1
|
Ramone T, Ghirri A, Prete A, Matrone A, Ciampi R, Piaggi P, Scutari M, Rago T, Torregrossa L, Romei C, Elisei R, Molinaro E. Molecular Profiling of Low-Risk Papillary Thyroid Carcinoma (mPTC) on Active Surveillance. J Clin Endocrinol Metab 2025; 110:685-692. [PMID: 39180363 DOI: 10.1210/clinem/dgae575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 08/26/2024]
Abstract
CONTEXT The active surveillance (AS) program for papillary thyroid carcinoma (≤1 cm) at low risk (mPTC) showed a low percentage of progression. OBJECTIVE The aim of this study was to find a molecular signature of cases that showed disease progression during AS, which would allow their early identification. METHODS We performed next-generation sequencing of 95 fine-needle aspiration cytology specimens from cases prospectively enrolled in the AS program to analyze key somatic driver alterations or gene fusions implicated in PTC tumorigenesis. TERT promoter analysis was performed using Sanger sequencing or droplet digital polymerase chain reaction. RESULTS BRAF p.V600E was found in 66.3% (63/95) of mPTC and was the most common somatic alteration, followed by RAS oncogene mutations detected in 3.2% of mPTC (3/95: 2 NRAS and 1 KRAS) and gene fusions detected in 3.2% of mPTC (3/95: 1 RET-PTC1, 1 TFG-NTRK1, 1 ALK imbalance). No TERT promoter mutations (C228T and C250T) were found in the analyzed mPTC (84/95). The comparison between the molecular profile and the clinical outcome of the mPTC (stable vs progressive disease) showed no correlation (P = .6) and did not identify a molecular signature able to identify progressive mPTC. CONCLUSION The molecular profile of mPTC is like that of bigger PTC with the exception that none of them showed a TERT promoter mutation. The identification of the most common driver mutations, such as BRAF, RAS, or gene fusions, is not helpful for the early identification of mPTC that will show disease progression during follow-up in the AS program.
Collapse
Affiliation(s)
- Teresa Ramone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Arianna Ghirri
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Alessandro Prete
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Antonio Matrone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Raffaele Ciampi
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Paolo Piaggi
- Department of Information Engineering, University of Pisa, Pisa 56124, Italy
| | - Maria Scutari
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Teresa Rago
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Liborio Torregrossa
- Department of Surgical, Medical and Molecular Pathology, University of Pisa, Pisa 56124, Italy
| | - Cristina Romei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Rossella Elisei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| | - Eleonora Molinaro
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Pisa 56124, Italy
| |
Collapse
|
2
|
Thangavelu L, Altamimi ASA, Ghaboura N, Babu MA, Roopashree R, Sharma P, Pal P, Choudhary C, Prasad GVS, Sinha A, Balaraman AK, Rawat S. Targeting the p53-p21 axis in liver cancer: Linking cellular senescence to tumor suppression and progression. Pathol Res Pract 2024; 263:155652. [PMID: 39437639 DOI: 10.1016/j.prp.2024.155652] [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: 09/06/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Liver cancer is a major health epidemic worldwide, mainly due to its high mortality rates and limited treatment options. The association of cellular senescence to tumorigenesis and the cancer hallmarks remains a subject of interest in cancer biology. The p53-p21 signalling axis is an important regulator in restoring the cell's balance by supporting tumor suppression and tumorigenesis in liver cancer. We review the novel molecular mechanisms that p53 and its downstream effector, p21, employ to induce cellular senescence, making it last longer, and halt the proliferation of damaged hepatocytes to become tumorous cells. We also examine how dysregulation of this pathway contributes to HCC pathogenesis, proliferation, survival, acquired resistance to apoptosis, and increased invasiveness. Furthermore, we comprehensively describe the molecular cross-talk between the p53-p21 signalling axis and major cell cycle signalling pathways, including Wnt/β-catenin, PI3K/Akt, and TGF-β in liver cancer and provide an overview of promising candidates for chemoprevention and future therapeutic strategies. This review article explores the roles of the p53-p21 pathway in liver cancer, examining its function in promoting cellular senescence under normal conditions and its potential role in cancer progression. It also highlights novel therapeutic drugs and drug targets within the pathway and discusses the implications for treatment strategies and prognosis in liver cancer.
Collapse
Affiliation(s)
- Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Pusparghya Pal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Chhavi Choudhary
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| |
Collapse
|
3
|
Splichal RC, Chen K, Walton SP, Chan C. The Role of Endoplasmic Reticulum Stress on Reducing Recombinant Protein Production in Mammalian Cells. Biochem Eng J 2024; 210:109434. [PMID: 39220803 PMCID: PMC11360842 DOI: 10.1016/j.bej.2024.109434] [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: 09/04/2024]
Abstract
Therapeutic recombinant protein production relies on industrial scale culture of mammalian cells to produce active proteins in quantities sufficient for clinical use. The combination of stresses from industrial cell culture environment and recombinant protein production can overwhelm the protein synthesis machinery in the endoplasmic reticulum (ER). This leads to a buildup of improperly folded proteins which induces ER stress. Cells respond to ER stress by activating the Unfolded Protein Response (UPR). To restore proteostasis, ER sensor proteins reduce global protein synthesis and increase chaperone protein synthesis, and if that is insufficient the proteins are degraded. If proteostasis is still not restored, apoptosis is initiated. Increasing evidence suggests crosstalk between ER proteostasis and DNA damage repair (DDR) pathways. External factors (e.g., metabolites) from the cellular environment as well as internal factors (e.g., transgene copy number) can impact genome stability. Failure to maintain genome integrity reduces cell viability and in turn protein production. This review focuses on the association between ER stress and processes that affect protein production and secretion. The processes mediated by ER stress, including inhibition of global protein translation, chaperone protein production, degradation of misfolded proteins, DNA repair, and protein secretion, impact recombinant protein production. Recombinant protein production can be reduced by ER stress through increased autophagy and protein degradation, reduced protein secretion, and reduced DDR response.
Collapse
Affiliation(s)
- R. Chauncey Splichal
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - Kevin Chen
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - S. Patrick Walton
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - Christina Chan
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, MI, USA
- Department of Computer Science and Engineering, Michigan State University, MI, USA
- Institute for Quantitative Health Science and Engineering, Division of Medical Devices, Michigan State University, MI, USA
| |
Collapse
|
4
|
Udah DC, Bakarey AS, Anetor GO, Omabe M, Edem VF, Ademowo OG, Anetor JI. Increased cancer risk in HIV-infected individuals occupationally exposed to chemicals: Depression of p53 as the key driver. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0002841. [PMID: 39042631 PMCID: PMC11265661 DOI: 10.1371/journal.pgph.0002841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 06/21/2024] [Indexed: 07/25/2024]
Abstract
The growing exposure to occupational chemicals and the spread of human immunodeficiency virus (HIV) infection are major global health issues. However, there is little data on the carcinogenic risk profile of HIV-infected individuals who have been occupationally exposed to chemical mixtures. This study therefore investigated the levels of cancer risk biomarkers in HIV-infected individuals exposed to occupational chemicals, exploring the relationship between apoptotic regulatory and oxidative response markers as a measure of cancer risk. Study participants (mean age 38.35±0.72 years) were divided into four groups according to their HIV status and occupational chemical exposure: 62 HIV-positive exposed (HPE), 66 HIV-positive unexposed (HPU), 60 HIV-negative exposed (HNE), and 60 HIV-negative unexposed (HNU). Serum p53, β-cell lymphoma-2 (bcl2), 8-hydroxydeoxyguanosine (8-OHdG), superoxide dismutase (SOD), and malondialdehyde (MDA) were measured using standard methods. Clusters of differentiation 4 (CD4+) T-lymphocytes were enumerated using flow cytometry. Serum p53 and bcl2 levels in HPE (0.91±0.11ng/ml and 122.37±15.77ng/ml) were significantly lower than HNU (1.49±0.15ng/ml and 225.52±33.67ng/ml) (p < 0.05), respectively. Wildtype p53 and bcl2 were positively and significantly correlated with 8-OHdG (r = 0.35, p<0.001; r = 0.36, p<0.001) and SOD (r = 0.38, p<0.001; r = 0.39, p<0.001). After controlling for gender, age, BMI, and cigarette smoking, both HIV status and SOD activity were significantly associated with wildtype p53 and bcl2 (p < 0.05). Malondialdehyde was significantly higher in the HPE (0.72 ± 0.01 mg/ml) than in the HNE (0.68 ± 0.01 mg/ml) and HNU (0.67 ± 0.01 mg/ml) groups (p < 0.05). The HPE group showed significantly lower CD4 counts than the HNE and HNU groups. Individuals who are HIV-infected and occupationally exposed to chemicals have a constellation of depressed immunity, elevated oxidative stress, and loss of tumour suppressive functions, which together intensify cancer risk, providing valuable scientific and public health bases for preventive measures in this vulnerable population.
Collapse
Affiliation(s)
- Donald C. Udah
- Department of Chemical Pathology, Laboratory for Toxicology and Micronutrient Metabolism, College of Medicine, University of Ibadan, Ibadan, Nigeria
- JSI Research & Training Institute Inc. (JSI), Abuja, Nigeria
| | - Adeleye S. Bakarey
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Biomedical Laboratory Science, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Gloria O. Anetor
- Department of Public Health Science, Faculty of Health Sciences, National Open University of Nigeria (NOUN), Abuja, Nigeria
| | - Maxwell Omabe
- Department of Medical Laboratory Sciences, School of Biomedical Science, Faculty of Health Science, Ebonyi State University, Nigeria
| | - Victory F. Edem
- Department of Immunology, University of Ibadan, Ibadan, Nigeria
| | - Olusegun G. Ademowo
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - John I. Anetor
- Department of Chemical Pathology, Laboratory for Toxicology and Micronutrient Metabolism, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
5
|
Erazo-Oliveras A, Muñoz-Vega M, Salinas ML, Wang X, Chapkin RS. Dysregulation of cellular membrane homeostasis as a crucial modulator of cancer risk. FEBS J 2024; 291:1299-1352. [PMID: 36282100 PMCID: PMC10126207 DOI: 10.1111/febs.16665] [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: 06/18/2022] [Revised: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Cellular membranes serve as an epicentre combining extracellular and cytosolic components with membranous effectors, which together support numerous fundamental cellular signalling pathways that mediate biological responses. To execute their functions, membrane proteins, lipids and carbohydrates arrange, in a highly coordinated manner, into well-defined assemblies displaying diverse biological and biophysical characteristics that modulate several signalling events. The loss of membrane homeostasis can trigger oncogenic signalling. More recently, it has been documented that select membrane active dietaries (MADs) can reshape biological membranes and subsequently decrease cancer risk. In this review, we emphasize the significance of membrane domain structure, organization and their signalling functionalities as well as how loss of membrane homeostasis can steer aberrant signalling. Moreover, we describe in detail the complexities associated with the examination of these membrane domains and their association with cancer. Finally, we summarize the current literature on MADs and their effects on cellular membranes, including various mechanisms of dietary chemoprevention/interception and the functional links between nutritional bioactives, membrane homeostasis and cancer biology.
Collapse
Affiliation(s)
- Alfredo Erazo-Oliveras
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Mónica Muñoz-Vega
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Michael L. Salinas
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Xiaoli Wang
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
- Center for Environmental Health Research; Texas A&M University; College Station, Texas, 77843; USA
| |
Collapse
|
6
|
Ma Y, Wang J, He X, Liu Y, Zhen S, An L, Yang Q, Niu F, Wang H, An B, Tai X, Yan Z, Wu C, Yang X, Liu X. Molecular mechanism of human ISG20L2 for the ITS1 cleavage in the processing of 18S precursor ribosomal RNA. Nucleic Acids Res 2024; 52:1878-1895. [PMID: 38153123 PMCID: PMC10899777 DOI: 10.1093/nar/gkad1210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/03/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023] Open
Abstract
The exonuclease ISG20L2 has been initially characterized for its role in the mammalian 5.8S rRNA 3' end maturation, specifically in the cleavage of ITS2 of 12S precursor ribosomal RNA (pre-rRNA). Here, we show that human ISG20L2 is also involved in 18S pre-rRNA maturation through removing the ITS1 region, and contributes to ribosomal biogenesis and cell proliferation. Furthermore, we determined the crystal structure of the ISG20L2 nuclease domain at 2.9 Å resolution. It exhibits the typical αβα fold of the DEDD 3'-5' exonuclease with a catalytic pocket located in the hollow near the center. The catalytic residues Asp183, Glu185, Asp267, His322 and Asp327 constitute the DEDDh motif in ISG20L2. The active pocket represents conformational flexibility in the absence of an RNA substrate. Using structural superposition and mutagenesis assay, we mapped RNA substrate binding residues in ISG20L2. Finally, cellular assays revealed that ISG20L2 is aberrantly up-regulated in colon adenocarcinoma and promotes colon cancer cell proliferation through regulating ribosome biogenesis. Together, these results reveal that ISG20L2 is a new enzymatic member for 18S pre-rRNA maturation, provide insights into the mechanism of ISG20L2 underlying pre-rRNA processing, and suggest that ISG20L2 is a potential therapeutic target for colon adenocarcinoma.
