DSB formation and DNA damage response (DDR) activation. Hence, we suggest dual targeting of RAC1/CDC42-dependent mechanisms in multiple cardiac cell types to mitigate DNA damage-dependent cardiac injury evoked by DOX-based anticancer therapy.
Collapse
Affiliation(s)
- Pelin Kücük
- Institute of Toxicology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstrasse 5, Düsseldorf 40225, Germany.
| | - Lena Abbey
- Institute of Toxicology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Joachim Schmitt
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Christian Henninger
- Institute of Toxicology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstrasse 5, Düsseldorf 40225, Germany
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstrasse 5, Düsseldorf 40225, Germany.
| |
Collapse
|
26
|
Ibragimova M, Kussainova A, Aripova A, Bersimbaev R, Bulgakova O. The Molecular Mechanisms in Senescent Cells Induced by Natural Aging and Ionizing Radiation. Cells 2024; 13:550. [PMID: 38534394 PMCID: PMC10969416 DOI: 10.3390/cells13060550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
This review discusses the relationship between cellular senescence and radiation exposure. Given the wide range of ionizing radiation sources encountered by people in professional and medical spheres, as well as the influence of natural background radiation, the question of the effect of radiation on biological processes, particularly on aging processes, remains highly relevant. The parallel relationship between natural and radiation-induced cellular senescence reveals the common aspects underlying these processes. Based on recent scientific data, the key points of the effects of ionizing radiation on cellular processes associated with aging, such as genome instability, mitochondrial dysfunction, altered expression of miRNAs, epigenetic profile, and manifestation of the senescence-associated secretory phenotype (SASP), are discussed. Unraveling the molecular mechanisms of cellular senescence can make a valuable contribution to the understanding of the molecular genetic basis of age-associated diseases in the context of environmental exposure.
Collapse
Affiliation(s)
- Milana Ibragimova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Assiya Kussainova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
- Department of Health Sciences, University of Genova, Via Pastore 1, 16132 Genoa, Italy
| | - Akmaral Aripova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Rakhmetkazhi Bersimbaev
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Olga Bulgakova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| |
Collapse
|
27
|
Dai GC, Wang H, Ming Z, Lu PP, Li YJ, Gao YC, Shi L, Cheng Z, Liu XY, Rui YF. Heterotopic mineralization (ossification or calcification) in aged musculoskeletal soft tissues: A new candidate marker for aging. Ageing Res Rev 2024; 95:102215. [PMID: 38325754 DOI: 10.1016/j.arr.2024.102215] [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: 11/19/2023] [Revised: 01/21/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Aging can lead to various disorders in organisms and with the escalating impact of population aging, the incidence of age-related diseases is steadily increasing. As a major risk factor for chronic illnesses in humans, the prevention and postponement of aging have become focal points of research among numerous scientists. Aging biomarkers, which mirror molecular alterations at diverse levels in organs, tissues, and cells, can be used to monitor and evaluate biological changes associated with aging. Currently, aging biomarkers are primarily categorized into physiological traits, imaging characteristics, histological features, cellular-level alterations, and molecular-level changes that encompass the secretion of aging-related factors. However, in the context of the musculoskeletal soft tissue system, aging-related biological indicators primarily involve microscopic parameters at the cellular and molecular levels, resulting in inconvenience and uncertainty in the assessment of musculoskeletal soft tissue aging. To identify convenient and effective indicators, we conducted a comprehensive literature review to investigate the correlation between ectopic mineralization and age-related changes in the musculoskeletal soft tissue system. Here, we introduce the concept of ectopic mineralization as a macroscopic, reliable, and convenient biomarker for musculoskeletal soft tissue aging and present novel targets and strategies for the future management of age-related musculoskeletal soft tissue disorders.
Collapse
Affiliation(s)
- Guang-Chun Dai
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Hao Wang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Zhang Ming
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Pan-Pan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Ying-Juan Li
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Yu-Cheng Gao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Liu Shi
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Zhang Cheng
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Xiao-Yu Liu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China
| | - Yun-Feng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China; Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, Jiangsu 210009, PR China.
| |
Collapse
|
28
|
Nitzsche A, Hennig CL, von Brandenstein K, Döding A, Schulze-Späte U, Symmank J, Jacobs C. GDF15 Modulates the Zoledronic-Acid-Induced Hyperinflammatory Mechanoresponse of Periodontal Ligament Fibroblasts. Cells 2024; 13:147. [PMID: 38247838 PMCID: PMC10814077 DOI: 10.3390/cells13020147] [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: 11/06/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
Orthodontic tooth movement (OTM) is thought to be impeded by bisphosphonate (BP) therapy, mainly due to increased osteoclast apoptosis and changes in the periodontal ligament (PdL), a connecting tissue between the alveolar bone and teeth. PdL cells, mainly fibroblasts (PdLFs), are crucial regulators in OTM by modulating force-induced local inflammatory processes. Recently, we identified the TGF-β/BMP superfamily member GDF15 as an important modulator in OTM, promoting the pro-inflammatory mechanoresponses of PdLFs. The precise impact of the highly potent BP zoledronate (ZOL) on the mechanofunctionality of PdLFs is still under-investigated. Therefore, the aim of this study was to further characterize the ZOL-induced changes in the initial inflammatory mechanoresponse of human PdLFs (hPdLFs) and to further clarify a potential interrelationship with GDF15 signaling. Thus, two-day in vitro treatment with 0.5 µM, 5 µM and 50 µM of ZOL altered the cellular properties of hPdLFs partially in a concentration-dependent manner. In particular, exposure to ZOL decreased their metabolic activity, the proliferation rate, detected using Ki-67 immunofluorescent staining, and survival, analyzed using trypan blue. An increasing occurrence of DNA strand breaks was observed using TUNEL and an activated DNA damage response was demonstrated using H2A.X (phosphoS139) staining. While the osteogenic differentiation of hPdLFs was unaffected by ZOL, increased cellular senescence was observed using enhanced p21Waf1/Cip1/Sdi1 and β-galactosidase staining. In addition, cytokine-encoding genes such as IL6, IL8, COX2 and GDF15, which are associated with a senescence-associated secretory phenotype, were up-regulated by ZOL. Subsequently, this change in the hPdLF phenotype promoted a hyperinflammatory response to applied compressive forces with an increased expression of the pro-inflammatory markers IL1β, IL6 and GDF15, as well as the activation of monocytic THP1 cells. GDF15 appeared to be particularly relevant to these changes, as siRNA-mediated down-regulation balanced these hyperinflammatory responses by reducing IL-1β and IL-6 expression (IL1B p-value < 0.0001; IL6 p-value < 0.001) and secretion (IL-1β p-value < 0.05; IL-6 p-value < 0.001), as well as immune cell activation (p-value < 0.0001). In addition, ZOL-related reduced RANKL/OPG values and inhibited osteoclast activation were enhanced in GDF15-deficient hPdLFs (both p-values < 0.0001; all statistical tests: one-way ANOVA, Tukey's post hoc test). Thus, GDF15 may become a promising new target in the personalized orthodontic treatment of bisphosphonatepatients.
