1
|
Chan HW, Lin WC, Kuo DY, Chuang HY. Beta-Caryophyllene Augments Radiotherapy Efficacy in GBM by Modulating Cell Apoptosis and DNA Damage Repair via PPARγ and NF-κB Pathways. Phytother Res 2024. [PMID: 39668701 DOI: 10.1002/ptr.8413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/30/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain malignancy with limited treatment options. Radiotherapy (RT) is often used for treating unresectable GBM; however, the outcomes are often limited due to the radioresistance of GBM. Therefore, the discovery of potential radiosensitizers to enhance GBM responses to RT is crucial. Beta-caryophyllene (BCP), a natural cannabinoid, promotes cancer apoptosis by upregulating the PPARγ signaling pathway and can cross the blood-brain barrier due to its lipophilic nature. This study aimed to evaluate the radiosensitizing potential of BCP in GBM cells. U87MG and GL261 cells and a GL261 tumor-bearing model were treated with RT, BCP, or both. Treatment efficacy was assessed using the MTT assay and tumor growth tracking, and the underlying mechanisms were investigated using western blotting, immunofluorescence staining, and other analyses. BCP synergistically enhanced the efficacy of RT in cell culture, as evidenced by the combination index determined through the MTT assay. This enhancement was mediated by the BCP-induced deceleration of DNA damage repair, as demonstrated by sustained γH2AX signal, upregulated PPARγ levels, and reduced expression of pAKT, pERK, and NF-κB, indicating apoptosis induction and inhibition of survival pathways. BCP significantly inhibited tumor growth in GL261 tumor-bearing mice with no discernible side effects. These findings indicate that BCP may serve as a potential radiosensitizer for improving RT outcomes in GBM by inhibiting DNA repair, inducing apoptosis, and suppressing anti-apoptotic and survival pathways.
Collapse
Affiliation(s)
- Hui-Wen Chan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chan Lin
- Department of Radiology, Cathay General Hospital, Taipei City, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Deng-Yu Kuo
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
2
|
Shaharudin NS, Surindar Singh GK, Kek TL, Sultan S. Targeting signaling pathways with andrographolide in cancer therapy (Review). Mol Clin Oncol 2024; 21:81. [PMID: 39301125 PMCID: PMC11411607 DOI: 10.3892/mco.2024.2779] [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: 03/19/2024] [Accepted: 07/10/2024] [Indexed: 09/22/2024] Open
Abstract
Terpenoids are a large group of naturally occurring organic compounds with a wide range of components. A phytoconstituent in this group, andrographolide, which is derived from a plant called Andrographis paniculate, offers a number of advantages, including anti-inflammatory, anticancer, anti-angiogenesis and antioxidant effects. The present review elucidates the capacity of andrographolide to inhibit signaling pathways, namely the nuclear factor-κB (NF-κB), hypoxia-inducible factor 1 (HIF-1), the Janus kinase (JAK)/signal transducer and activator of transcription (STAT), phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), Wnt/β-catenin and mitogen-activated protein kinase (MAPK) pathways, which are involved in cellular processes and responses such as the inflammatory response, apoptosis and angiogenesis. Inhibiting pathways enables andrographolide to exhibit its anticancer effects against breast, colorectal and lung cancer. The present review focuses on the anticancer effects of andrographolide, specifically in breast, colorectal and lung cancer through the NF-κB, HIF-1 and JAK/STAT signaling pathways. Therefore, the Google Scholar, PubMed and ScienceDirect databases were used to search for references to these prevalent types of cancer and the anticancer mechanisms of andrographolide associated with them. The following key words were used: Andrographolide, anticancer, JAK/STAT, HIF-1, NF-κB, PI3K/AKT/mTOR, Wnt/β-catenin and MAPK pathways, and the literature was limited to studies published between 2010 to 2023. The present review article provides details about the different involvements of signaling pathways in the anticancer mechanisms of andrographolide.