Collapse
Affiliation(s)
- Yinliang Ma
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Jiaxu Wang
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
- College of Life Sciences, State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, Xinxiang 453002 Henan, China
| | - Xingyi He
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Yuhang Liu
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Shuo Zhen
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Lina An
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Qian Yang
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Fumin Niu
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Hong Wang
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Boran An
- Affiliated Hospital of Hebei University, Hebei University, Baoding 071002 Hebei, China
| | - Xinyue Tai
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Zhenzhen Yan
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Chen Wu
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| | - Xiaoyun Yang
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
- Department of Biology, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Xiuhua Liu
- College of Life Sciences, Hebei Innovation Center for Bioengineering and Biotechnology, Institute of Life Sciences and Green Development, Hebei University, Baoding 071002 Hebei, China
| |
Collapse
|
7
|
Singh K, Agrawal L, Gupta R, Singh D, Kathpalia M, Kaur N. Lectins as a promising therapeutic agent for breast cancer: A review. Breast Dis 2024; 43:193-211. [PMID: 38905027 PMCID: PMC11307042 DOI: 10.3233/bd-230047] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
Efficient treatment of cancer has been a subject of research by scientists for many years. Current treatments for cancer, such as radiotherapy, chemotherapy and surgery have been used in traditional combination therapy, but they have major setbacks like non-specificity, non-responsiveness in certain cancer types towards treatment, tumor recurrence, etc. Epidemiological data has shown that breast cancer accounts for 14% of cancer cases occurring in Indian women. In recent years, scientists have started to focus on the use of natural compounds like lectins obtained from various sources to counter the side effects of traditional therapy. Lectins like Sambucus nigra Agglutinin, Maackia amurensis lectin, Okra lectins, Haliclona caerulea lectin, Sclerotium rolfsii lectin, etc., have been discovered to have both diagnostic and therapeutic potential for breast cancer patients. Lectins have been found to have inhibitory effects on various cancer cell activities such as neo-angiogenesis, causing cell cycle arrest at the G1 phase, and inducing apoptosis. The major idea behind the use of lectins in cancer diagnostics and therapeutics is their capability to bind to glycosylated proteins that are expressed on the cell surface. This review focuses on an exploration of the roles of post-translational modification in cancer cells, especially glycosylation, and the potential of lectins in cancer diagnosis and therapeutics.
Collapse
Affiliation(s)
- Keerti Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Lokita Agrawal
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Rhea Gupta
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Divyam Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Meghavi Kathpalia
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Navkiran Kaur
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
8
|
Chowdhury SR, Chuong P, Mgbemena VE, Statsyuk A. Development of a PROTAC Targeting Chk1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573733. [PMID: 38260247 PMCID: PMC10802242 DOI: 10.1101/2023.12.30.573733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
A series of Chk1 degraders were designed and synthesized. The degraders were developed through the conjugation of a promiscuous kinase binder and thalidomide. One of the degraders PROTAC-2 was able to decrease Chk1 levels in a concentration-dependent manner in A375 cells. The developed probes can be useful for the development of selective and more potent Chk1 degraders.
Collapse
Affiliation(s)
- Sandipan Roy Chowdhury
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Health 2, 4349 Martin Luther King Boulevard, Houston, Texas, 77204
| | - Patrick Chuong
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Health 2, 4349 Martin Luther King Boulevard, Houston, Texas, 77204
| | - Victoria E Mgbemena
- Department of Biology, MD and S Brailsford College of Arts and Sciences, Prairie View A&M University, Prairie View, TX 77446, USA
| | - Alexander Statsyuk
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Health 2, 4349 Martin Luther King Boulevard, Houston, Texas, 77204
| |
Collapse
|
9
|
Shaheer K, Prabhu BS, Ali HS, Lakshmanan-M D. Breast cancer cells are sensitized by piperine to radiotherapy through estrogen receptor-α mediated modulation of a key NHEJ repair protein- DNA-PK. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155126. [PMID: 37913642 DOI: 10.1016/j.phymed.2023.155126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/03/2023] [Accepted: 09/27/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Non-homologous end joining, an important DNA-double-stranded break repair pathway, plays a prominent role in conferring resistance to radiotherapeutic agents, resulting in cancer progression and relapse. PURPOSE The molecular players involved in the radio-sensitizing effects of piperine and many other phytocompounds remain evasive to a great extent. The study is designed to assess if piperine, a plant alkaloid can alter the radioresistance by modulating the expression of non-homologous end-joining machinery. METHODS AND MATERIALS Estrogen receptor-positive/negative, breast cancer cells were cultured to understand the synergetic effects of piperine with radiotherapy. Cisplatin and Bazedoxifene were used as positive controls. Cells were exposed to γ- radiation using Low Dose gamma Irradiator-2000. The piperine effect on Estrogen receptor modulation, DNA-Damage, DNA-Damage-Response, and apoptosis was done by western blotting, immunofluorescence, yeast-based-estrogen-receptor-LacZ-reporter assay, and nuclear translocation analysis. Micronuclei assay was done for DNA damage and genotoxicity, and DSBs were quantified by γH2AX-foci-staining using confocal microscopy. Flow cytometry analysis was done to determine the cell cycle, mitochondrial membrane depolarization, and Reactive oxygen species generation. Pharmacophore analysis and protein-ligand interaction studies were done using Schrodinger software. Synergy was computed by compusyn-statistical analysis. Standard errors/deviation/significance were computed with GraphPad prism. RESULTS Using piperine, we propose a new strategy for overcoming acquired radioresistance through estrogen receptor-mediated modulation of the NHEJ pathway. This is the first comprehensive study elucidating the mechanism of radio sensitizing potential of piperine. Piperine enhanced the radiation-induced cell death and enhanced the expression and activation of Estrogen receptor β, while Estrogen receptor α expression and activation were reduced. In addition, piperine shares common pharmacophore features with most of the known estrogen agonists and antagonists. It altered the estrogen receptor α/β ratio and the expression of estrogen-responsive proteins of DDR and NHEJ pathway. Enhanced expression of DDR proteins, ATM, p53, and P-p53 with low DNA-PK repair complex (comprising of DNA-PKcs/Ku70/Ku80), resulted in the accumulation of radiation-induced DNA double-stranded breaks (as evidenced by MNi and γH2AX-foci) culminating in cell cycle arrest and mitochondrial-pathway of apoptosis. CONCLUSION In conclusion, our study for the first time reported that piperine sensitizes breast cancer cells to radiation by accumulating DNA breaks, through altering the expression of DNA-PK Complex, and DDR proteins, via selective estrogen receptor modulation, offering a novel strategy for combating radioresistance.
Collapse
Affiliation(s)
- Koniyan Shaheer
- Division of Cancer Research and Therapeutics (CaRT), Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - Br Swathi Prabhu
- Division of Cancer Research and Therapeutics (CaRT), Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - H Shabeer Ali
- Department of Biotechnology and Microbiology, Kannur University, Kannur, Kerala, India
| | - Divya Lakshmanan-M
- Division of Cancer Research and Therapeutics (CaRT), Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India.
| |
Collapse
|
10
|
Coll RP, Bright SJ, Martinus DKJ, Georgiou DK, Sawakuchi GO, Manning HC. Alpha Particle-Emitting Radiopharmaceuticals as Cancer Therapy: Biological Basis, Current Status, and Future Outlook for Therapeutics Discovery. Mol Imaging Biol 2023; 25:991-1019. [PMID: 37845582 PMCID: PMC12054971 DOI: 10.1007/s11307-023-01857-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 10/18/2023]
Abstract
Critical advances in radionuclide therapy have led to encouraging new options for cancer treatment through the pairing of clinically useful radiation-emitting radionuclides and innovative pharmaceutical discovery. Of the various subatomic particles used in therapeutic radiopharmaceuticals, alpha (α) particles show great promise owing to their relatively large size, delivered energy, finite pathlength, and resulting ionization density. This review discusses the therapeutic benefits of α-emitting radiopharmaceuticals and their pairing with appropriate diagnostics, resulting in innovative "theranostic" platforms. Herein, the current landscape of α particle-emitting radionuclides is described with an emphasis on their use in theranostic development for cancer treatment. Commonly studied radionuclides are introduced and recent efforts towards their production for research and clinical use are described. The growing popularity of these radionuclides is explained through summarizing the biological effects of α radiation on cancer cells, which include DNA damage, activation of discrete cell death programs, and downstream immune responses. Examples of efficient α-theranostic design are described with an emphasis on strategies that lead to cellular internalization and the targeting of proteins involved in therapeutic resistance. Historical barriers to the clinical deployment of α-theranostic radiopharmaceuticals are also discussed. Recent progress towards addressing these challenges is presented along with examples of incorporating α-particle therapy in pharmaceutical platforms that can be easily converted into diagnostic counterparts.
Collapse
Affiliation(s)
- Ryan P Coll
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | - Scott J Bright
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - David K J Martinus
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - Dimitra K Georgiou
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | - Gabriel O Sawakuchi
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - H Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA.
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA.
| |
Collapse
|
11
|
Hemmati F, Akinpelu A, Song J, Amiri F, McDaniel A, McMurray C, Afthinos A, Andreadis ST, Aitken AV, Biancardi VC, Gerecht S, Mistriotis P. Downregulation of YAP Activity Restricts P53 Hyperactivation to Promote Cell Survival in Confinement. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302228. [PMID: 37267923 PMCID: PMC10427377 DOI: 10.1002/advs.202302228] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Indexed: 06/04/2023]
Abstract
Cell migration through confining three dimensional (3D) topographies can lead to loss of nuclear envelope integrity, DNA damage, and genomic instability. Despite these detrimental phenomena, cells transiently exposed to confinement do not usually die. Whether this is also true for cells subjected to long-term confinement remains unclear at present. To investigate this, photopatterning and microfluidics are employed to fabricate a high-throughput device that circumvents limitations of previous cell confinement models and enables prolonged culture of single cells in microchannels with physiologically relevant length scales. The results of this study show that continuous exposure to tight confinement can trigger frequent nuclear envelope rupture events, which in turn promote P53 activation and cell apoptosis. Migrating cells eventually adapt to confinement and evade cell death by downregulating YAP activity. Reduced YAP activity, which is the consequence of confinement-induced YAP1/2 translocation to the cytoplasm, suppresses the incidence of nuclear envelope rupture and abolishes P53-mediated cell death. Cumulatively, this work establishes advanced, high-throughput biomimetic models for better understanding cell behavior in health and disease, and underscores the critical role of topographical cues and mechanotransduction pathways in the regulation of cell life and death.
Collapse
Affiliation(s)
- Farnaz Hemmati
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | - Ayuba Akinpelu
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Farshad Amiri
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | - Anya McDaniel
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | - Collins McMurray
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | | | - Stelios T. Andreadis
- Departments of Chemical and Biological EngineeringThe State University of New YorkBuffaloNY14260USA
- Department of Biomedical EngineeringUniversity at BuffaloThe State University of New YorkBuffaloNY14228USA
- Center of Excellence in Bioinformatics and Life SciencesBuffaloNY14203USA
- Center for Cell Gene and Tissue Engineering (CGTE)University at BuffaloThe State University of New YorkBuffaloNY14260USA
| | - Andrew V. Aitken
- Department of AnatomyPhysiology and PharmacologyCollege of Veterinary MedicineAuburn UniversityAuburnAL36849USA
- Center for Neurosciences InitiativeAuburn UniversityAuburnAL36849USA
| | - Vinicia C. Biancardi
- Department of AnatomyPhysiology and PharmacologyCollege of Veterinary MedicineAuburn UniversityAuburnAL36849USA
- Center for Neurosciences InitiativeAuburn UniversityAuburnAL36849USA
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | |
Collapse
|
12
|
Bernardo T, Behrends C, Klein D, Kuntze A, Timmermann B, von Neubeck C. Similar additive effects of doxorubicin in combination with photon or proton irradiation in soft tissue sarcoma models. Front Oncol 2023; 13:1211984. [PMID: 37503316 PMCID: PMC10368985 DOI: 10.3389/fonc.2023.1211984] [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: 04/25/2023] [Accepted: 06/19/2023] [Indexed: 07/29/2023] Open
Abstract
High-precision radiotherapy with proton beams is frequently used in the management of aggressive soft tissue sarcoma (STS) and is often combined with doxorubicin (Dox), the first-line chemotherapy for STS. However, current treatment approaches continue to result in high local recurrence rates often occurring within the treatment field. This strongly indicates the need of optimized treatment protocols taking the vast heterogeneity of STS into account, thereby fostering personalized treatment approaches. Here, we used preclinical STS models to investigate the radiation response following photon (X) or proton (H) irradiation alone and in combination with different treatment schedules of Dox. As preclinical models, fibrosarcoma (HT-1080), undifferentiated pleiomorphic sarcoma (GCT), and embryonal rhabdomyosarcoma (RD) cell lines were used; the latter two are mutated for TP53. The cellular response regarding clonogenic survival, apoptosis, cell-cycle distribution, proliferation, viability, morphology, and motility was investigated. The different STS cell types revealed a dose-dependent radiation response with reduced survival, proliferation, viability, and motility whereas G2/M phase arrest as well as apoptosis were induced. RD cells showed the most radiosensitive phenotype; the linear quadratic model fit could not be applied. In combined treatment schedules, Dox showed the highest efficiency when applied after or before and after radiation; Dox treatment only before radiation was less efficient. GCT cells were the most chemoresistant cell line in this study most probably due to their TP53 mutation status. Interestingly, similar additive effects could be observed for X or H irradiation in combination with Dox treatment. However, the additive effects were determined more frequently for X than for H irradiation. Thus, further investigations are needed to specify alternative drug therapies that display superior efficacy when combined with H therapy.