Collapse
Affiliation(s)
- Ann Nitzsche
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany; (A.N.); (C.-L.H.); (K.v.B.); (C.J.)
| | - Christoph-Ludwig Hennig
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany; (A.N.); (C.-L.H.); (K.v.B.); (C.J.)
| | - Katrin von Brandenstein
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany; (A.N.); (C.-L.H.); (K.v.B.); (C.J.)
| | - Annika Döding
- Section of Geriodontics, Department of Conservative Dentistry and Periodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany; (A.D.); (U.S.-S.)
| | - Ulrike Schulze-Späte
- Section of Geriodontics, Department of Conservative Dentistry and Periodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany; (A.D.); (U.S.-S.)
| | - Judit Symmank
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany; (A.N.); (C.-L.H.); (K.v.B.); (C.J.)
| | - Collin Jacobs
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany; (A.N.); (C.-L.H.); (K.v.B.); (C.J.)
| |
Collapse
|
29
|
Liu J, Liu K, Wang Y, Shi Z, Xu R, Zhang Y, Li J, Liu C, Xue B. Death receptor 5 is required for intestinal stem cell activity during intestinal epithelial renewal at homoeostasis. Cell Death Dis 2024; 15:27. [PMID: 38199990 PMCID: PMC10782029 DOI: 10.1038/s41419-023-06409-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024]
Abstract
Intestinal epithelial renewal, which depends on the proliferation and differentiation of intestinal stem cells (ISCs), is essential for epithelial homoeostasis. Understanding the mechanism controlling ISC activity is important. We found that death receptor 5 (DR5) gene deletion (DR5-/-) mice had impaired epithelial absorption and barrier function, resulting in delayed weight gain, which might be related to the general reduction of differentiated epithelial cells. In DR5-/- mice, the expression of ISC marker genes, the number of Olfm4+ ISCs, and the number of Ki67+ and BrdU+ cells in crypt were reduced. Furthermore, DR5 deletion inhibited the expression of lineage differentiation genes driving ISC differentiation into enterocytes, goblet cells, enteroendocrine cells, and Paneth cells. Therefore, DR5 gene loss may inhibit the intestinal epithelial renewal by dampening ISC activity. The ability of crypts from DR5-/- mice to form organoids decreased, and selective DR5 activation by Bioymifi promoted organoid growth and the expression of ISC and intestinal epithelial cell marker genes. Silencing of endogenous DR5 ligand TRAIL in organoids down-regulated the expression of ISC and intestinal epithelial cell marker genes. So, DR5 expressed in intestinal crypts was involved in the regulation of ISC activity. DR5 deletion in vivo or activation in organoids inhibited or enhanced the activity of Wnt, Notch, and BMP signalling through regulating the production of Paneth cell-derived ISC niche factors. DR5 gene deletion caused apoptosis and DNA damage in transit amplifying cells by inhibiting ERK1/2 activity in intestinal crypts. Inhibition of ERK1/2 with PD0325901 dampened the ISC activity and epithelial regeneration. In organoids, when Bioymifi's effect in activating ERK1/2 activity was completely blocked by PD0325901, its role in stimulating ISC activity and promoting epithelial regeneration was also eliminated. In summary, DR5 in intestinal crypts is essential for ISC activity during epithelial renewal under homoeostasis.
Collapse
Affiliation(s)
- Jianbo Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kaixuan Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Wang
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ziru Shi
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Runze Xu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yundi Zhang
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingxin Li
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuanyong Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bing Xue
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
30
|
Yadav P, Shah R, Roy A, Jani S, Chatterjee K, Saini DK. Cellular Senescence Program is Sensitive to Physical Differences in Polymeric Tissue Scaffolds. ACS MATERIALS AU 2024; 4:35-44. [PMID: 38221924 PMCID: PMC10786134 DOI: 10.1021/acsmaterialsau.3c00057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 01/16/2024]
Abstract
A typical cellular senescence program involves exposing cells to DNA-damaging agents such as ionization radiation or chemotherapeutic drugs, which cause multipronged changes, including increased cell size and volume, the onset of enhanced oxidative stress, and inflammation. In the present study, we examined if the senescence onset decision is sensitive to the design, porosity, and architecture of the substrate. To address this, we generated a library of polymeric scaffolds widely used in tissue engineering of varied stiffness, architecture, and porosity. Using irradiated A549 lung cancer cells, we examined the differences between cellular responses in these 3D scaffold systems and observed that senescence onset is equally diminished. When compared to the two-dimensional (2D) culture formats, there were profound changes in cell size and senescence induction in three-dimensional (3D) scaffolds. We further establish that these observed differences in the senescence state can be attributed to the altered cell spreading and cellular interactions on these substrates. This study elucidates the role of scaffold architecture in the cellular senescence program.
Collapse
Affiliation(s)
- Parul Yadav
- Department
of Bioengineering, Indian Institute of Science, C.V Raman Avenue, Bangalore, India 560012
| | - Rahul Shah
- Department
of Materials Engineering, Indian Institute
of Science, C.V Raman
Avenue, Bangalore, India 560012
| | - Anindo Roy
- Department
of Materials Engineering, Indian Institute
of Science, C.V Raman
Avenue, Bangalore, India 560012
| | - Sibani Jani
- Department
of Bioengineering, Indian Institute of Science, C.V Raman Avenue, Bangalore, India 560012
| | - Kaushik Chatterjee
- Department
of Bioengineering, Indian Institute of Science, C.V Raman Avenue, Bangalore, India 560012
- Department
of Materials Engineering, Indian Institute
of Science, C.V Raman
Avenue, Bangalore, India 560012
| | - Deepak Kumar Saini
- Department
of Bioengineering, Indian Institute of Science, C.V Raman Avenue, Bangalore, India 560012
- Department
of Developmental Biology and Genetics, C.V Raman Avenue, Indian Institute of Science, Bangalore, India 560012
| |
Collapse
|
31
|
Maruyama N, Fukunaga I, Kogo T, Endo T, Fujii W, Kanai-Azuma M, Naito K, Sugiura K. Accumulation of senescent cells in the stroma of aged mouse ovary. J Reprod Dev 2023; 69:328-336. [PMID: 37926520 PMCID: PMC10721854 DOI: 10.1262/jrd.2023-021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Senescent cells play a detrimental role in age-associated pathogenesis by producing factors involved in senescence-associated secretory phenotype (SASP). The present study was conducted to examine the possibility that senescent cells are present in aged ovaries and, if so, to determine the tissue region where senescent cells accumulate using a mouse model. Female mice at 2-4 and 8-10 months were used as reproductively young and aged models, respectively; the latter included mice with and without reproductive experience. Cells positive for senescence-associated β-galactosidase (SA-β-Gal) staining, one of the markers of cellular senescence, were detected in the stromal region of aged, but not young, ovaries regardless of reproductive experience. Likewise, the localization of cells expressing CDKN2A (cyclin dependent kinase inhibitor 2A), another senescence marker, in the stromal region of aged ovaries was detected with immunohistochemistry. CDKN2A expression detected by western blotting was significantly higher in the ovaries of aged mice with reproductive experience than in those without the experience. Moreover, cells positive for both γH2AX (a senescence marker) and fluorescent SA-β-Gal staining were present in those isolated from aged ovaries. In addition, the transcript levels of several SASP factors were significantly increased in aged ovaries. These results suggest that senescent cells accumulate in the ovarian stroma and may affect ovarian function in aged mice. Additionally, reproductive experience may promote accumulation.