Collapse
Affiliation(s)
- Nur Shahirah Shaharudin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300, Malaysia
| | - Gurmeet Kaur Surindar Singh
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300, Malaysia
- Faculty of Pharmacy, Brain Degeneration and Therapeutics Research Center, Universiti Teknologi MARA, Shah Alam, Selangor 40450, Malaysia
| | - Teh Lay Kek
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300, Malaysia
| | - Sadia Sultan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300, Malaysia
- Faculty of Pharmacy, Biotransformation Research Center, Universiti Teknologi MARA, Shah Alam, Selangor 40450, Malaysia
| |
Collapse
|
3
|
Wu YQ, Ding KX, Lv ZC, Cao ZY, Zhao K, Gao HY, Sun HY, Li JJ, Li SY, Zhao XW, Xue Y, Xiang SS, Zheng XF, Yang XM, Li CY. Single-Cell Transcriptomics Reveals Early Effects of Ionizing Radiation on Bone Marrow Mononuclear Cells in Mice. Int J Mol Sci 2024; 25:9287. [PMID: 39273235 PMCID: PMC11395520 DOI: 10.3390/ijms25179287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Ionizing radiation exposure can cause damage to diverse tissues and organs, with the hematopoietic system being the most sensitive. However, limited information is available regarding the radiosensitivity of various hematopoietic cell populations in the bone marrow due to the high heterogeneity of the hematopoietic system. In this study, we observed that granulocyte-macrophage progenitors, hematopoietic stem/progenitor cells, and B cells within the bone marrow showed the highest sensitivity, exhibiting a rapid decrease in cell numbers following irradiation. Nonetheless, neutrophils, natural killer (NK) cells, T cells, and dendritic cells demonstrated a certain degree of radioresistance, with neutrophils exhibiting the most pronounced resistance. By employing single-cell transcriptome sequencing, we investigated the early responsive genes in various cell types following irradiation, revealing that distinct gene expression profiles emerged between radiosensitive and radioresistant cells. In B cells, radiation exposure led to a specific upregulation of genes associated with mitochondrial respiratory chain complexes, suggesting a connection between these complexes and cell radiosensitivity. In neutrophils, radiation exposure resulted in fewer gene alterations, indicating their potential for distinct mechanisms in radiation resistance. Collectively, this study provides insights into the molecular mechanism for the heterogeneity of radiosensitivity among the various bone marrow hematopoietic cell populations.
Collapse
Affiliation(s)
- Yun-Qiang Wu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ke-Xin Ding
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhi-Chun Lv
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zheng-Yue Cao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ke Zhao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hui-Ying Gao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hui-Ying Sun
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jing-Jing Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Si-Yu Li
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Xiong-Wei Zhao
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Yang Xue
- School of Life Sciences, Hebei University, No. 180 Wusi Dong Road, Lian Chi District, Baoding 071000, China
| | - Shen-Si Xiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiao-Fei Zheng
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiao-Ming Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China
- School of Life Sciences, Hebei University, No. 180 Wusi Dong Road, Lian Chi District, Baoding 071000, China
| |
Collapse
|
4
|
Wang Y, Cheng Y, Zhang P, Huang D, Zhai X, Feng Z, Fang D, Liu C, Du J, Cai J. FG-4592 protected haematopoietic system from ionising radiation in mice. Immunology 2024; 172:614-626. [PMID: 38685744 DOI: 10.1111/imm.13797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
Ionising radiation exposure can lead to acute haematopoietic radiation syndrome. Despite significant advancements in the field of radioprotection, no drugs with high efficacy and low toxicity have yet been approved by the Food and Drug Administration. FG-4592, as a proline hydroxylase inhibitor, may play an important role in radioprotection of the haematopoietic system. Mice were peritoneal injected with FG-4592 or normal saline. After irradiation, the survival time, body weight, peripheral blood cell and bone marrow cell (BMC) count, cell apoptosis, pathology were analysed and RNA-sequence technique (RNA-Seq) was conducted to explore the mechanism of FG-4592 in the haematopoietic system. Our results indicated that FG-4592 improved the survival rate and weight of irradiated mice and protected the spleen, thymus and bone marrow from IR-induced injury. The number of BMCs was increased and protected against IR-induced apoptosis. FG-4592 also promoted the recovery of the blood system and erythroid differentiation. The results of RNA-Seq and Western blot showed that the NF-κB signalling pathway and hypoxia-inducible factor-1 (HIF-1) signalling pathway were upregulated by FG-4592. Meanwhile, RT-PCR results showed that FG-4592 could promote inflammatory response significantly. FG-4592 exhibited radioprotective effects in the haematopoietic system by promoting inflammatory response and targeting the NF-κB, HIF signalling pathway.