Collapse
Affiliation(s)
- Teresa Bernardo
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carina Behrends
- West German Proton Therapy Center Essen (WPE), Essen, Germany
- West German Cancer Centre (WTZ), University Hospital Essen, Essen, Germany
- Faculty of Physics, Technical University (TU) Dortmund University, Dortmund, Germany
| | - Diana Klein
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anna Kuntze
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Beate Timmermann
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- West German Proton Therapy Center Essen (WPE), Essen, Germany
- West German Cancer Centre (WTZ), University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Essen, Germany
| | - Cläre von Neubeck
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
13
|
Yeo XH, Sundararajan V, Wu Z, Phua ZJC, Ho YY, Peh KLE, Chiu YC, Tan TZ, Kappei D, Ho YS, Tan DSP, Tam WL, Huang RYJ. The effect of inhibition of receptor tyrosine kinase AXL on DNA damage response in ovarian cancer. Commun Biol 2023; 6:660. [PMID: 37349576 PMCID: PMC10287694 DOI: 10.1038/s42003-023-05045-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/14/2023] [Indexed: 06/24/2023] Open
Abstract
AXL is a receptor tyrosine kinase that is often overexpressed in cancers. It contributes to pathophysiology in cancer progression and therapeutic resistance, making it an emerging therapeutic target. The first-in-class AXL inhibitor bemcentinib (R428/BGB324) has been granted fast track designation by the U.S. Food and Drug Administration (FDA) in STK11-mutated advanced metastatic non-small cell lung cancer and was also reported to show selective sensitivity towards ovarian cancers (OC) with a Mesenchymal molecular subtype. In this study, we further explored AXL's role in mediating DNA damage responses by using OC as a disease model. AXL inhibition using R428 resulted in the increase of DNA damage with the concurrent upregulation of DNA damage response signalling molecules. Furthermore, AXL inhibition rendered cells more sensitive to the inhibition of ATR, a crucial mediator for replication stress. Combinatory use of AXL and ATR inhibitors showed additive effects in OC. Through SILAC co-immunoprecipitation mass spectrometry, we identified a novel binding partner of AXL, SAM68, whose loss in OC cells harboured phenotypes in DNA damage responses similar to AXL inhibition. In addition, AXL- and SAM68-deficiency or R428 treatment induced elevated levels of cholesterol and upregulated genes in the cholesterol biosynthesis pathway. There might be a protective role of cholesterol in shielding cancer cells against DNA damage induced by AXL inhibition or SMA68 deficiency.
Collapse
Affiliation(s)
- Xun Hui Yeo
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Republic of Singapore
| | - Vignesh Sundararajan
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Republic of Singapore
| | - Zhengwei Wu
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Republic of Singapore
| | - Zi Jin Cheryl Phua
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Yin Ying Ho
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros, Singapore, 138668, Republic of Singapore
| | - Kai Lay Esther Peh
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros, Singapore, 138668, Republic of Singapore
| | - Yi-Chia Chiu
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Republic of Singapore
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Republic of Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117597, Republic of Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros, Singapore, 138668, Republic of Singapore
| | - David Shao Peng Tan
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Republic of Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Republic of Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Republic of Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117597, Republic of Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Ruby Yun-Ju Huang
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
14
|
Boos A, Gahr BM, Park DD, Braun V, Bühler A, Rottbauer W, Just S. Hdac1-deficiency affects the cell cycle axis Cdc25-Cdk1 causing impaired G2/M phase progression and reduced cardiomyocyte proliferation in zebrafish. Biochem Biophys Res Commun 2023; 665:98-106. [PMID: 37149988 DOI: 10.1016/j.bbrc.2023.04.116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/11/2023] [Accepted: 04/29/2023] [Indexed: 05/09/2023]
Abstract
Zebrafish have the ability to fully regenerate their hearts after injury since cardiomyocytes subsequently dedifferentiate, re-enter cell cycle, and proliferate to replace damaged myocardial tissue. Recent research identified the reactivation of dormant developmental pathways during cardiac regeneration in adult zebrafish, suggesting pro-proliferative pathways important for developmental heart growth to be also critical for regenerative heart growth after injury. Histone deacetylase 1 (Hdac1) was recently shown to control both, embryonic as well as adult regenerative cardiomyocyte proliferation in the zebrafish model. Nevertheless, regulatory pathways controlled by Hdac1 are not defined yet. By analyzing RNA-seq-derived transcriptional profiles of the Hdac1-deficient zebrafish mutant baldrian, we here identified DNA damage response (DDR) pathways activated in baldrian mutant embryos. Surprisingly, although the DDR signaling pathway was transcriptionally activated, we found the complete loss of protein expression of the known DDR effector and cell cycle inhibitor p21. Consequently, we observed an upregulation of the p21-downstream target Cdk2, implying elevated G1/S phase transition in Hdac1-deficient zebrafish hearts. Remarkably, Cdk1, another p21-but also Cdc25-downstream target was downregulated. Here, we found the significant downregulation of Cdc25 protein expression, explaining reduced Cdk1 levels and suggesting impaired G2/M phase progression in Hdac1-deficient zebrafish embryos. To finally prove defective cell cycle progression due to Hdac1 loss, we conducted Cytometer-based cell cycle analyses in HDAC1-deficient murine HL-1 cardiomyocytes and indeed found impaired G2/M phase transition resulting in defective cardiomyocyte proliferation. In conclusion, our results suggest a critical role of Hdac1 in maintaining both, regular G1/S and G2/M phase transition in cardiomyocytes by controlling the expression of essential cell cycle regulators such as p21 and Cdc25.
Collapse
Affiliation(s)
- Alena Boos
- Molecular Cardiology, Department of Internal Medicine II, Ulm University, Ulm, Germany
| | - Bernd Martin Gahr
- Molecular Cardiology, Department of Internal Medicine II, Ulm University, Ulm, Germany
| | - Deung-Dae Park
- Molecular Cardiology, Department of Internal Medicine II, Ulm University, Ulm, Germany
| | - Verena Braun
- Molecular Cardiology, Department of Internal Medicine II, Ulm University, Ulm, Germany
| | - Anja Bühler
- Molecular Cardiology, Department of Internal Medicine II, Ulm University, Ulm, Germany
| | | | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, Ulm University, Ulm, Germany.
| |
Collapse
|
15
|
Yuan B, Kikuchi H, Li J, Kawabata A, Yao K, Takagi N, Okazaki M. Cytotoxic Effects of Darinaparsin, a Novel Organic Arsenical, against Human Leukemia Cells. Int J Mol Sci 2023; 24:2282. [PMID: 36768603 PMCID: PMC9916914 DOI: 10.3390/ijms24032282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
To explore the molecular mechanisms of action underlying the antileukemia activities of darinaparsin, an organic arsenical approved for the treatment of peripheral T-cell lymphoma in Japan, cytotoxicity of darinaparsin was evaluated in leukemia cell lines NB4, U-937, MOLT-4 and HL-60. Darinaparsin was a more potent cytotoxic than sodium arsenite, and induced apoptosis/necrosis in NB4 and HL-60 cells. In NB4 cells exhibiting the highest susceptibility to darinaparsin, apoptosis induction was accompanied by the activation of caspase-8/-9/-3, a substantial decrease in Bid expression, and was suppressed by Boc-D-FMK, a pancaspase inhibitor, suggesting that darinaparsin triggered a convergence of the extrinsic and intrinsic pathways of apoptosis via Bid truncation. A dramatic increase in the expression level of γH2AX, a DNA damage marker, occurred in parallel with G2/M arrest. Activation of p53 and the inhibition of cdc25C/cyclin B1/cdc2 were concomitantly observed in treated cells. Downregulation of c-Myc, along with inactivation of E2F1 associated with the activation of Rb, was observed, suggesting the critical roles of p53 and c-Myc in darinaparsin-mediated G2/M arrest. Trolox, an antioxidative reagent, suppressed the apoptosis induction but failed to correct G2/M arrest, suggesting that oxidative stress primarily contributed to apoptosis induction. Suppression of Notch1 signaling was also confirmed. Our findings provide novel insights into molecular mechanisms underlying the cytotoxicity of darinaparsin and strong rationale for its new clinical application for patients with different types of cancer.
Collapse
Affiliation(s)
- Bo Yuan
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| | - Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| | - Jingmei Li
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| | - Atsushi Kawabata
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| | - Kozo Yao
- Product Development Division, Solasia Pharma K.K., Tokyo 105-0011, Japan
| | - Norio Takagi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Hachioji 192-0392, Japan
| | - Mari Okazaki
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| |
Collapse
|
16
|
Mondal P, Natesh J, Penta D, Meeran SM. Extract of Murraya koenigii selectively causes genomic instability by altering redox-status via targeting PI3K/AKT/Nrf2/caspase-3 signaling pathway in human non-small cell lung cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154272. [PMID: 35728387 DOI: 10.1016/j.phymed.2022.154272] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/01/2022] [Accepted: 06/11/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related death worldwide. Dietary bioactives have been used as alternative therapeutics to overcome various adverse effects caused by chemotherapeutics. Curry leaves are a widely used culinary spice and different parts of this plant have been used in traditional medicines. Curry leaves are a rich source of multiple bioactives, especially polyphenols and alkaloids. Therefore, extraction processes play a key role in obtaining the optimum yield of bioactives and their efficacy. PURPOSE We aim to select an extraction process that achieves the optimum yield of bioactives in curry leaves crude extract (CLCE) with minimum solvent usage and in a shorter time. Further, to investigate the anticancer properties of CLCE and its mechanism against lung cancer. METHODS Different extraction processes were performed and analyzed polyphenol content. The bioactives and essential oils present in curry leaves were identified through LC-MS/MS and GC-MS analysis. The cytotoxicity of microwave-assisted CLCE (MA-CLCE) was investigated through MTT and colony-forming assays. The DNA damage was observed by comet assay. The apoptotic mechanisms of MA-CLCE were investigated by estimating ROS production, depolarization of mitochondrial membrane potential (MMP), and apoptotic proteins. The glutathione assay estimated the antioxidant potential of MA-CLCE in normal cells. RESULTS Generally, conventional extraction methods require high temperatures, extra energy input, and time. Recently, green extraction processes are getting wider attention as alternative extraction methods. This study compared different extraction processes and found that the microwave-assisted extraction (MAE) method yields the highest polyphenols from curry leaves among other extraction processes with minimum processing. The MA-CLCE functions as an antioxidant under normal physiological conditions but pro-oxidant to cancer cells. MA-CLCE scavenges free radicals and enhances the intracellular GSH level in alveolar macrophages in situ. We found that MA-CLCE selectively inhibits cell proliferation and induces apoptosis in cancer cells by altering cellular redox status. MA-CLCE induces chromatin condensation and genotoxicity through ROS-induced depolarization of MMP. The depolarization of MMP causes the release of cytochrome c into the cytosol and activates the apoptotic pathway in lung cancer cells. However, pretreatment with ascorbic acid, an antioxidant, inhibits the MA-CLCE-induced apoptosis by reducing ROS production, which impedes mitochondrial membrane disruption, preventing BAX/BCL-2 expression alteration. Simultaneously, MA-CLCE downregulates the expression of survival signaling regulator PI3K/AKT, which modulates Nrf-2. MA-CLCE also diminishes intracellular antioxidant proficiency by suppressing Nrf-2 expression, followed by HO-1 expressions. CONCLUSION Among several extraction methods, MA-CLCE is rich in several bioactives, especially polyphenols, alkaloids, and essential oils. Here, we reported for the first time that MA-CLCE functions as a pro-oxidant to lung cancer cells and acts as an antioxidant to normal cells by regulating different cellular programs and signaling pathways. Therefore, it can be further developed as a promising phytomedicine against lung cancer.