Collapse
Affiliation(s)
- Natsumi Maruyama
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Isuzu Fukunaga
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomoaki Kogo
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tsutomu Endo
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Experimental Animal Model for Human Disease, Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo, Japan
| | - Wataru Fujii
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Present address: Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masami Kanai-Azuma
- Department of Experimental Animal Model for Human Disease, Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kunihiko Naito
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Koji Sugiura
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
32
|
Gao J, Wang H, Shen J, Liu X, Zhu X, Huang C, Li G, Sun Y, Liu Z, Sun YE, Liu H. Mutual regulation between GDF11 and TET2 prevents senescence of mesenchymal stem cells. J Cell Physiol 2023; 238:2827-2840. [PMID: 37801347 DOI: 10.1002/jcp.31132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/07/2023]
Abstract
Growth differentiation factor 11 (GDF11) is a putative systemic rejuvenation factor. In this study, we characterized the mechanism by which GDF11 reversed aging of mesenchymal stem cells (MSCs). In culture, aged MSCs proliferate slower and are positive for senescence markers senescence-associated β-galactosidase and P16ink4a . They have shortened telomeres, decreased GDF11 expression, and reduced osteogenic potential. GDF11 can block MSC aging in vitro and reverse age-dependent bone loss in vivo. The antiaging effect of GDF11 is via activation of the Smad2/3-PI3K-AKT-mTOR pathway. Unexpectedly, GDF11 also upregulated a DNA demethylase Tet2, which served as a key mediator for GDF11 to autoregulate itself via demethylation of the GDF11 promoter. Mutation of Tet2 facilitates MSC aging by blocking GDF11 expression. Mutagenesis of Tet2-regulated CpG sites also blocks GDF11 expression, leading to MSC aging. Together, a novel mutual regulatory relationship between GDF11 and an epigenetic factor Tet2 unveiled their antiaging roles.
Collapse
Affiliation(s)
- Jiaming Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junyan Shen
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaojing Liu
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoqi Zhu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ce Huang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Gongchen Li
- Department of Implantology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, School of Stomatology, Tongji University, Shanghai, China
| | - Yao Sun
- Department of Implantology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, School of Stomatology, Tongji University, Shanghai, China
| | - Zhongmin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Eve Sun
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Implantology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, School of Stomatology, Tongji University, Shanghai, China
- Department of Psychiatry and Biobehavioral Sciences, UCLA Medical School, Los Angeles, California, USA
| | - Hailiang Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Qian J, Zhou X, Tanaka K, Takahashi A. Alteration in the chromatin landscape during the DNA damage response: Continuous rotation of the gear driving cellular senescence and aging. DNA Repair (Amst) 2023; 131:103572. [PMID: 37742405 DOI: 10.1016/j.dnarep.2023.103572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/26/2023]
Abstract
The DNA damage response (DDR) is a crucial biological mechanism for maintaining cellular homeostasis in living organisms. This complex process involves a cascade of signaling pathways that orchestrate the sensing and processing of DNA lesions. Perturbations in this process may cause DNA repair failure, genomic instability, and irreversible cell cycle arrest, known as cellular senescence, potentially culminating in tumorigenesis. Persistent DDR exerts continuous and cumulative pressure on global chromatin dynamics, resulting in altered chromatin structure and perturbed epigenetic regulations, which are highly associated with cellular senescence and aging. Sustained DDR activation and heterochromatin changes further promote senescence-associated secretory phenotype (SASP), which is responsible for aging-related diseases and cancer development. In this review, we discuss the diverse mechanisms by which DDR leads to cellular senescence and triggers SASP, together with the evidence for DDR-induced chromatin remodeling and epigenetic regulation in relation to aging.
Collapse
Affiliation(s)
- Jianghao Qian
- Division of Cellular Senescence, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, Miyagi 980-8575, Japan
| | - Xiangyu Zhou
- Division of Cellular Senescence, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, Miyagi 980-8575, Japan
| | - Akiko Takahashi
- Division of Cellular Senescence, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; Cancer Cell Communication Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| |
Collapse
|
34
|
Alves-Silva JM, Moreira P, Cavaleiro C, Pereira C, Cruz MT, Salgueiro L. Effect of Ferulago lutea (Poir.) Grande Essential Oil on Molecular Hallmarks of Skin Aging. PLANTS (BASEL, SWITZERLAND) 2023; 12:3741. [PMID: 37960097 PMCID: PMC10648677 DOI: 10.3390/plants12213741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
With the increase in global life expectancy, maintaining health into old age becomes a challenge, and research has thus concentrated on various strategies which aimed to mitigate the effects of skin aging. Aromatic plants stand out as promising sources of anti-aging compounds due to their secondary metabolites, particularly essential oils (EOs). The aim of this study was to ascribe to Ferulago lutea EO several biological activities that could be useful in the context of skin aging. The EO was obtained using hydrodistillation and characterized by gas chromatography-mass spectrometry (GC/MS). The anti-inflammatory potential was assessed using lipopolysaccharide (LPS)-stimulated macrophages. The effect on cell migration was disclosed using scratch wound assay. Lipogenesis was induced using T0901317, hyperpigmentation with 3-isobutyl-1-methylxantine (IBMX) and senescence with etoposide. Our results show that the EO was characterized mainly by α-pinene and limonene. The EO was able to decrease nitric oxide (NO) release as well as iNOS and pro-IL-1β protein levels. The EO promoted wound healing while decreasing lipogenesis and having depigmenting effects. The EO also reduced senescence-associated β-galactosidase, p21/p53 protein levels and the nuclear accumulation of γH2AX. Overall, our study highlights the properties of F. lutea EO that make it a compelling candidate for dermocosmetics applications.
Collapse
Affiliation(s)
- Jorge M. Alves-Silva
- Univ Coimbra, Institute for Clinical and Biomedical Research, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, 3000-548 Coimbra, Portugal; (P.M.); (C.P.)
| | - Patrícia Moreira
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, 3000-548 Coimbra, Portugal; (P.M.); (C.P.)