Collapse
Affiliation(s)
- Priya Mondal
- Department of Biochemistry, Laboratory of Nutritional Epigenetics, CSIR-Central Food Technological Research Institute, Mysore, Karnataka 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jagadish Natesh
- Department of Biochemistry, Laboratory of Nutritional Epigenetics, CSIR-Central Food Technological Research Institute, Mysore, Karnataka 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dhanamjai Penta
- Department of Biochemistry, Laboratory of Nutritional Epigenetics, CSIR-Central Food Technological Research Institute, Mysore, Karnataka 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Syed Musthapa Meeran
- Department of Biochemistry, Laboratory of Nutritional Epigenetics, CSIR-Central Food Technological Research Institute, Mysore, Karnataka 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
17
|
Integration of synthetic and natural derivatives revives the therapeutic potential of temozolomide against glioma- an in vitro and in vivo perspective. Life Sci 2022; 301:120609. [DOI: 10.1016/j.lfs.2022.120609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/02/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
|
18
|
Zerboni A, Rossi T, Bengalli R, Catelani T, Rizzi C, Priola M, Casadei S, Mantecca P. Diesel exhaust particulate emissions and in vitro toxicity from Euro 3 and Euro 6 vehicles. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 297:118767. [PMID: 34974087 DOI: 10.1016/j.envpol.2021.118767] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Incomplete combustion processes in diesel engines produce particulate matter (PM) that significantly contributes to air pollution. Currently, there remains a knowledge gap in relation to the physical and chemical characteristics and also the biological reactivity of the PM emitted from old- and new-generation diesel vehicles. In this study, the emissions from a Euro 3 diesel vehicle were compared to those from a Euro 6 car during the regeneration of a diesel particulate filter (DPF). Different driving cycles were used to collect two types of diesel exhaust particles (DEPs). The particle size distribution was monitored using an engine exhaust particle sizer spectrometer and an electrical low-pressure impactor. Although the Euro 6 vehicle emitted particulates only during DPF regeneration that primarily occurs for a few minutes at high speeds, such emissions are characterized by a higher number of ultrafine particles (<0.1 μm) compared to those from the Euro 3 diesel vehicle. The emitted particles possess different characteristics. For example, Euro 6 DEPs exhibit a lower PAH content than do Euro 3 samples; however, they are enriched in metals that were poorly detected or undetected in Euro 3 emissions. The biological effects of the two DEPs were investigated in human bronchial BEAS-2B cells exposed to 50 μg/mL of PM (corresponding to 5.2 μg/cm2), and the results revealed that Euro 3 DEPs activated the typical inflammatory and pro-carcinogenic pathways induced by combustion-derived particles, while Euro 6 DEPs were less effective in regard to activating such biological responses. Although further investigations are required, it is evident that the different in vitro effects elicited by Euro 3 and Euro 6 DEPs can be correlated with the variable chemical compositions (metals and PAHs) of the emitted particles that play a pivotal role in the inflammatory and carcinogenic potential of airborne PM.
Collapse
Affiliation(s)
- Alessandra Zerboni
- Polaris Research Centre, Dept. of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, 20126, Milan, Italy.
| | - Tommaso Rossi
- Innovhub-SSI Fuels Department, Via Galileo Galilei, 1, 20097, San Donato Milanese, Milan, Italy
| | - Rossella Bengalli
- Polaris Research Centre, Dept. of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, 20126, Milan, Italy
| | - Tiziano Catelani
- Microscopy Facility, University of Milano-Bicocca, Piazza della Scienza 3, 20126, Milano, Italy
| | - Cristiana Rizzi
- Dept. of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, 20126, Milan, Italy
| | - Marco Priola
- Innovhub-SSI Fuels Department, Via Galileo Galilei, 1, 20097, San Donato Milanese, Milan, Italy
| | - Simone Casadei
- Innovhub-SSI Fuels Department, Via Galileo Galilei, 1, 20097, San Donato Milanese, Milan, Italy
| | - Paride Mantecca
- Polaris Research Centre, Dept. of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, 20126, Milan, Italy
| |
Collapse
|
19
|
Abdallah AE, Mabrouk RR, Elnagar MR, Farrag AM, Kalaba MH, Sharaf MH, El-Fakharany EM, Bakhotmah DA, Elkaeed EB, Al Ward MMS. New Series of VEGFR-2 Inhibitors and Apoptosis Enhancers: Design, Synthesis and Biological Evaluation. Drug Des Devel Ther 2022; 16:587-606. [PMID: 35281317 PMCID: PMC8904266 DOI: 10.2147/dddt.s344750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/28/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Cancer is still a major world health threat, causing a high rate of mortality. VEGFR-2 inhibitor anticancer agents are of great significance. However, they showed some serious side effects. PURPOSE To discover new effective and safer anticancer agents, a new series of piperazinylquinoxaline-based derivatives was designed and synthesized on the basis of the pharmacophoric features of VEGFR-2 inhibitor drugs. METHODS The new candidates were evaluated against A549 lung cancer cells, HepG-2 hepatoma cells, Caco-2 colon cancer cells, MDA breast cancer cells, and VEGFR-2 kinase. Moreover, cell cycle kinetics and apoptosis rates were studied in HepG-2 cells treated with compound 11, which was the most promising candidate. RESULTS The new derivatives revealed better antitumor results (IC50 from 6.48 to 38.58 µM) against the aforementioned cancer cell lines than sorafenib. Also, the new candidates showed VEGFR-2 inhibition with IC50 values ranging from 0.19 to 0.60 µM compared to 0.08 µM for sorafenib. Compound 11, meanwhile, showed IC50 values equal to 10.61, 9.52, 12.45, 11.52, and 0.19 µM against the cancer cell lines and VEGFR-2, respectively. Moreover, compound 11 raised the apoptosis rate in HepG-2 cells from 5% to 44% and caused 4, 2.3, and 3-fold increases in BAX/Bcl-2 ratio, caspase-3 level, and P53 expression, respectively, compared to control untreated cells. Finally, the new derivatives displayed the correct binding mode into VEGFR-2 kinase pocket, giving interactions with the essential residues. CONCLUSION This work suggests that compound 11 is a very significant anticancer candidate, and piperazinylquinoxaline is an important scaffold in the development of new potential effective and safer VEGFR-2 inhibitor agents.
Collapse
Affiliation(s)
- Abdallah E Abdallah
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Reda R Mabrouk
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Mohamed R Elnagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, 11884, Egypt
| | - Amel Mostafa Farrag
- Pharmaceutical Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Mohamed H Kalaba
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, 11884, Egypt
| | - Mohamed H Sharaf
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, 11884, Egypt
| | - Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg El Arab, Egypt
| | - Dina Abed Bakhotmah
- Chemistry Department, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, 13713, Saudi Arabia
| | - Maged Mohammed Saleh Al Ward
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| |
Collapse
|
20
|
Biological evaluation of complexes of cyclopentadienyl M(CO) 3+ (M = Re, 99mTc) with high blood-brain barrier penetration potential as brain cancer agents. Invest New Drugs 2022; 40:497-505. [PMID: 35024984 DOI: 10.1007/s10637-022-01211-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
To address the major medical need for effective chemotherapeutics/diagnostics for brain cancer, in this work three cyclopentadienyl M(CO)3+ (M = Re, 99mTc) complexes, which cross the blood-brain barrier (BBB) in high % and are designed to mimic the anticancer agent 2-phenylbenzothiazole, are in vitro and in vivo evaluated for anticancer action. The study includes cytotoxicity and uptake studies in cancer and healthy neuronal cell lines, mechanistic investigation of potential anticancer pathways, and biodistribution studies in mice bearing glioblastoma xenografts. The stable Re complexes exhibit selective uptake and significant antiproliferative effect, particularly against U-251 MG glioblastoma cells, with no significant toxicity in healthy neurons, demonstrating the suitability of this type of complexes to serve as selective therapeutic/imaging agents for brain cancer. Furthermore, they result in the generation of elevated Reactive Oxygen Species (ROS) levels, and lead to significant G2/M arrest followed by apoptosis. Biodistribution studies in U-251 MG xenograft bearing mice with the radioactive 99mTc complex that exhibits the highest BBB penetration, show retention at the tumor-site offering a diagnostic prospect and, in addition, indicating the capability of the Re analogue to accumulate at the tumor site for therapeutic action. Overall, the complexes demonstrate significant anticancer properties that, combined with their high BBB penetration potential, render them strong candidates for further evaluation as brain cancer agents.
Collapse
|
21
|
Sp N, Kang DY, Jo ES, Lee JM, Bae SW, Jang KJ. Pivotal Role of Iron Homeostasis in the Induction of Mitochondrial Apoptosis by 6-Gingerol Through PTEN Regulated PD-L1 Expression in Embryonic Cancer Cells. Front Oncol 2021; 11:781720. [PMID: 34804985 PMCID: PMC8595921 DOI: 10.3389/fonc.2021.781720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/15/2021] [Indexed: 01/07/2023] Open
Abstract
Embryonic cancer stem cells (CSCs) can differentiate into any cancer type. Targeting CSCs with natural compounds is a promising approach as it suppresses cancer recurrence with fewer adverse effects. 6-Gingerol is an active component of ginger, which exhibits well-known anti-cancer activities. This study determined the mechanistic aspects of cell death induction by 6-gingerol. To analyze cellular processes, we used Western blot and real-time qPCR for molecular signaling studies and conducted flow cytometry. Our results suggested an inhibition of CSC marker expression and Wnt/β-catenin signaling by 6-gingerol in NCCIT and NTERA-2 cells. 6-Gingerol induced reactive oxygen species generation, the DNA damage response, cell cycle arrest, and the intrinsic pathway of apoptosis in embryonic CSCs. Furthermore, 6-gingerol inhibited iron metabolism and induced PTEN, which both played vital roles in the induction of cell death. The activation of PTEN resulted in the inhibition of PD-L1 expression through PI3K/AKT/p53 signaling. The induction of PTEN also mediated the downregulation of microRNAs miR-20b, miR-21, and miR-130b to result in PD-L1 suppression by 6-gingerol. Hence, 6-gingerol may be a promising candidate to target CSCs by regulating PTEN-mediated PD-L1 expression.
Collapse
Affiliation(s)
- Nipin Sp
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, South Korea
| | - Dong Young Kang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, South Korea
| | - Eun Seong Jo
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju-si, South Korea
| | - Jin-Moo Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju-si, South Korea.,SK Bioscience, Seongnam-si, South Korea
| | - Se Won Bae
- Department of Chemistry and Cosmetics, Jeju National University, Jeju, South Korea
| | - Kyoung-Jin Jang
- Department of Pathology, School of Medicine, Institute of Biomedical Science and Technology, Konkuk University, Chungju, South Korea
| |
Collapse
|
22
|
Jaiswal SK, Raj S, DePamphilis ML. Developmental Acquisition of p53 Functions. Genes (Basel) 2021; 12:genes12111675. [PMID: 34828285 PMCID: PMC8622856 DOI: 10.3390/genes12111675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Remarkably, the p53 transcription factor, referred to as “the guardian of the genome”, is not essential for mammalian development. Moreover, efforts to identify p53-dependent developmental events have produced contradictory conclusions. Given the importance of pluripotent stem cells as models of mammalian development, and their applications in regenerative medicine and disease, resolving these conflicts is essential. Here we attempt to reconcile disparate data into justifiable conclusions predicated on reports that p53-dependent transcription is first detected in late mouse blastocysts, that p53 activity first becomes potentially lethal during gastrulation, and that apoptosis does not depend on p53. Furthermore, p53 does not regulate expression of genes required for pluripotency in embryonic stem cells (ESCs); it contributes to ESC genomic stability and differentiation. Depending on conditions, p53 accelerates initiation of apoptosis in ESCs in response to DNA damage, but cell cycle arrest as well as the rate and extent of apoptosis in ESCs are p53-independent. In embryonic fibroblasts, p53 induces cell cycle arrest to allow repair of DNA damage, and cell senescence to prevent proliferation of cells with extensive damage.
Collapse
Affiliation(s)
- Sushil K. Jaiswal
- National Institute of Child Health and Human Development, Bethesda, MD 20892, USA;
- National Human Genome Research Institute, Bethesda, MD 20892, USA
| | - Sonam Raj
- National Cancer Institute, Bethesda, MD 20892, USA;
| | - Melvin L. DePamphilis
- National Institute of Child Health and Human Development, Bethesda, MD 20892, USA;
- Correspondence:
| |
Collapse
|
23
|
Sp N, Kang DY, Jo ES, Lee JM, Jang KJ. Iron Metabolism as a Potential Mechanism for Inducing TRAIL-Mediated Extrinsic Apoptosis Using Methylsulfonylmethane in Embryonic Cancer Stem Cells. Cells 2021; 10:cells10112847. [PMID: 34831070 PMCID: PMC8616102 DOI: 10.3390/cells10112847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/11/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
Embryonic cancer stem cells (CSCs) can differentiate into any cancer type. Targeting CSC using natural compounds is a good approach as it suppresses cancer recurrence with fewer adverse effects, and methylsulfonylmethane (MSM) is a sulfur-containing compound with well-known anticancer activities. This study determined the mechanistic aspects of the anticancer activity of MSM. We used Western blotting and real-time qPCR for molecular signaling studies and conducted flow cytometry for analyzing the processes in cells. Our results suggested an inhibition in the expression of CSC markers and Wnt/β-catenin signaling. MSM induced TRAIL-mediated extrinsic apoptosis in NCCIT and NTERA-2 cells rather than an intrinsic pathway. Inhibition of iron metabolism-dependent reactive oxygen species (ROS) generation takes part in TRAIL-mediated apoptosis induction by MSM. Suppressing iron metabolism by MSM also regulated p38/p53/ERK signaling and microRNA expressions, such as upregulating miR-130a and downregulating miR-221 and miR-222, which resulted in TRAIL induction and thereby extrinsic pathway of apoptosis. Hence, MSM could be a good candidate for neoadjuvant therapy by targeting CSCs by inhibiting iron metabolism.
Collapse
Affiliation(s)
- Nipin Sp
- Department of Pathology, Institute of Biomedical Science and Technology, School of Medicine, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.)
| | - Dong Young Kang
- Department of Pathology, Institute of Biomedical Science and Technology, School of Medicine, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.)
| | - Eun Seong Jo
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju-si 28159, Korea; (E.S.J.); (J.-M.L.)
| | - Jin-Moo Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation, Osong Health Technology Administration Complex, Cheongju-si 28159, Korea; (E.S.J.); (J.-M.L.)
- SK Bioscience, Seongnam-si 13493, Korea
| | - Kyoung-Jin Jang
- Department of Pathology, Institute of Biomedical Science and Technology, School of Medicine, Konkuk University, Chungju 27478, Korea; (N.S.); (D.Y.K.)