- Univ Coimbra, Center for Neuroscience and Cell Biology, Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal;
| | - Carlos Cavaleiro
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, 3030-790 Coimbra, Portugal
| | - Cláudia Pereira
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, 3000-548 Coimbra, Portugal; (P.M.); (C.P.)
- Univ Coimbra, Center for Neuroscience and Cell Biology, Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal;
- Univ Coimbra, Faculty of Medicine, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
| | - Maria Teresa Cruz
- Univ Coimbra, Center for Neuroscience and Cell Biology, Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal;
| | - Lígia Salgueiro
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, 3030-790 Coimbra, Portugal
| |
Collapse
|
35
|
Tan J, Sun X, Zhao H, Guan H, Gao S, Zhou P. Double-strand DNA break repair: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2023; 4:e388. [PMID: 37808268 PMCID: PMC10556206 DOI: 10.1002/mco2.388] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
Double-strand break (DSB), a significant DNA damage brought on by ionizing radiation, acts as an initiating signal in tumor radiotherapy, causing cancer cells death. The two primary pathways for DNA DSB repair in mammalian cells are nonhomologous end joining (NHEJ) and homologous recombination (HR), which cooperate and compete with one another to achieve effective repair. The DSB repair mechanism depends on numerous regulatory variables. DSB recognition and the recruitment of DNA repair components, for instance, depend on the MRE11-RAD50-NBS1 (MRN) complex and the Ku70/80 heterodimer/DNA-PKcs (DNA-PK) complex, whose control is crucial in determining the DSB repair pathway choice and efficiency of HR and NHEJ. In-depth elucidation on the DSB repair pathway's molecular mechanisms has greatly facilitated for creation of repair proteins or pathways-specific inhibitors to advance precise cancer therapy and boost the effectiveness of cancer radiotherapy. The architectures, roles, molecular processes, and inhibitors of significant target proteins in the DSB repair pathways are reviewed in this article. The strategy and application in cancer therapy are also discussed based on the advancement of inhibitors targeted DSB damage response and repair proteins.
Collapse
Affiliation(s)
- Jinpeng Tan
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Xingyao Sun
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Hongling Zhao
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Hua Guan
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Shanshan Gao
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Ping‐Kun Zhou
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceChina
- Department of Radiation BiologyBeijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| |
Collapse
|
36
|
Michiba A, Gi M, Yokohira M, Sakurai E, Teramoto A, Kiriyama Y, Yamada S, Wanibuchi H, Tsukamoto T. Early detection of genotoxic hepatocarcinogens in rats using γH2AX and Ki-67: prediction by machine learning. Toxicol Sci 2023; 195:202-212. [PMID: 37527026 DOI: 10.1093/toxsci/kfad073] [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] [Indexed: 08/03/2023] Open
Abstract
Direct DNA double-strand breaks result in phosphorylation of H2AX, a variant of the histone H2 protein. Phosphorylated H2AX (γH2AX) may be a potential indicator in the evaluation of genotoxicity and hepatocarcinogenicity. In this study, γH2AX and Ki-67 were detected in the short-term responses (24 h after chemical administration) to classify genotoxic hepatocarcinogens (GHs) from non-GH chemicals. One hundred and thirty-five 6-week-old Crl: CD(SD) (SPF) male rats were treated with 22 chemicals including 11 GH and 11 non-GH, sacrificed 24 h later, and immunostained with γH2AX and Ki-67. Positivity rates of these markers were measured in the 3 liver ZONEs 1-3; portal, lobular, and central venous regions. These values were input into 3 machine learning models-Naïve Bayes, Random Forest, and k-Nearest Neighbor to classify GH and non-GH using a 10-fold cross-validation method. All 11 and 10 out of 11 GH caused significant increase in γH2AX and Ki-67 levels, respectively (P < .05). Of the 3 machine learning models, Random Forest performed the best. GH were identified with 95.0% sensitivity (76/80 GH-treated rats), 90.9% specificity (50/55 non-GH-treated rats), and 90.0% overall correct response rate using γH2AX staining, and 96.2% sensitivity (77/80), 81.8% specificity (45/55), and 90.4% overall correct response rate using Ki-67 labeling. Random Forest model using γH2AX and Ki-67 could independently predict GH in the early stage with high accuracy.
Collapse
Affiliation(s)
- Ayano Michiba
- Department of Diagnostic Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Min Gi
- Department of Environmental Risk Assessment, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Osaka 545-8585, Japan
| | - Masanao Yokohira
- Departments of Medical Education and Pathology and Host-Defense, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa 761-0793, Japan
| | - Eiko Sakurai
- Department of Diagnostic Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Atsushi Teramoto
- Faculty of Information Engineering, Meijo University, Nagoya, Aichi 468-8502, Japan
| | - Yuka Kiriyama
- Department of Diagnostic Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Department of Pathology, Narita Memorial Hospital, Toyohashi, Aichi 441-8029, Japan
| | - Seiji Yamada
- Department of Diagnostic Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Osaka 545-8585, Japan
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
37
|
Alessandra-Perini J, Machado DE, Palmero CY, Claudino MC, de Moura RS, Palumbo Junior A, Perini JA, Nasciutti LE. Euterpe oleracea extract (açaí) exhibits cardioprotective effects after chemotherapy treatment in a breast cancer model. BMC Complement Med Ther 2023; 23:301. [PMID: 37626388 PMCID: PMC10463785 DOI: 10.1186/s12906-023-04104-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Açaí, a Brazilian native fruit, has already been demonstrated to play a role in the progress of breast cancer and cardiotoxicity promoted by chemotherapy agents. Thus, the present study aimed to evaluate the combined use of açaí and the FAC-D chemotherapy protocol in a breast cancer model in vivo. METHODS Mammary carcinogenesis was induced in thirty female Wistar rats by subcutaneous injection of 25 mg/kg 7,12-dimethylbenzanthracene (DMBA) in the mammary gland. After sixty days, the rats were randomized into two groups: treated with 200 mg/kg of either açaí extract or vehicle, via gastric tube for 45 consecutive days. The FAC-D protocol was initiated after 90 days of induction by intraperitoneal injection for 3 cycles with a 7-day break each. After treatment, blood was collected for haematological and biochemical analyses, and tumours were collected for macroscopic and histological analyses. In the same way, heart, liver, and kidney samples were also collected for macroscopic and histological analyses. RESULTS Breast cancer was found as a cystic mass with a fibrotic pattern in the mammary gland. The histological analysis showed an invasive carcinoma area in both groups; however, in the saline group, there was a higher presence of inflammatory clusters. No difference was observed regarding body weight, glycaemia, aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, and urea in either group. However, açaí treatment decreased creatine kinase (CK), creatine kinase MB (CKMB), troponin I and C-reactive protein levels and increased the number of neutrophils and monocytes. Heart histopathology showed normal myocardium in the açaí treatment, while the saline group presented higher toxicity effects with loss of architecture of cardiac tissue. Furthermore, the açaí treatment presented greater collagen distribution, increased hydroxyproline concentration and lower H2AX immunostaining in the heart samples. CONCLUSION Açaí decreased the number of inflammatory cells in the tumor environment and exhibited protection against chemotherapy drug cardiotoxicity with an increased immune response in animals. Thus, açaí can be considered a promising low-cost therapeutic treatment that can be used in association with chemotherapy agents to avoid heart damage.
Collapse
Affiliation(s)
- Jéssica Alessandra-Perini
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
- Laboratório de Pesquisa em Ciências Farmacêuticas (LAPESF), Universidade do Estado do Rio de Janeiro (UERJ), Manuel Caldeira de Alvarenga Avenue, 1.203, Rio de Janeiro, RJ, 23070-200, Brazil.
| | - Daniel Escorsim Machado
- Laboratório de Pesquisa em Ciências Farmacêuticas (LAPESF), Universidade do Estado do Rio de Janeiro (UERJ), Manuel Caldeira de Alvarenga Avenue, 1.203, Rio de Janeiro, RJ, 23070-200, Brazil
| | - Celia Yelimar Palmero
- Laboratório Integrado de Morfologia, Instituto de Biodiversidade e Sustentabilidade, Universidade Federal do Rio de Janeiro 9UFRJ), Rio de Janeiro, RJ, Brazil
| | - Marllow Caetano Claudino
- Laboratório de Pesquisa em Ciências Farmacêuticas (LAPESF), Universidade do Estado do Rio de Janeiro (UERJ), Manuel Caldeira de Alvarenga Avenue, 1.203, Rio de Janeiro, RJ, 23070-200, Brazil
| | - Roberto Soares de Moura
- Departamento de Farmacologia e Psicobiologia, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Antônio Palumbo Junior
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Jamila Alessandra Perini
- Laboratório de Pesquisa em Ciências Farmacêuticas (LAPESF), Universidade do Estado do Rio de Janeiro (UERJ), Manuel Caldeira de Alvarenga Avenue, 1.203, Rio de Janeiro, RJ, 23070-200, Brazil
| | - Luiz Eurico Nasciutti
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
38
|
Xing X, Liang Y, Li Y, Zhao Y, Zhang Y, Li Z, Li Z, Wu Z. Fisetin Delays Postovulatory Oocyte Aging by Regulating Oxidative Stress and Mitochondrial Function through Sirt1 Pathway. Molecules 2023; 28:5533. [PMID: 37513404 PMCID: PMC10384696 DOI: 10.3390/molecules28145533] [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: 06/13/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The quality of oocytes determines the development potential of an embryo and is dependent on their timely fertilization after ovulation. Postovulatory oocyte aging is an inevitable factor during some assisted reproduction technology procedures, which results in poor fertilization rates and impairs embryo development. We found that fisetin, a bioactive flavonol contained in fruits and vegetables, delayed postovulatory oocyte aging in mice. Fisetin improved the development of aged oocytes after fertilization and inhibited the Sirt1 reduction in aged oocytes. Fisetin increased the GSH level and Sod2 transcription level to inhibit ROS accumulation in aged oocytes. Meanwhile, fisetin attenuated aging-induced spindle abnormalities, mitochondrial dysfunction, and apoptosis. At the molecular level, fisetin decreased aging-induced aberrant expression of H3K9me3. In addition, fisetin increased the expression levels of the mitochondrial transcription factor Tfam and the mitochondrial genes Co2 and Atp8 by upregulating Sirt1 in aged oocytes. Finally, inhibition of Sirt1 reversed the anti-aging effects of fisetin. Taken together, fisetin delayed postovulatory oocyte aging by upregulating Sirt1.
Collapse
Affiliation(s)
- Xupeng Xing
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yalin Liang
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yanan Li
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yaolu Zhao
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yuxing Zhang
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zheng Li
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Gene Bank of GuangDong Local Livestock and Poultry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
39
|
Kang T, Moore EC, Kopania EEK, King CD, Schilling B, Campisi J, Good JM, Brem RB. A natural variation-based screen in mouse cells reveals USF2 as a regulator of the DNA damage response and cellular senescence. G3 (BETHESDA, MD.) 2023; 13:jkad091. [PMID: 37097016 PMCID: PMC10320765 DOI: 10.1093/g3journal/jkad091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/26/2023]
Abstract
Cellular senescence is a program of cell cycle arrest, apoptosis resistance, and cytokine release induced by stress exposure in metazoan cells. Landmark studies in laboratory mice have characterized a number of master senescence regulators, including p16INK4a, p21, NF-κB, p53, and C/EBPβ. To discover other molecular players in senescence, we developed a screening approach to harness the evolutionary divergence between mouse species. We found that primary cells from the Mediterranean mouse Mus spretus, when treated with DNA damage to induce senescence, produced less cytokine and had less-active lysosomes than cells from laboratory Mus musculus. We used allele-specific expression profiling to catalog senescence-dependent cis-regulatory variation between the species at thousands of genes. We then tested for correlation between these expression changes and interspecies sequence variants in the binding sites of transcription factors. Among the emergent candidate senescence regulators, we chose a little-studied cell cycle factor, upstream stimulatory factor 2 (USF2), for molecular validation. In acute irradiation experiments, cells lacking USF2 had compromised DNA damage repair and response. Longer-term senescent cultures without USF2 mounted an exaggerated senescence regulatory program-shutting down cell cycle and DNA repair pathways, and turning up cytokine expression, more avidly than wild-type. We interpret these findings under a model of pro-repair, anti-senescence regulatory function by USF2. Our study affords new insights into the mechanisms by which cells commit to senescence, and serves as a validated proof of concept for natural variation-based regulator screens.