- Correspondence: ; Tel.: +82-2-2030-7839
| |
Collapse
|
24
|
Conedera FM, Pousa AMQ, Mercader N, Tschopp M, Enzmann V. The TGFβ/Notch axis facilitates Müller cell-to-epithelial transition to ultimately form a chronic glial scar. Mol Neurodegener 2021; 16:69. [PMID: 34593012 PMCID: PMC8482586 DOI: 10.1186/s13024-021-00482-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/13/2021] [Indexed: 01/04/2023] Open
Abstract
Background Contrasting with zebrafish, retinal regeneration from Müller cells (MCs) is largely limited in mammals, where they undergo reactive gliosis that consist of a hypertrophic response and ultimately results in vision loss. Transforming growth factor β (TGFβ) is essential for wound healing, including both scar formation and regeneration. However, targeting TGFβ may affect other physiological mechanisms, owing its pleiotropic nature. The regulation of various cellular activities by TGFβ relies on its interaction with other pathways including Notch. Here, we explore the interplay of TGFβ with Notch and how this regulates MC response to injury in zebrafish and mice. Furthermore, we aimed to characterize potential similarities between murine and human MCs during chronic reactive gliosis. Methods Focal damage to photoreceptors was induced with a 532 nm diode laser in TgBAC (gfap:gfap-GFP) zebrafish (ZF) and B6-Tg (Rlbp1-GFP) mice. Transcriptomics, immunofluorescence, and flow cytometry were employed for a comparative analysis of MC response to laser-induced injury between ZF and mouse. The laser-induced injury was paired with pharmacological treatments to inhibit either Notch (DAPT) or TGFβ (Pirfenidone) or TGFβ/Notch interplay (SIS3). To determine if the murine laser-induced injury model translates to the human system, we compared the ensuing MC response to human donors with early retinal degeneration. Results Investigations into injury-induced changes in murine MCs revealed TGFβ/Notch interplay during reactive gliosis. We found that TGFβ1/2 and Notch1/2 interact via Smad3 to reprogram murine MCs towards an epithelial lineage and ultimately to form a glial scar. Similar to what we observed in mice, we confirmed the epithelial phenotype of human Müller cells during gliotic response. Conclusion The study indicates a pivotal role for TGFβ/Notch interplay in tuning MC stemness during injury response and provides novel insights into the remodeling mechanism during retinal degenerative diseases. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00482-z.
Collapse
Affiliation(s)
- Federica Maria Conedera
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.,Advanced Microscopy Program, Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA.,Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ana Maria Quintela Pousa
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Markus Tschopp
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of Ophthalmology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Volker Enzmann
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland. .,Department of BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
25
|
Stanic M, Mekhail K. Integration of DNA damage responses with dynamic spatial genome organization. Trends Genet 2021; 38:290-304. [PMID: 34598804 DOI: 10.1016/j.tig.2021.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 11/28/2022]
Abstract
The maintenance of genome stability and cellular homeostasis depends on the temporal and spatial coordination of successive events constituting the classical DNA damage response (DDR). Recent findings suggest close integration and coordination of DDR signaling with specific cellular processes. The mechanisms underlying such coordination remain unclear. We review emerging crosstalk between DNA repair factors, chromatin remodeling, replication, transcription, spatial genome organization, cytoskeletal forces, and liquid-liquid phase separation (LLPS) in mediating DNA repair. We present an overarching DNA repair framework within which these dynamic processes intersect in nuclear space over time. Collectively, this interplay ensures the efficient assembly of DNA repair proteins onto shifting genome structures to preserve genome stability and cell survival.
Collapse
Affiliation(s)
- Mia Stanic
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, MaRS Centre, West Tower, 661 University Avenue, Toronto, ON M5G 1M1, Canada
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, MaRS Centre, West Tower, 661 University Avenue, Toronto, ON M5G 1M1, Canada; Canada Research Chairs Program, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
26
|
Mohiuddin M, Kasahara K. Paclitaxel Impedes EGFR-mutated PC9 Cell Growth via Reactive Oxygen Species-mediated DNA Damage and EGFR/PI3K/AKT/mTOR Signaling Pathway Suppression. Cancer Genomics Proteomics 2021; 18:645-659. [PMID: 34479917 DOI: 10.21873/cgp.20287] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND/AIM Paclitaxel is used as a first-line and subsequent therapy for the treatment of various cancers. However, the function and mechanisms of action of paclitaxel in non-small-cell lung cancer (NSCLC) remain unknown. In this study, the molecular mechanism underlying the anticancer activity of paclitaxel was investigated in vitro in a human NSCLC cell line carrying the EGFR exon 19 deletion (PC9). MATERIALS AND METHODS PC9 cells were treated with paclitaxel and then evaluated with a cell viability assay, DAPI staining, Giemsa staining, apoptosis assay, reactive oxygen species (ROS) assay, terminal deoxynucleotidyl transferase dUTP nick-end labeling assay and Western blotting. RESULTS Paclitaxel markedly decreased the viability of PC9 cells and induced morphological signs of apoptosis. The apoptotic effects of paclitaxel were observed through caspase cascade activation, along with ROS generation and loss of mitochondrial membrane potential (MMP). Furthermore, paclitaxel induced ROS-mediated DNA damage that triggered the activation of the extrinsic pathway of apoptosis via the up-regulation of death receptor (DR5) and caspase-8 activation. In addition, we found that paclitaxel effectively suppressed the EGFR/PI3K/AKT/mTOR signaling pathway to impede PC9 cell growth. Paclitaxel induced cell cycle arrest at the G1 phase in response to DNA damage, in association with the suppression of CDC25A, Cdk2 and Cyclin E1 protein expression. CONCLUSION Paclitaxel showed anticancer effects against NSCLC by activating extrinsic and intrinsic apoptotic pathways through enhancing ROS generation, inducing cell cycle arrest, and suppressing EGFR/PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Md Mohiuddin
- Department of Respiratory Medicine, Kanazawa University, Ishikawa, Japan
| | - Kazuo Kasahara
- Department of Respiratory Medicine, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
27
|
Barnoud T, Indeglia A, Murphy ME. Shifting the paradigms for tumor suppression: lessons from the p53 field. Oncogene 2021; 40:4281-4290. [PMID: 34103683 PMCID: PMC8238873 DOI: 10.1038/s41388-021-01852-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 01/20/2023]
Abstract
The TP53 gene continues to hold distinction as the most frequently mutated gene in cancer. Since its discovery in 1979, hundreds of research groups have devoted their efforts toward understanding why this gene is so frequently selected against by tumors, with the hopes of harnessing this information toward improved therapy of cancer. The result is that this protein has been meticulously analyzed in tumor and normal cells, resulting in over one hundred thousand publications, with an average of five thousand papers published on p53 every year for the past decade. The journey toward understanding p53 function has been anything but straightforward; in fact, the field is notable for the numerous times that established paradigms not only have been shifted, but in fact have been shattered or reversed. In this review, we will discuss the manuscripts, or series of manuscripts, that have most radically changed our thinking about how this tumor suppressor functions, and we will delve into the emerging challenges for the future in this important area of research. It is hoped that this review will serve as a useful historical reference for those interested in p53, and a useful lesson on the need to be flexible in the face of established paradigms.
Collapse
Affiliation(s)
- Thibaut Barnoud
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Alexandra Indeglia
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA.,Department of Biochemistry and Molecular Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Maureen E Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Hur S, Jang E, Lee JH. Beneficial Actions of Orostachys japonica and Its Compounds against Tumors via MAPK Signaling Pathways. Nutrients 2021; 13:nu13020555. [PMID: 33567572 PMCID: PMC7915109 DOI: 10.3390/nu13020555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/30/2022] Open
Abstract
Tumors are one of the most life-threatening diseases, and a variety of cancer treatment options have been continuously introduced in order to overcome cancer and improve conventional therapy. Orostachys japonica (O. japonica), which is a perennial plant belonging to the genus Orostachys of the Crassulaceae family, has been revealed to exhibit pharmacological properties against various tumors in numerous studies. The present review aimed to discuss the biological actions and underlying molecular mechanisms of O. japonica and its representative compounds-kaempferol and quercetin-against tumors. O. japonica reportedly has antiproliferative, anti-angiogenic, and antimetastatic activities against various types of malignant tumors through the induction of apoptosis and cell cycle arrest, a blockade of downstream vascular endothelial growth factor (VEGF)-VEGFR2 pathways, and the regulation of epithelial-to-mesenchymal transition. In addition, emerging studies have highlighted the antitumor efficacy of kaempferol and quercetin. Interestingly, it was found that alterations of the mitogen-activated protein kinase (MAPK) signaling cascades are involved in the pivotal mechanisms of the antitumor effects of O. japonica and its two compounds against cancer cell overgrowth, angiogenesis, and metastasis. In summary, O. japonica could be considered a preventive and therapeutic medicinal plant which exhibits antitumor actions by reversing altered patterns of MAPK cascades, and kaempferol and quercetin might be potential components that can contribute to the efficacy and underlying mechanism of O. japonica.
Collapse
Affiliation(s)
- Soyoung Hur
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - Eungyeong Jang
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
- Department of Internal Medicine, Kyung Hee University Korean Medicine Hospital, Seoul 02447, Korea
| | - Jang-Hoon Lee
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
- Correspondence: ; Tel.: +82-2-958-9118; Fax: +82-2-958-9258
| |
Collapse
|
29
|
Balagholi S, Aletaha M, Kanavi MR, Karami S, Dabbaghi R, Nakhaie PA. Optimizing the effective doses of mitomycin C, 5-fluorouracil, and their combination on cultivated basal cell carcinoma. Arq Bras Oftalmol 2021; 84:481-489. [PMID: 33567038 PMCID: PMC11878417 DOI: 10.5935/0004-2749.20210049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/21/2020] [Indexed: 11/20/2022] Open
Abstract
PURPOSE This study aimed to optimize the effective doses of mitomycin C, 5-fluorouracil, and their combination on cultivated basal cell carcinoma. METHODS Cultivated basal cell carcinoma and fibroblastic cells were treated with different concentrations of mitomycin C, 5-fluorouracil, and their combination. Cell viability, cell cycle, apoptosis, and expression levels of TP53, CDKN1A, and CDK6 were investigated. The most effective drug with its optimum dosage was administered via multiple intralesional injections to a 65-year-old woman with advanced periorbital nodulo-ulcerative BCC. RESULTS The concentrations of 0.00312 and 0.312 mg/mL were considered optimum for mitomycin C and 5-fluorouracil, respectively. The mean viabilities of basal cell carcinoma treated with mitomycin C alone and its combination with 5-fluorouracil were significantly less than those of the controls (p=0.002 and p=0.04, respectively). The cell cycle of all the treated basal cell carcinoma groups was arrested in the S phase. The apoptotic rates (p=0.002) of mitomycin C treated basal cell carcinoma were higher than those of the other treated cells, and their TP53 was significantly upregulated (p=0.0001). Moreover, CDKN1A was upregulated, whereas CDK6 was downregulated in basal cell carcinoma treated with either 5-fluorouracil (p=0.0001 and p=0.01, respectively) or the combination of 5-fluorouracil and mitomycin C (p=0.007 and p=0.001, respectively). Basal cell carcinoma lesions were significantly alleviated following mitomycin C injections in the reported patient. CONCLUSION Our in vitro results revealed that the effective doses of mitomycin C and 5-fluorouracil on cultivated basal cell carcinoma were optimized. Mitomycin C was more effective in inducing the apoptosis of basal cell carcinoma than 5-fluorouracil and their combination. The intralesional injections of the optimum dose of mitomycin C could be proposed for the nonsurgical treatment of advanced eyelid basal cell carcinoma.