Collapse
Affiliation(s)
- Taekyu Kang
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
| | - Emily C Moore
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Emily E K Kopania
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | | | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jeffrey M Good
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Rachel B Brem
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
40
|
Tong X, Li D, Liu N, Huang W, Zhao X, Zhang D, Xue X, Fu J. Rad1 attenuates DNA double-strand breaks and cell cycle arrest in type II alveolar epithelial cells of rats with bronchopulmonary dysplasia. Mol Med 2023; 29:70. [PMID: 37226090 PMCID: PMC10207718 DOI: 10.1186/s10020-023-00660-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/27/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is the most common and serious chronic lung disease in preterm infants with pathological characteristics of arrested lung development. DNA double-strand breaks (DSBs) are a serious manifestation of oxidative stress damage, but little is known about the role of DSBs in BPD. The current study set out to detect DSB accumulation and cell cycle arrest in BPD and study the expression of genes related to DNA damage and repair in BPD through DNA damage signaling pathway-based PCR array to determine a suitable target to improve arrested lung development associated with BPD. METHODS DSB accumulation and cell cycle arrest were detected in a BPD animal model and primary cells, then a DNA damage signaling pathway-based PCR array was used to identify the target of DSB repair in BPD. RESULTS DSB accumulation and cell cycle arrest were shown in BPD animal model, primary type II alveolar epithelial cells (AECII) and cultured cells after exposure to hyperoxia. Of the 84 genes in the DNA damage-signaling pathway PCR array, eight genes were overexpressed and 11 genes were repressed. Rad1, an important protein for DSB repair, was repressed in the model group. Real-time PCR and western blots were used to verify the microarray results. Next, we confirmed that silencing Rad1 expression aggravated the accumulation of DSBs and cell cycle arrest in AECII cells, whereas its overexpression alleviated DSB accumulation and cell cycle arrest. CONCLUSIONS The accumulation of DSBs in AECII might be an important cause of alveolar growth arrest associated with BPD. Rad1 could be an effective target for intervention to improve this arrest in lung development associated with BPD.
Collapse
Affiliation(s)
- Xin Tong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Na Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanjie Huang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinyi Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dan Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
41
|
Uruski P, Matuszewska J, Leśniewska A, Rychlewski D, Niklas A, Mikuła-Pietrasik J, Tykarski A, Książek K. An integrative review of nonobvious puzzles of cellular and molecular cardiooncology. Cell Mol Biol Lett 2023; 28:44. [PMID: 37221467 DOI: 10.1186/s11658-023-00451-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/17/2023] [Indexed: 05/25/2023] Open
Abstract
Oncologic patients are subjected to four major treatment types: surgery, radiotherapy, chemotherapy, and immunotherapy. All nonsurgical forms of cancer management are known to potentially violate the structural and functional integrity of the cardiovascular system. The prevalence and severity of cardiotoxicity and vascular abnormalities led to the emergence of a clinical subdiscipline, called cardiooncology. This relatively new, but rapidly expanding area of knowledge, primarily focuses on clinical observations linking the adverse effects of cancer therapy with deteriorated quality of life of cancer survivors and their increased morbidity and mortality. Cellular and molecular determinants of these relations are far less understood, mainly because of several unsolved paths and contradicting findings in the literature. In this article, we provide a comprehensive view of the cellular and molecular etiology of cardiooncology. We pay particular attention to various intracellular processes that arise in cardiomyocytes, vascular endothelial cells, and smooth muscle cells treated in experimentally-controlled conditions in vitro and in vivo with ionizing radiation and drugs representing diverse modes of anti-cancer activity.
Collapse
Affiliation(s)
- Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Aleksandra Leśniewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Daniel Rychlewski
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Arkadiusz Niklas
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland.
| |
Collapse
|
42
|
Zhao Y, Lin X, Zeng W, Qin X, Miao B, Gao S, Liu J, Li Z. Berberine inhibits the progression of renal cell carcinoma cells by regulating reactive oxygen species generation and inducing DNA damage. Mol Biol Rep 2023:10.1007/s11033-023-08381-w. [PMID: 37217616 DOI: 10.1007/s11033-023-08381-w] [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: 07/22/2022] [Accepted: 03/09/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Berberine is a natural isoquinoline alkaloid that has been shown to have antitumor properties in a growing number of studies. However, its role in renal cell carcinoma remains unclear. This study investigates berberine's effect and mechanism in renal cell carcinoma. METHODS The methyl-tetrazolium, colony formation, and lactate dehydrogenase assay were used to detect proliferation and cytotoxicity, respectively. Flow cytometry, caspase-Glo 3/7 assay, and adenosine triphosphate assay were used to detect apoptosis and the adenosine triphosphate levels. Wound healing and transwell assay were used to examine the migration ability of renal cell carcinoma cells. Besides, the level of reactive oxygen species (ROS) was explored using a DCFH-DA-based kit. Additionally, western blot and Immunofluorescence assay was taken to determine the levels of relative proteins. RESULTS In vitro, our findings indicated that the proliferation and migration of renal cell carcinoma cells treated with berberine in various concentrations were inhibited, while the level of ROS and apoptosis rate were increased. Furthermore, The results of western blot showed that the expression of Bax, Bad, Bak, Cyto c, Clv-Caspase 3, Clv-Caspase 9, E-cadherin, TIMP-1and γH2AX were up-regulated, while Bcl-2, N-cadherin, Vimentin, Snail, Rad51 and PCNA were down-regulated after treating with berberine with various concentration. CONCLUSION The result of this study revealed that berberine inhibits renal cell carcinoma progression via regulating ROS generation and inducing DNA break.
Collapse
Affiliation(s)
- Yuwan Zhao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Xinghua Lin
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Wenfeng Zeng
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Xingzhang Qin
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Bailiang Miao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Sheng Gao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Jianjun Liu
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
| | - Zhuo Li
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
| |
Collapse
|
43
|
Vigašová K, Durdík M, Jakl L, Dolinská Z, Pobijaková M, Fekete M, Závacká I, Belyaev I, Marková E. Chemotherapy and cryopreservation affects DNA repair foci in lymphocytes of breast cancer patients. Int J Radiat Biol 2023; 99:1660-1668. [PMID: 37145321 DOI: 10.1080/09553002.2023.2211140] [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: 02/28/2022] [Accepted: 04/20/2023] [Indexed: 05/06/2023]
Abstract
PURPOSE Although breast cancer (BC) patients benefit from radiotherapy (RT), some radiosensitive (RS) patients suffer from side effects caused by ionizing radiation in healthy tissues. It is thought that RS is underlaid by a deficiency in the repair of DNA double-strand breaks (DSB). DNA repair proteins such as p53-binding protein 1 (53BP1) and phosphorylated histone H2AX (γH2AX), form DNA repair foci at the DSB locations and thus serve as DSB biomarkers. Peripheral blood lymphocytes (PBL) are commonly believed to be an appropriate cell system for RS assessment using DNA repair foci. The amount of DSB may also be influenced by chemotherapy (CHT), which is often chosen as the first treatment modality before RT. As it is not always possible to analyze blood samples immediately after collection, there is a need for cryopreservation of PBL in liquid nitrogen. However, cryopreservation may potentially affect the number of DNA repair foci. In this work, we studied the effect of cryopreservation and CHT on the amount of DNA repair foci in PBL of BC patients undergoing radiotherapy. MATERIALS AND METHODS The effect of cryopreservation was studied by immunofluorescence analysis of 53BP1 and γH2AX proteins at different time intervals after in vitro irradiation. The effect of chemotherapy was analyzed by fluorescent labelling of 53BP1 and γH2AX proteins in PBL collected before, during, and after RT. RESULTS Higher number of primary 53BP1/γH2AX foci was observed in frozen cells indicating that cryopreservation affects the formation of DNA repair foci in PBL of BC patients. In CHT-treated patients, a higher number of foci were found before RT, but no differences were observed during and after the RT. CONCLUSIONS Cryopreservation is the method of choice for analyzing DNA repair residual foci, but only similarly treated and preserved cells should be used for comparison of primary foci. CHT induces DNA repair foci in PBL of BC patients, but this effect disappears during radiotherapy.