Collapse
Affiliation(s)
- Sahar Balagholi
- Blood Transfusion Research Center, High Institute for Research and
Education in Transfusion Medicine, Tehran, Iran
| | - Maryam Aletaha
- Ophthalmic Research Center, Shahid Beheshti University of Medical
Sciences, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Shahid Beheshti
University of Medical Sciences, Tehran, Iran
| | - Samira Karami
- Ocular Tissue Engineering Research Center, Shahid Beheshti
University of Medical Sciences, Tehran, Iran
| | - Rasul Dabbaghi
- Ocular Tissue Engineering Research Center, Shahid Beheshti
University of Medical Sciences, Tehran, Iran
| | - Parisa Ashtar Nakhaie
- Ophthalmic Research Center, Shahid Beheshti University of Medical
Sciences, Tehran, Iran
| |
Collapse
|
30
|
Hegazy MEF, Dawood M, Mahmoud N, Elbadawi M, Sugimoto Y, Klauck SM, Mohamed N, Efferth T. 2α-Hydroxyalantolactone from Pulicaria undulata: activity against multidrug-resistant tumor cells and modes of action. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153409. [PMID: 33341310 DOI: 10.1016/j.phymed.2020.153409] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/03/2020] [Accepted: 11/10/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Sesquiterpene lactones having α-methylene-γ-lactone moiety are promising natural metabolites showing various biological activity. One of the major metabolites isolated from Pulicaria undulata, 2α-hydroxyalantolactone (PU-1), has not been investigated in detail yet. Multidrug resistance (MDR) represents a major obstacle for cancer chemotherapy and the capability of novel natural products to overcoming MDR is of great interest. PURPOSE Exploring the molecular modes of action for potent natural product metabolites. METHODS The resazurin reduction assay was employed to evaluate the cytotoxicity of PU-1 on sensitive and their corresponding drug-resistant cell lines (overexpressing P-glycoprotein, BCRP, ABCB5, ΔEGFR, or TP53 knockout). Gene expression profiling was performed by transcriptome-wide mRNA microarray in the human CCRF-CEM leukemic cells after treatment with PU-1. The top significantly up- or down-regulated genes were identified by Chipster program and analyzed using Ingenuity Pathway Analysis (IPA) software. Finally, flow cytometry and Western blotting were performed for cell cycle analyses and apoptosis detection. RESULTS The sesquiterpene lactone, PU-1, showed potent cytotoxicity towards the drug-sensitive and -resistant cell lines. Transcriptome-wide mRNA expression profiling and pathway analysis pointed to genes involved in DNA damage response and G2/M cell cycle arrest. G2/M arrest was verified by flow cytometry and further confirmed by the upregulation of p21 and downregulation of p-CDC25C expression in Western blotting. Moreover, the suggested DNA damage checkpoint regulation was confirmed by immunofluorescence and Western blotting by upregulation of pS345 Chk1, p-H3 and γ-H2AX. Furthermore, PU-1 inhibited PI3K/AKT pathway, which is involved in signaling DNA damage and G2/M arrest. Cells ultimately induced apoptosis upon PU-1 treatment. CONCLUSIONS PU-1 is a potent natural product inhibiting otherwise drug-resistant human tumor cell growth through DNA damage, G2/M cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- Mohamed-Elamir F Hegazy
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; Phytochemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Nuha Mahmoud
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Mohamed Elbadawi
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Yoshikazu Sugimoto
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Nagla Mohamed
- Chemistry Department, Faculty of Science, Aswan University, Egypt
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
31
|
Kciuk M, Marciniak B, Mojzych M, Kontek R. Focus on UV-Induced DNA Damage and Repair-Disease Relevance and Protective Strategies. Int J Mol Sci 2020; 21:ijms21197264. [PMID: 33019598 PMCID: PMC7582305 DOI: 10.3390/ijms21197264] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023] Open
Abstract
The protective ozone layer is continually depleting due to the release of deteriorating environmental pollutants. The diminished ozone layer contributes to excessive exposure of cells to ultraviolet (UV) radiation. This leads to various cellular responses utilized to restore the homeostasis of exposed cells. DNA is the primary chromophore of the cells that absorbs sunlight energy. Exposure of genomic DNA to UV light leads to the formation of multitude of types of damage (depending on wavelength and exposure time) that are removed by effectively working repair pathways. The aim of this review is to summarize current knowledge considering cellular response to UV radiation with special focus on DNA damage and repair and to give a comprehensive insight for new researchers in this field. We also highlight most important future prospects considering application of the progressing knowledge of UV response for the clinical control of diverse pathologies.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Doctoral School of Exact and Natural Sciences, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (B.M.); (R.K.)
- Correspondence:
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (B.M.); (R.K.)
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, 3 Maja 54, 08-110 Siedlce, Poland;
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (B.M.); (R.K.)
| |
Collapse
|
32
|
Mehta S, Algie M, Al-Jabry T, McKinney C, Kannan S, Verma CS, Ma W, Zhang J, Bartolec TK, Masamsetti VP, Parker K, Henderson L, Gould ML, Bhatia P, Harfoot R, Chircop M, Kleffmann T, Cohen SB, Woolley AG, Cesare AJ, Braithwaite A. Critical Role for Cold Shock Protein YB-1 in Cytokinesis. Cancers (Basel) 2020; 12:cancers12092473. [PMID: 32882852 PMCID: PMC7565962 DOI: 10.3390/cancers12092473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Y-box-binding protein-1, YB-1, plays an important role in regulating the cell cycle, although precisely how it does the is unknown. Using live cell imaging, we show that YB-1 is essential for initiating the last step of cell division (cytokinesis), required for creation of two daughter cells. Using confocal microscopy we showed that YB-1 regulates the spatial distribution of key proteins essential for cytokinesis to occur and that this required YB-1 to be phosphorylated on several residues. In-silico modeling demonstrated that modifications at these residues resulted in conformational changes in YB-1 protein allowing it to interact with proteins essential for cytokinesis. As many cancers have high levels YB-1 and these are associated with poor prognosis, our data suggest developing small molecule inhibitors to block YB-1 phosphorylation could be a novel approach to cancer therapy. Abstract High levels of the cold shock protein Y-box-binding protein-1, YB-1, are tightly correlated with increased cell proliferation and progression. However, the precise mechanism by which YB-1 regulates proliferation is unknown. Here, we found that YB-1 depletion in several cancer cell lines and in immortalized fibroblasts resulted in cytokinesis failure and consequent multinucleation. Rescue experiments indicated that YB-1 was required for completion of cytokinesis. Using confocal imaging we found that YB-1 was essential for orchestrating the spatio-temporal distribution of the microtubules, β-actin and the chromosome passenger complex (CPC) to define the cleavage plane. We show that phosphorylation at six serine residues was essential for cytokinesis, of which novel sites were identified using mass spectrometry. Using atomistic modelling we show how phosphorylation at multiple sites alters YB-1 conformation, allowing it to interact with protein partners. Our results establish phosphorylated YB-1 as a critical regulator of cytokinesis, defining precisely how YB-1 regulates cell division.
Collapse
Affiliation(s)
- Sunali Mehta
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Maurice Wilkins Centre for Biodiscovery, University of Otago, 9016 Dunedin, New Zealand
- Correspondence: ; Tel.: +64-3-4797169
| | - Michael Algie
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Centre for Protein Research, Department of Biochemistry, University of Otago, 9054 Dunedin, New Zealand;
| | - Tariq Al-Jabry
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Cushla McKinney
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Srinivasaraghavan Kannan
- Department of Biomolecular Modelling and Design, Bioinformatics Institute (A*STAR), 30 Biopolis Street, 07-01 Matrix, Singapore 138671, Singapore; (S.K.); (C.S.V.)
| | - Chandra S Verma
- Department of Biomolecular Modelling and Design, Bioinformatics Institute (A*STAR), 30 Biopolis Street, 07-01 Matrix, Singapore 138671, Singapore; (S.K.); (C.S.V.)
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117543, Singapore
| | - Weini Ma
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Jessie Zhang
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Tara K. Bartolec
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - V. Pragathi Masamsetti
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Kim Parker
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Luke Henderson
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Maurice Wilkins Centre for Biodiscovery, University of Otago, 9016 Dunedin, New Zealand
| | - Maree L Gould
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Puja Bhatia
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Rhodri Harfoot
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
| | - Megan Chircop
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Torsten Kleffmann
- Centre for Protein Research, Department of Biochemistry, University of Otago, 9054 Dunedin, New Zealand;
| | - Scott B Cohen
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Adele G Woolley
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Maurice Wilkins Centre for Biodiscovery, University of Otago, 9016 Dunedin, New Zealand
| | - Anthony J Cesare
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
| | - Antony Braithwaite
- Department of Pathology, University of Otago, 9016 Dunedin, New Zealand; (M.A.); (C.M.); (K.P.); (L.H.); (M.L.G.); (P.B.); (R.H.); (A.G.W.); (A.B.)
- Maurice Wilkins Centre for Biodiscovery, University of Otago, 9016 Dunedin, New Zealand
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW 2145, Australia; (T.S.-J.); (W.M.); (J.Z.); (T.K.B.); (V.P.M.); (M.C.); (S.B.C.); (A.J.C.)
- Malaghan Institute of Medical Research, 6242 Wellington, New Zealand
| |
Collapse
|
33
|
Solek P, Koszla O, Mytych J, Badura J, Chelminiak Z, Cuprys M, Fraczek J, Tabecka-Lonczynska A, Koziorowski M. Neuronal life or death linked to depression treatment: the interplay between drugs and their stress-related outcomes relate to single or combined drug therapies. Apoptosis 2020; 24:773-784. [PMID: 31278507 PMCID: PMC6711955 DOI: 10.1007/s10495-019-01557-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Depression is a serious medical condition, typically treated by antidepressants. Conventional monotherapy can be effective only in 60–80% of patients, thus modern psychiatry deals with the challenge of new methods development. At the same moment, interactions between antidepressants and the occurrence of potential side effects raise serious concerns, which are even more exacerbated by the lack of relevant data on exact molecular mechanisms. Therefore, the aims of the study were to provide up-to-date information on the relative mechanisms of action of single antidepressants and their combinations. In this study, we evaluated the effect of single and combined antidepressants administration on mouse hippocampal neurons after 48 and 96 h in terms of cellular and biochemical features in vitro. We show for the first time that co-treatment with amitriptyline/imipramine + fluoxetine initiates in cells adaptation mechanisms which allow cells to adjust to stress and finally exerts less toxic events than in cells treated with single antidepressants. Antidepressants treatment induces in neuronal cells oxidative and nitrosative stress, which leads to micronuclei and double-strand DNA brakes formation. At this point, two different mechanistic events are initiated in cells treated with single and combined antidepressants. Single antidepressants (amitriptyline, imipramine or fluoxetine) activate cell cycle arrest resulting in proliferation inhibition. On the other hand, treatment with combined antidepressants (amitriptyline/imipramine + fluoxetine) initiates p16-dependent cell cycle arrest, overexpression of telomere maintenance proteins and finally restoration of proliferation. In conclusion, our findings may pave the way to better understanding of the stress-related effects on neurons associated with mono- and combined therapy with antidepressants.
Collapse
Affiliation(s)
- Przemyslaw Solek
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland.
| | - Oliwia Koszla
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland.,Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Lab, Faculty of Pharmacy with Division of Medical Analytics, Medical University of Lublin, Chodzki 4A, 20-093, Lublin, Poland
| | - Jennifer Mytych
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| | - Joanna Badura
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| | - Zaneta Chelminiak
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| | - Magdalena Cuprys
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| | - Joanna Fraczek
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| | - Anna Tabecka-Lonczynska
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| | - Marek Koziorowski
- Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| |
Collapse
|
34
|
The dichotomous effects of caffeine on homologous recombination in mammalian cells. DNA Repair (Amst) 2020; 88:102805. [PMID: 32062581 DOI: 10.1016/j.dnarep.2020.102805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 01/15/2020] [Accepted: 01/19/2020] [Indexed: 11/23/2022]
Abstract
This study was initiated to examine the effects of caffeine on the DNA damage response (DDR) and homologous recombination (HR) in mammalian cells. A 5 mM caffeine treatment caused the cell cycle to stall at G2/M and cells eventually underwent apoptosis. Caffeine exposure also induced a strong DDR along with subsequent activation of wildtype p53 protein. An unexpected observation was the caffeine-induced depletion of Rad51 (and Brca2) proteins. Consequently, caffeine-treated cells were expected to be inefficient in HR. However, a dichotomy in the HR response of cells to caffeine treatment was revealed. Caffeine treatment rendered cells significantly better at performing the nascent DNA synthesis that accompanies the early strand invasion steps of HR. Additionally, caffeine treatment increased chromatin accessibility and elevated the efficiency of illegitimate recombination. Conversely, the increase in nascent DNA synthesis did not translate into a higher number of gene targeting events. Thus, prolonged caffeine exposure stalls the cell cycle, induces a p53-mediated apoptotic response and a down-regulation of critical HR proteins, and for reasons discussed, stimulates early steps of HR, but not the formation of complete recombination products.
Collapse
|
35
|
Liu C, Huang X, Wang P, Pan Y, Cao D, Liu YY, Chen AJ. Interference of Hsp27 Results in Apoptosis Induced by Photodamage via Regulation of Subcellular Localization of p21 in Immortalized Human Keratinocytes. Med Sci Monit 2019; 25:7557-7566. [PMID: 31592001 PMCID: PMC6795107 DOI: 10.12659/msm.917164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Owing to the increased incidence of photodermatosis caused by ultraviolet light in recent years, it is necessary to clarify the mechanisms potential photodamage to the skin and reveal possible therapeutic targets. Heat shock protein 27 (Hsp27) is well known for suppressing apoptosis. The aim of present study was to elucidate possible photoprotective mechanism between Hsp27 and p21 on ultraviolet B (UVB)-induced photodamage. MATERIAL AND METHODS The Hsp27 gene was interfered to assess the expression of its downstream effectors, cell apoptosis, and cell proliferation ability. The cell apoptosis was tested using flow cytometry method. The cell proliferation ability was tested using Cell Counting Kit-8 (CCK-8) assay. The expression of protein was tested using western-blotting method. The expression of mRNA was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR). The subcellular localization was elucidated using immunofluorescence. RESULTS Hsp27 knockdown decreased cell viability and increased the incidence of UVB-induced apoptosis. Compared with control group, activation of phosphorylated-Akt (p-Akt)-dependent pathway resulted in the nuclear accumulation of p21 and suppression of cell proliferation, while promoting apoptosis in Hsp27 knockdown group. In addition, Hsp27 knockdown increased p53 expression and the Bax: Bcl-2 ratio, which further accelerated the apoptotic process. CONCLUSIONS These findings complemented the mechanism of skin photodamage and demonstrated the photoprotective mechanisms of Hsp27 in HaCaT cells, which might implicate a potential therapeutic target of photodamage and photodermatosis.