Collapse
Affiliation(s)
- Katarína Vigašová
- Department of Radiobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Matúš Durdík
- Department of Radiobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lukáš Jakl
- Department of Radiobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Zuzana Dolinská
- Department of Radiation Oncology, Radiological Centrum, National Cancer Institute, Bratislava, Slovakia
| | - Margita Pobijaková
- Department of Radiation Oncology, Radiological Centrum, National Cancer Institute, Bratislava, Slovakia
| | - Marta Fekete
- Department of Radiation Oncology, Radiological Centrum, National Cancer Institute, Bratislava, Slovakia
| | - Ingrid Závacká
- Department of Radiation Oncology, Radiological Centrum, National Cancer Institute, Bratislava, Slovakia
| | - Igor Belyaev
- Department of Radiobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Eva Marková
- Department of Radiobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
44
|
Zhang C, Zhou Y, Zheng J, Ning N, Liu H, Jiang W, Yu X, Mu K, Li Y, Guo W, Hu H, Li J, Chen D. Inhibition of GABAA receptors in intestinal stem cells prevents chemoradiotherapy-induced intestinal toxicity. J Exp Med 2022; 219:213480. [PMID: 36125780 PMCID: PMC9499828 DOI: 10.1084/jem.20220541] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 08/07/2022] [Accepted: 08/26/2022] [Indexed: 11/04/2022] Open
Abstract
Lethal intestinal tissue toxicity is a common side effect and a dose-limiting factor in chemoradiotherapy. Chemoradiotherapy can trigger DNA damage and induce P53-dependent apoptosis in LGR5+ intestinal stem cells (ISCs). Gamma-aminobutyric acid (GABA) and its A receptors (GABAAR) are present in the gastrointestinal tract. However, the functioning of the GABAergic system in ISCs is poorly defined. We found that GABAAR α1 (GABRA1) levels increased in the murine intestine after chemoradiotherapy. GABRA1 depletion in LGR5+ ISCs protected the intestine from chemoradiotherapy-induced P53-dependent apoptosis and prolonged animal survival. The administration of bicuculline, a GABAAR antagonist, prevented chemoradiotherapy-induced ISC loss and intestinal damage without reducing the chemoradiosensitivity of tumors. Mechanistically, it was associated with the reduction of reactive oxygen species-induced DNA damage via the L-type voltage-dependent Ca2+ channels. Notably, flumazenil, a GABAAR antagonist approved by the U.S. Food and Drug Administration, rescued human colonic organoids from chemoradiotherapy-induced toxicity. Therefore, flumazenil may be a promising drug for reducing the gastrointestinal side effects of chemoradiotherapy.
Collapse
Affiliation(s)
- Cuiyu Zhang
- Department of Physiology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuping Zhou
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junjie Zheng
- Department of Physiology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Nannan Ning
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Haining Liu
- Department of Liver Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenyang Jiang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xin Yu
- Department of Biotherapy, State Key laboratory of Biotherapy and cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kun Mu
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yan Li
- Translational Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Wei Guo
- Department of Colorectal Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Huili Hu
- Department of Systems Biomedicine and Research Center of Stem Cell and Regenerative Medicine, Shandong University Cheeloo Medical College, School of Basic Medical Sciences, Jinan, China
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Jingxin Li:
| | - Dawei Chen
- Department of Physiology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Laboratory of Medical Chemistry, GIGA-Stem Cells, Faculty of Medicine, University of Liège, CHU, Sart-Tilman, Liège, Belgium
- Correspondence to Dawei Chen:
| |
Collapse
|
45
|
Tamanoi F, Yoshikawa K. Summary of volume 51: DNA damage and double-strand breaks. Enzymes 2022; 52:1-10. [PMID: 36423960 DOI: 10.1016/bs.enz.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
DNA damage and breaks are events that happen to DNA which exert a variety of influence on cell physiology including inhibition of DNA synthesis, repair response, cell cycle effect and cell death. Thus, it is important to deepened understanding of these events. In volume 51, we discussed topics including (1) assays to detect double-strand breaks, (2) conditions leading to double-strand breaks, (3) effects of irradiation, (4) DNA structure and chromatins, and (5) direct and indirect effect on DNA. Contributing authors and a table of contents for volume 51 are mentioned. We also discuss further issues and topics that need to be featured in future volumes. These include DNA damage sensors, DNA damage response proteins, and double-strand break repair pathways.
Collapse
Affiliation(s)
- Fuyuhiko Tamanoi
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan.
| | - Kenichi Yoshikawa
- Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan; Center for Integrative Medicine and Physics, Kyoto University, Kyoto, Japan
| |
Collapse
|
46
|
Toyoda T, Ogawa K. Early detection of urinary bladder carcinogens in rats by immunohistochemistry for γ-H2AX: a review from analyses of 100 chemicals. J Toxicol Pathol 2022; 35:283-298. [PMID: 36406171 PMCID: PMC9647216 DOI: 10.1293/tox.2022-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022] Open
Abstract
In safety evaluations of chemicals, there is an urgent need to develop short-term methods to replace long-term carcinogenicity tests. We have reported that immunohistochemistry for γ-H2AX, a well-established biomarker of DNA damage, can detect bladder carcinogens at an early stage using histopathological specimens from 28-day repeated-dose oral toxicity studies in rats. Given the markedly low level of γ-H2AX formation in the bladder urothelium of untreated rats, an increase in γ-H2AX-positive cells following chemical exposure can be relatively easy to identify. Among the 100 compounds examined to date, bladder carcinogens can be detected with high sensitivity (33/39; 84.6%) and specificity (58/61; 95.1%). As expected, γ-H2AX formation levels tended to be high following exposure to genotoxic bladder carcinogens, whereas nongenotoxic bladder carcinogens also increased the number of γ-H2AX-positive cells, probably through secondary DNA damage associated with sustained proliferative stimulation. γ-H2AX formation in the bladder urothelium reflects species differences in susceptibility to bladder carcinogenesis between rats and mice and shows a clear dose-dependency associated with the intensity of tumor development as well as high reproducibility. Some of the bladder carcinogens that showed false-negative results in the evaluation of γ-H2AX alone could be detected by combined evaluation with immunostaining for bladder stem cell markers, including aldehyde dehydrogenase 1A1. This method may be useful for the early detection of bladder carcinogens, as it can be performed by simple addition of conventional immunostaining using formalin-fixed paraffin-embedded tissues from 28-day repeated-dose toxicity studies in rodents, which are commonly used in safety evaluations of chemical substances.