Collapse
Affiliation(s)
- Chuan Liu
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China (mainland)
| | - Xin Huang
- Prescriptions Department, College of Traditional Chinese Medicine, Chongqing Medical University, Yuzhong, Chongqing, China (mainland)
| | - Ping Wang
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China (mainland)
| | - Yun Pan
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China (mainland)
| | - Di Cao
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China (mainland)
| | - Yi-Yi Liu
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China (mainland)
| | - Ai-Jun Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China (mainland)
| |
Collapse
|
36
|
The Use of Genetically Engineered Mouse Models for Studying the Function of Mutated Driver Genes in Pancreatic Cancer. J Clin Med 2019; 8:jcm8091369. [PMID: 31480737 PMCID: PMC6780401 DOI: 10.3390/jcm8091369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is often treatment-resistant, with the emerging standard of care, gemcitabine, affording only a few months of incrementally-deteriorating survival. Reflecting on the history of failed clinical trials, genetically engineered mouse models (GEMMs) in oncology research provides the inspiration to discover new treatments for pancreatic cancer that come from better knowledge of pathogenesis mechanisms, not only of the derangements in and consequently acquired capabilities of the cancer cells, but also in the aberrant microenvironment that becomes established to support, sustain, and enhance neoplastic progression. On the other hand, the existing mutational profile of pancreatic cancer guides our understanding of the disease, but leaves many important questions of pancreatic cancer biology unanswered. Over the past decade, a series of transgenic and gene knockout mouse modes have been produced that develop pancreatic cancers with features reflective of metastatic pancreatic ductal adenocarcinoma (PDAC) in humans. Animal models of PDAC are likely to be essential to understanding the genetics and biology of the disease and may provide the foundation for advances in early diagnosis and treatment.
Collapse
|
37
|
Mohammadzadeh A, Mirza-Aghazadeh-Attari M, Hallaj S, Saei AA, Alivand MR, Valizadeh A, Yousefi B, Majidinia M. Crosstalk between P53 and DNA damage response in ageing. DNA Repair (Amst) 2019; 80:8-15. [DOI: 10.1016/j.dnarep.2019.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/19/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023]
|
38
|
Lee W, Cho JH, Lee Y, Lee S, Kim DH, Ha S, Kondo Y, Ishigami A, Chung HY, Lee J. Dibutyl phthalate impairs neural progenitor cell proliferation and hippocampal neurogenesis. Food Chem Toxicol 2019; 129:239-248. [DOI: 10.1016/j.fct.2019.04.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/03/2019] [Accepted: 04/22/2019] [Indexed: 01/18/2023]
|
39
|
Bartold K, Pietrzyk-Le A, Lisowski W, Golebiewska K, Siklitskaya A, Borowicz P, Shao S, D'Souza F, Kutner W. Promoting bioanalytical concepts in genetics: A TATA box molecularly imprinted polymer as a small isolated fragment of the DNA damage repairing system. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:1-10. [PMID: 30948043 DOI: 10.1016/j.msec.2019.02.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 11/25/2022]
Abstract
We demonstrate that a new, stable, artificial TATA (T - thymine, A - adenine) box is recognized by amino acids recognizing the natural TATA box. Here, the former mimicked, as a minimal motif, oligodeoxyribonucleotide interactions with amino acids of proteins involved in repairing of damaged dsDNA. By electropolymerization, we molecularly imprinted non-labeled 5'-TATAAA-3' via Watson-Crick nucleobase pairing, thus synthesizing, in a one-step procedure, the hexakis[bis(2,2'-bithien-5-yl)] TTTATA and simultaneously hybridizing it with the 5'-TATAAA-3' template. That is, a stable dsDNA analog having a controlled sequence of nucleobases was formed in the molecularly imprinted polymer (MIP). The 5'-TATAAA-3' was by the X-ray photoelectron spectroscopy (XPS) depth profiling found to be homogeneously distributed both in the bulk of the MIP film and on its surface. The 5'-TATAAA-3' concentration in the 2.8(±0.2)-nm relative surface area, ~140-nm thick MIP film was 2.1 mM. The MIP served as a matrix of an artificial TATA box with the TATAAA-promoter sequence. We comprehensively characterized this artificial DNA hybrid by the polarization-modulation infrared reflection-absorption spectroscopy (PM-IRRAS) and X-ray photoelectron spectroscopy (XPS). Further, we examined interactions of DNA repairing TATA binding protein (TBP) amino acids with the artificial TATA box prepared. That is, molecules of l-phenylalanine aromatic amino acid were presumably engaged in stacking interactions with nucleobase steps of this artificial TATA box. The nitrogen-to‑phosphorus atomic % ratio on the surface of the MIP-(5'-TATAAA-3') film increased by ~1.6 times after film immersing in the l-glutamic acid solution, as determined using the XPS depth profiling. Furthermore, l-lysine and l-serine preferentially interacted with the phosphate moiety of 5'-TATAAA-3'. We monitored amino acids interactions with the artificial TATA box using real-time piezoelectric microgravimetry at a quartz crystal microbalance (QCM) and surface plasmon resonance (SPR) spectroscopy under flow injection analysis (FIA) conditions.
Collapse
Affiliation(s)
- Katarzyna Bartold
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | | | - Wojciech Lisowski
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | | | | | - Pawel Borowicz
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Shuai Shao
- Department of Chemistry, University of North Texas, Denton TX, USA
| | - Francis D'Souza
- Department of Chemistry, University of North Texas, Denton TX, USA
| | - Wlodzimierz Kutner
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland; Faculty of Mathematics and Natural Sciences, School of Sciences, Cardinal Stefan Wyszynski University in Warsaw, Poland
| |
Collapse
|
40
|
Nag S, Manna K, Saha KD. Tannic acid-stabilized gold nano-particles are superior to native tannic acid in inducing ROS-dependent mitochondrial apoptosis in colorectal carcinoma cells via the p53/AKT axis. RSC Adv 2019; 9:8025-8038. [PMID: 35547831 PMCID: PMC9087445 DOI: 10.1039/c9ra00808j] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/22/2019] [Indexed: 12/19/2022] Open
Abstract
Tannic acid and AuNP-TA lead to death of colon cancer cells via the ROS/p53/Akt pathway, and AuNP-TA is more potent.
Collapse
Affiliation(s)
- Sayoni Nag
- Cancer Biology and Inflammatory Disorder Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
| | - Krishnendu Manna
- Cancer Biology and Inflammatory Disorder Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
| | - Krishna Das Saha
- Cancer Biology and Inflammatory Disorder Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
| |
Collapse
|
41
|
Eslamparast A, Abbasgholizadeh R, Ostad SN, Gharghabi M, Ghahremani MH. N-Terminal Domain of Fragile Histidine Triad Exerts Potent Cytotoxic Effect in HT1080 Cells and Increases Doxorubicin Cytotoxicity. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2019; 18:254-262. [PMID: 31089360 PMCID: PMC6487436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Fragile histidine triad (FHIT) serves a critical function as a tumor suppressor that inhibits p53 degradation by mouse double minute 2 (MDM2). The functional domains of FHIT involved in tumor inhibition was interpreted. In-silico screening data were employed to construct truncated forms of FHIT to assess their cytotoxic effects on the HT1080 cell line. Full FHIT expression was confirmed by western blotting and expression of two FHIT truncates were confirmed by RT-PCR. Transfection of these truncated forms into HT1080 cells showed that the N-terminal truncated form (amino acids 17-102) better inhibited proliferation than the full-length FHIT. The combined effects of these truncated forms augmented doxorubicin-induced cytotoxicity. Functional analysis demonstrated that these fragments and their combination with doxorubicin can arrest cells in the G2 phase of the cell cycle as specified by flow cytometry. The FHIT functional domains can be used as lead compounds for development of drug designs and gene transfer for cancer therapy.
Collapse
Affiliation(s)
- Ameneh Eslamparast
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran. ,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Reza Abbasgholizadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Nasser Ostad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Gharghabi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Corresponding author: E-mail:
| |
Collapse
|
42
|
Wang XJ, Chen W, Fu XT, Ma JK, Wang MH, Hou YJ, Tian DC, Fu XY, Fan CD. Reversal of homocysteine-induced neurotoxicity in rat hippocampal neurons by astaxanthin: evidences for mitochondrial dysfunction and signaling crosstalk. Cell Death Discov 2018; 4:50. [PMID: 30374413 PMCID: PMC6197197 DOI: 10.1038/s41420-018-0114-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/29/2018] [Accepted: 10/01/2018] [Indexed: 11/09/2022] Open
Abstract
Elevated plasma level of homocysteine (Hcy) represents an independent risk for neurological diseases, and induction of oxidative damage is considered as one of the most important pathomechanisms. Astaxanthin (ATX) exhibits strong antioxidant activity in kinds of experimental models. However, the potential of ATX against Hcy-induced neurotoxicity has not been well explored yet. Herein, the neuroprotective effect of ATX against Hcy-induced neurotoxicity in rat hippocampal neurons was examined, and the underlying mechanism was evaluated. The results showed that ATX pre-treatment completely reversed Hcy-induced neurotoxicity through inhibiting cell apoptosis in rat primary hippocampal neurons. The mechanical investigation revealed that ATX effectively blocked Hcy-induced mitochondrial dysfunction by regulating Bcl-2 family and opening of mitochondrial permeability transition pore (MPTP). ATX pre-treatment also attenuated Hcy-induced oxidative damage via inhibiting the release of intracellular reactive oxide species (ROS) and superoxide anion through regulating MPTP opening. Moreover, normalization of MAPKs and PI3K/AKT pathways also contributed to ATX-mediated protective effects. Taken together, these results above suggested that ATX has the potential to reverse Hcy-induced neurotoxicity and apoptosis by inhibiting mitochondrial dysfunction, ROS-mediated oxidative damage and regulation of MAKPs and AKT pathways, which validated the strategy of using ATX could be a highly effective way in combating Hcy-mediated neurological disorders.
Collapse
Affiliation(s)
- Xian-Jun Wang
- 1Department of Neurology, People's Hospital of Linyi Affiliated to Qingdao University, Linyi, 276000 Shandong China
| | - Wang Chen
- 1Department of Neurology, People's Hospital of Linyi Affiliated to Qingdao University, Linyi, 276000 Shandong China
| | - Xiao-Ting Fu
- 2School of Basic Medicine, Taishan Medical University, Taian, Shandong 271000 China
| | - Jin-Kui Ma
- 3Faculty of Bioresource Sciences, Akita Prefectural University, 241-438 Kaidobata-Nishi, Shimoshinjo-Nakano, Akita-shi, Akita 010-0195 Japan
| | - Mei-Hong Wang
- Department of Neurology, People's Hospital of Yishui, Linyi, 276400 Shandong China
| | - Ya-Jun Hou
- 2School of Basic Medicine, Taishan Medical University, Taian, Shandong 271000 China
| | - Da-Chen Tian
- 1Department of Neurology, People's Hospital of Linyi Affiliated to Qingdao University, Linyi, 276000 Shandong China
| | - Xiao-Yan Fu
- 2School of Basic Medicine, Taishan Medical University, Taian, Shandong 271000 China
| | - Cun-Dong Fan
- 2School of Basic Medicine, Taishan Medical University, Taian, Shandong 271000 China
| |
Collapse
|
43
|
Niklaus NJ, Humbert M, Tschan MP. Cisplatin sensitivity in breast cancer cells is associated with particular DMTF1 splice variant expression. Biochem Biophys Res Commun 2018; 503:2800-2806. [PMID: 30100063 DOI: 10.1016/j.bbrc.2018.08.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/05/2018] [Indexed: 01/08/2023]
Abstract
The cyclin D binding myb-like transcription factor 1 (DMTF1) is a tumor suppressor gene that activates p14ARF transcription and thereby stabilizing the p53 tumor suppressor. The DMTF1 gene locus encodes for three different alternatively spliced isoforms, namely DMTF1α, β and γ. The oncogenic DMTF1β isoform negatively interferes with the transcriptional activity of DMTF1α. Increased DMTF1β is associated with increased cell proliferation in a variety of cancer cell types. In this study, we aimed at identifying the role of DMTF1 isoforms in response to cisplatin treatment in breast cancer cells. First, we used SKBR3 (cisplatin sensitive) and MCF7 (cisplatin resistant) breast cancer cell lines to quantify DMTF1 expression in response to cisplatin treatment. Total DMTF1 mRNA levels increased in a dose dependent manner in both cell lines upon cisplatin treatment. However, the mRNA levels of the isoforms revealed that the sensitive cell line, SKBR3, showed increased levels of both isoforms, whereas the resistant cell, MCF7, only showed increased levels of the oncogenic DMTF1β isoform. Silencing all DMTF1 isoforms led to increased cell survival upon cisplatin treatment. Furthermore, we found a significant increase in the percentage of quiescent cells in SKBR3 shDMTF1. Together, our data suggest that DMTF1 expression levels are associated with increased cisplatin resistance.
Collapse
Affiliation(s)
- Nicolas J Niklaus
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Magali Humbert
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland
| | - Mario P Tschan
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland.
| |
Collapse
|
44
|
Nattress CB, Halldén G. Advances in oncolytic adenovirus therapy for pancreatic cancer. Cancer Lett 2018; 434:56-69. [PMID: 29981812 DOI: 10.1016/j.canlet.2018.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023]
Abstract
Survival rates for pancreatic cancer patients have remained unchanged for the last four decades. The most aggressive, and most common, type of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC), which has the lowest 5-year survival rate of all cancers globally. The poor prognosis is typically due to late presentation of often non-specific symptoms and rapid development of resistance to all current therapeutics, including the standard-of-care cytotoxic drug gemcitabine. While early surgical intervention can significantly prolong patient survival, there are few treatment options for late-stage non-resectable metastatic disease, resulting in mostly palliative care. In addition, a defining feature of pancreatic cancer is the immunosuppressive and impenetrable desmoplastic stroma that blocks access to tumour cells by therapeutic drugs. The limited effectiveness of conventional chemotherapeutics reveals an urgent need to develop novel therapies with different mechanisms of action for this malignancy. An emerging alternative to current therapeutics is oncolytic adenoviruses; these engineered biological agents have proven efficacy and tumour-selectivity in preclinical pancreatic cancer models, including models of drug-resistant cancer. Safety of oncolytic adenoviral mutants has been extensively assessed in clinical trials with only limited toxicity to normal healthy tissue being reported. Promising efficacy in combination with gemcitabine was demonstrated in preclinical and clinical studies. A recent surge in novel adenoviral mutants entering clinical trials for pancreatic cancer indicates improved efficacy through activation of the host anti-tumour responses. The potential for adenoviruses to synergise with chemotherapeutics, activate anti-tumour immune responses, and contribute to stromal dissemination render these mutants highly attractive candidates for improved patient outcomes. Currently, momentum is gathering towards the development of systemically-deliverable mutants that are able to overcome anti-viral host immune responses, erythrocyte binding and hepatic uptake, to promote elimination of primary and metastatic lesions. This review will cover the key components of pancreatic cancer oncogenesis; novel oncolytic adenoviruses; clinical trials; and the current progress in overcoming the challenges of systemic delivery.
Collapse
Affiliation(s)
- Callum Baird Nattress
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, United Kingdom
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, United Kingdom.
| |
Collapse
|
45
|
Kalathil D, Prasad M, Chelladurai M, John S, Nair AS. Thiostrepton degrades mutant p53 by eliciting an autophagic response in SW480 cells. J Cell Physiol 2018; 233:6938-6950. [PMID: 29665004 DOI: 10.1002/jcp.26601] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 03/14/2018] [Indexed: 12/18/2022]
Abstract
Mutations in p53 gene are one of the hallmarks of tumor development. Specific targeting of mutant p53 protein has a promising role in cancer therapeutics. Our preliminary observation showed destabilization of mutant p53 protein in SW480, MiaPaCa and MDAMB231 cell lines upon thiostrepton treatment. In order to elucidate the mechanism of thiostrepton triggered mutant p53 degradation, we explored the impact of proteasome inhibition on activation of autophagy. Combined treatment of thiostrepton and cycloheximide/chloroquine prevented the degradation of mutant p53 protein, reinforcing autophagy as the means of mutant p53 destabilization. Our initial studies suggested that mutant p53 degradation post THSP treatment was carried out by BAG3 mediated autophagy, based on the evidence of BAG1 to BAG3 switching. Subsequent interactome analysis performed post thiostrepton treatment revealed an association of p53 with autophagosome complex associated proteins such as BAG3, p62 and HSC70. Reaccumulation of p53 was seen in BAG3 silenced cells treated with thiostrepton, thereby confirming the role of BAG3 in destabilization of this molecule. Further, localization of p53 into the lysosome upon THSP treatment substantiated our findings that mutant p53 was degraded by an autopahgic process.
Collapse
Affiliation(s)
- Dhanya Kalathil
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Manu Prasad
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Maharrish Chelladurai
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Samu John
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Asha S Nair
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
46
|
Li JY, Liu CP, Shiao WC, Jayakumar T, Li YS, Chang NC, Huang SY, Hsieh CY. Inhibitory effect of PDGF-BB and serum-stimulated responses in vascular smooth muscle cell proliferation by hinokitiol via up-regulation of p21 and p53. Arch Med Sci 2018; 14:579-587. [PMID: 29765446 PMCID: PMC5949921 DOI: 10.5114/aoms.2018.75085] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/30/2015] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Vascular smooth muscle cell (VSMC) proliferation plays a major role in the progression of vascular diseases. In the present study, we established the efficacy and the mechanisms of action of hinokitiol, a tropolone derivative found in Chamaecyparis taiwanensis, Cupressaceae, in relation to platelet-derived growth factor-BB (PDGF-BB) and serum-dependent VSMC proliferation. MATERIAL AND METHODS Primary cultured rat VSMCs were pre-treated with hinokitiol and then stimulated by PDGF-BB (10 ng/ml) or serum (10% fetal bovine serum). Cell proliferation and cytotoxicity were determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and lactose dehydrogenase assay, respectively. The degree of DNA synthesis was evaluated by BrdU-incorporation measurements and observed using confocal microscopy. Immunoblotting was utilized to determine the protein level of p-extracellular signal-regulated kinase (ERK) 1/2, p-Akt, p-phosphoinositide 3-kinase (PI3K), p-Janus kinase 2 (JAK2), p-p53, and p21Cip1. The promoter activity of p21 and p53 activity were measured by dual luciferase reporter assay. RESULTS Treatment with hinokitiol (1-10 μM) inhibited PDGF-BB and serum-induced VSMC proliferation and DNA synthesis in a concentration-dependent manner. Cytotoxicity was not observed in hinokitiol-treated VSMCs at the studied concentrations. Pre-incubation of VSMCs with hinokitiol did not alter PDGF-BB-induced phosphorylation of ERK1/2, Akt, PI3K or JAK2. Interestingly, hinokitiol induced promoter activity of p21 and p21 protein expression in VSMCs. Furthermore, hinokitiol augmented p53 protein phosphorylation and subsequently led to enhanced p53 activity. CONCLUSIONS These data suggest that the anti-proliferative effects of hinokitiol in VSMCs may be mediated by activation of p21 and p53 signaling pathways, and it may contribute to the prevention of vascular diseases associated with VSMC proliferation.
Collapse
Affiliation(s)
- Jiun-Yi Li
- Department of Cardiovascular Surgery, Mackay Memorial Hospital, and Mackay Medical College, Taipei, Taiwan
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Ping Liu
- Department of Cardiology, Yuan’s General Hospital, Kaohsiung, Taiwan
| | - Wei-Cheng Shiao
- Department of Internal Medicine, Yuan’s General Hospital, Kaohsiung, Taiwan
| | - Thanasekaran Jayakumar
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Shin Li
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nen-Chung Chang
- Department of Cardiology, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Ying Hsieh
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
47
|
Zhang JJ, Muenzner JK, Abu El Maaty MA, Karge B, Schobert R, Wölfl S, Ott I. A multi-target caffeine derived rhodium(i) N-heterocyclic carbene complex: evaluation of the mechanism of action. Dalton Trans 2018; 45:13161-8. [PMID: 27334935 DOI: 10.1039/c6dt02025a] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A rhodium(i) and a ruthenium(ii) complex with a caffeine derived N-heterocyclic carbene (NHC) ligand were biologically investigated as organometallic conjugates consisting of a metal center and a naturally occurring moiety. While the ruthenium(ii) complex was largely inactive, the rhodium(i) NHC complex displayed selective cytotoxicity and significant anti-metastatic and in vivo anti-vascular activities and acted as both a mammalian and an E. coli thioredoxin reductase inhibitor. In HCT-116 cells it increased the reactive oxygen species level, leading to DNA damage, and it induced cell cycle arrest, decreased the mitochondrial membrane potential, and triggered apoptosis. This rhodium(i) NHC derivative thus represents a multi-target compound with promising anti-cancer potential.
Collapse
Affiliation(s)
- Jing-Jing Zhang
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstr. 55, D-38106 Braunschweig, Germany. and Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Julienne K Muenzner
- Department of Organic Chemistry, University Bayreuth, Universitätsstr. 30, D-95440 Bayreuth, Germany
| | - Mohamed A Abu El Maaty
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Bianka Karge
- Department of Chemical Biology, Helmholtz Centre for Infection Research GmbH, Inhoffenstr. 7, D-38124 Braunschweig, Germany
| | - Rainer Schobert
- Department of Organic Chemistry, University Bayreuth, Universitätsstr. 30, D-95440 Bayreuth, Germany
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstr. 55, D-38106 Braunschweig, Germany.
| |
Collapse
|
48
|
Yan R, Zhu K, Dang C, Lan K, Wang H, Yuan D, Chen W, Meltzer SJ, Li K. Paf15 expression correlates with rectal cancer prognosis, cell proliferation and radiation response. Oncotarget 2018; 7:38750-38761. [PMID: 27246972 PMCID: PMC5122426 DOI: 10.18632/oncotarget.9606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 04/26/2016] [Indexed: 01/12/2023] Open
Abstract
Paf15, which participates in DNA repair, is overexpressed in numerous solid tumors. Blocking of Paf15 inhibits the growth of many types of cancer cells; while simultaneously enhancing cellular sensitivity to UV radiation. However, its expression and function in rectal cancer (RC) remain unknown. The current study was undertaken to assess the association of Paf15 expression with RC prognosis, as well as to explore the participation of Paf15 in the response of RC cells to irradiation. Increased Paf15 expression was observed in RC tissues and associated with pTNM stage and poor survival. In vitro, Paf15 induced increased RC cell proliferation while accelerating cell cycle progression, inhibiting cell death, and protecting against gamma radiation-induced DNA damage in RC cells. In conclusion, increased Paf15 expression is associated with increased RC proliferation, decreased patient survival, and a worse radiotherapeutic response.
Collapse
Affiliation(s)
- Rong Yan
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China.,Department of Medicine (GI Division) and Oncology, Johns Hopkins School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Kun Zhu
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Ke Lan
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Haonan Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Dawei Yuan
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Wei Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Stephen J Meltzer
- Department of Medicine (GI Division) and Oncology, Johns Hopkins School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Kang Li
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
49
|
Hiramoto K, Yamate Y, Yokoyama S. Ultraviolet A Eye Irradiation Ameliorates Atopic Dermatitis via p53 and Clock Gene Proteins in NC/Nga Mice. Photochem Photobiol 2018; 94:378-383. [PMID: 29105092 DOI: 10.1111/php.12853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/01/2017] [Indexed: 11/29/2022]
Abstract
Atopic dermatitis (AD) is a widespread chronic skin condition that severely affects quality of life and can lead to more serious complications. Although ultraviolet (UV)A eye irradiation can exert various effects on the skin, it is unknown whether UVA can affect AD. To investigate potential associations, we used an NC/Nga mouse model of AD to study the effects of UVA eye irradiation. The eyes of mice were irradiated with a UVA dose of 100 kJ m-2 using a FL20SBLB-A lamp. Our histological data demonstrated that AD symptoms could be ameliorated by UVA eye irradiation. We also observed an increase in the levels of adrenocorticotropic hormone (ACTH), p53 and retinoid X receptor α (RXRα) in mice with UVA-irradiated eyes. In contrast, the levels of thymic stromal lymphopoietin (TSLP), period 2 (PER2) and differentiated embryo chondrocytes 1 (DEC1) protein were decreased in mice treated with UVA irradiation. Furthermore, UVA eye-irradiated mice exhibited reduced DEC1 and RXRα colocalization compared with nonirradiated mice. These results suggested that p53 and various clock gene proteins played important roles in the amelioration of AD symptoms observed after UVA eye irradiation; this technique may have therapeutic applications in AD.
Collapse
Affiliation(s)
- Keiichi Hiramoto
- Department of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Yurika Yamate
- Department of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Satoshi Yokoyama
- Department of Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
50
|
Rehm T, Rothemund M, Bär A, Dietel T, Kempe R, Kostrhunova H, Brabec V, Kasparkova J, Schobert R. N,N-Dialkylbenzimidazol-2-ylidene platinum complexes – effects of alkyl residues and ancillary cis-ligands on anticancer activity. Dalton Trans 2018; 47:17367-17381. [DOI: 10.1039/c8dt03360a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Benzimidazol-2-ylidene platinum complexes exhibit anticancer activity, which is tuneable via N-alkyl residues and ancillary ligands and is different from that of cisplatin.
Collapse
Affiliation(s)
- Tobias Rehm
- Department of Chemistry
- University Bayreuth
- 95440 Bayreuth
- Germany
| | | | - Alexander Bär
- Department of Chemistry
- University Bayreuth
- 95440 Bayreuth
- Germany
| | - Thomas Dietel
- Lehrstuhl fuer Anorganische Chemie II
- University Bayreuth
- 95440 Bayreuth
- Germany
| | - Rhett Kempe
- Lehrstuhl fuer Anorganische Chemie II
- University Bayreuth
- 95440 Bayreuth
- Germany
| | - Hana Kostrhunova
- Institute of Biophysics
- Academy of Sciences of the Czech Republic
- CZ-61265 Brno
- Czech Republic
| | - Viktor Brabec
- Institute of Biophysics
- Academy of Sciences of the Czech Republic
- CZ-61265 Brno
- Czech Republic
| | - Jana Kasparkova
- Institute of Biophysics
- Academy of Sciences of the Czech Republic
- CZ-61265 Brno
- Czech Republic
| | - Rainer Schobert
- Department of Chemistry
- University Bayreuth
- 95440 Bayreuth
- Germany
| |
Collapse
|