Collapse
Affiliation(s)
- Takeshi Toyoda
- Division of Pathology, National Institute of Health
Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health
Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan
| |
Collapse
|
47
|
Targeting the USP7/RRM2 axis drives senescence and sensitizes melanoma cells to HDAC/LSD1 inhibitors. Cell Rep 2022; 40:111396. [PMID: 36130505 DOI: 10.1016/j.celrep.2022.111396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 07/01/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
Deubiquitinating enzymes are key regulators of the ubiquitin-proteasome system and cell cycle, and their dysfunction leads to tumorigenesis. Our in vivo drop-out screens in patient-derived xenograft models identify USP7 as a regulator of melanoma. We show that USP7 downregulation induces cellular senescence, arresting melanoma growth in vivo and proliferation in vitro in BRAF- and NRAS-mutant melanoma. We provide a comprehensive understanding of targets and networks affected by USP7 depletion by performing a global transcriptomic and proteomics analysis. We show that RRM2 is a USP7 target and is regulated by USP7 during S phase of the cell cycle. Ectopic expression of RRM2 in USP7-depleted cells rescues the senescent phenotype. Pharmacological inhibition of USP7 by P5091 phenocopies the shUSP7-induced senescent phenotype. We show that the bifunctional histone deacetylase (HDAC)/LSD1 inhibitor domatinostat has an additive antitumor effect, eliminating P5091-induced senescent cells, paving the way to a therapeutic combination for individuals with melanoma.
Collapse
|
48
|
Cao Y, Sun Q, Chen Z, Lu J, Geng T, Ma L, Zhang Y. CDKN2AIP is critical for spermiogenesis and germ cell development. Cell Biosci 2022; 12:136. [PMID: 35989335 PMCID: PMC9394077 DOI: 10.1186/s13578-022-00861-z] [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: 03/18/2022] [Accepted: 07/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background As a member of RNA-binding protein, CDKN2AIP has been shown to play a critical role in stem cell pluripotency and somatic differentiation. Recent studies indicate that Cdkn2aip is essential for spermatogonial self-renewal and proliferation through the activating Wnt-signaling pathway. However, the mechanisms of how Cdkn2aip regulate spermatogenesis is poorly characterized. Results We discovered that the CDKN2AIP was expressed in spermatocyte as well as spermatids and participated in spermiogenesis. Cdkn2aip−/− mice exhibited multiple sperm head defects accompanied by age dependent germ cell loss that might be result of protamine replacement failure and impaired SUN1 expression. Loss of Cdkn2aip expression in male mice resulted in synapsis failure in 19% of all spermatocytes and increased apoptosis due to damaged DNA double-strand break (DSB) repair and crossover formation. In vitro, knockdown of Cdkn2aip was associated with extended S phase, increased DNA damage and apoptosis. Conclusions Our findings not only identified the importance of CDKN2AIP in spermiogenesis and germ cell development, but also provided insight upon the driving mechanism. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00861-z.
Collapse
|
49
|
Hedyotis diffusae Herba-Andrographis Herba inhibits the cellular proliferation of nasopharyngeal carcinoma and triggers DNA damage through activation of p53 and p21. Cancer Gene Ther 2022; 29:973-983. [PMID: 34754077 DOI: 10.1038/s41417-021-00385-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 08/08/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022]
Abstract
Dysregulation of the cell cycle and the resulting aberrant cellular proliferation has been highlighted as a hallmark of cancer. Certain traditional Chinese medicines can inhibit cancer growth by inducing cell cycle arrest. In this study we explore the effect of Hedyotis diffusae Herba-Andrographis Herba on the cell cycle of nasopharyngeal carcinoma (NPC). Hedyotis diffusae Herba-Andrographis Herba-containing serum was prepared and then added to the cell culture medium. BrdU, comet, and FUCCI assays, western blot analysis and flow cytometry analysis revealed that Hedyotis diffusae Herba-Andrographis Herba treatment significantly alters cell proliferation, DNA damage, and cell cycle distribution. Xenograft mouse model experiments were performed, confirming these in vitro findings in vivo. Treatment with Hedyotis diffusae Herba-Andrographis Herba inhibited cell proliferation, promoted DNA damage, and arrested NPC cells progression from G1 to S phase. Further examination of the underlying molecular mechanisms revealed that treatment with Hedyotis diffusae Herba-Andrographis Herba increased the expression of p53 and p21, while reducing that of CCND1, Phospho-Rb, E2F1, γH2AX, and Ki-67 both in vivo and in vitro. Conversely, the inhibition of p53 and p21 could abolish the promoting effect of Hedyotis diffusae Herba-Andrographis Herba on the NPC cell cycle arrest at the G1 phase, contributing to the proliferation of NPC cells. Hedyotis diffusae Herba-Andrographis Herba suppressed the tumor growth in vivo. Overall, these findings suggest that Hedyotis Diffusae Herba-Andrographis prevent the progression of NPC by inducing NPC cell cycle arrest at the G1 phase through a p53/p21-dependent mechanism, providing a novel potential therapeutic treatment against NPC.
Collapse
|
50
|
Shafqat S, Arana Chicas E, Shafqat A, Hashmi SK. The Achilles' heel of cancer survivors: fundamentals of accelerated cellular senescence. J Clin Invest 2022; 132:e158452. [PMID: 35775492 PMCID: PMC9246373 DOI: 10.1172/jci158452] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent improvements in cancer treatment have increased the lifespan of pediatric and adult cancer survivors. However, cancer treatments accelerate aging in survivors, which manifests clinically as the premature onset of chronic diseases, such as endocrinopathies, osteoporosis, cardiac dysfunction, subsequent cancers, and geriatric syndromes of frailty, among others. Therefore, cancer treatment-induced early aging accounts for significant morbidity, mortality, and health expenditures among cancer survivors. One major mechanism driving this accelerated aging is cellular senescence; cancer treatments induce cellular senescence in tumor cells and in normal, nontumor tissue, thereby helping mediate the onset of several chronic diseases. Studies on clinical monitoring and therapeutic targeting of cellular senescence have made considerable progress in recent years. Large-scale clinical trials are currently evaluating senotherapeutic drugs, which inhibit or eliminate senescent cells to ameliorate cancer treatment-related aging. In this article, we survey the recent literature on phenotypes and mechanisms of aging in cancer survivors and provide an up-to-date review of the major preclinical and translational evidence on cellular senescence as a mechanism of accelerated aging in cancer survivors, as well as insight into the potential of senotherapeutic drugs. However, only with time will the clinical effect of senotherapies on cancer survivors be visible.
Collapse
Affiliation(s)
| | - Evelyn Arana Chicas
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Shahrukh K. Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Clinical Affairs, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| |
Collapse
|