Collapse
Affiliation(s)
- Yuedong Wang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Ying Cheng
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Pei Zhang
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, China
| | - Daqian Huang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Xuanlu Zhai
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Zhenlan Feng
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Duo Fang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Cong Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Jicong Du
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Jianming Cai
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
5
|
Gupta K, Perkerson RB, Parsons TM, Angom R, Amerna D, Burgess JD, Ren Y, McLean PJ, Mukhopadhyay D, Vibhute P, Wszolek ZK, Zubair AC, Quiñones-Hinojosa A, Kanekiyo T. Secretome from iPSC-derived MSCs exerts proangiogenic and immunosuppressive effects to alleviate radiation-induced vascular endothelial cell damage. Stem Cell Res Ther 2024; 15:230. [PMID: 39075600 PMCID: PMC11287895 DOI: 10.1186/s13287-024-03847-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Radiation therapy is the standard of care for central nervous system tumours. Despite the success of radiation therapy in reducing tumour mass, irradiation (IR)-induced vasculopathies and neuroinflammation contribute to late-delayed complications, neurodegeneration, and premature ageing in long-term cancer survivors. Mesenchymal stromal cells (MSCs) are adult stem cells that facilitate tissue integrity, homeostasis, and repair. Here, we investigated the potential of the iPSC-derived MSC (iMSC) secretome in immunomodulation and vasculature repair in response to radiation injury utilizing human cell lines. METHODS We generated iPSC-derived iMSC lines and evaluated the potential of their conditioned media (iMSC CM) to treat IR-induced injuries in human monocytes (THP1) and brain vascular endothelial cells (hCMEC/D3). We further assessed factors in the iMSC secretome, their modulation, and the molecular pathways they elicit. RESULTS Increasing doses of IR disturbed endothelial tube and spheroid formation in hCMEC/D3. When IR-injured hCMEC/D3 (IR ≤ 5 Gy) were treated with iMSC CM, endothelial cell viability, adherence, spheroid compactness, and proangiogenic sprout formation were significantly ameliorated, and IR-induced ROS levels were reduced. iMSC CM augmented tube formation in cocultures of hCMEC/D3 and iMSCs. Consistently, iMSC CM facilitated angiogenesis in a zebrafish model in vivo. Furthermore, iMSC CM suppressed IR-induced NFκB activation, TNF-α release, and ROS production in THP1 cells. Additionally, iMSC CM diminished NF-kB activation in THP1 cells cocultured with irradiated hCMEC/D3, iMSCs, or HMC3 microglial lines. The cytokine array revealed that iMSC CM contains the proangiogenic and immunosuppressive factors MCP1/CCL2, IL6, IL8/CXCL8, ANG (Angiogenin), GROα/CXCL1, and RANTES/CCL5. Common promoter regulatory elements were enriched in TF-binding motifs such as androgen receptor (ANDR) and GATA2. hCMEC/D3 phosphokinome profiling revealed increased expression of pro-survival factors, the PI3K/AKT/mTOR modulator PRAS40 and β-catenin in response to CM. The transcriptome analysis revealed increased expression of GATA2 in iMSCs and the enrichment of pathways involved in RNA metabolism, translation, mitochondrial respiration, DNA damage repair, and neurodevelopment. CONCLUSIONS The iMSC secretome is a comodulated composite of proangiogenic and immunosuppressive factors that has the potential to alleviate radiation-induced vascular endothelial cell damage and immune activation.
Collapse
Affiliation(s)
- Kshama Gupta
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
| | - Ralph B Perkerson
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Tammee M Parsons
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Ramacharan Angom
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Danilyn Amerna
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Jeremy D Burgess
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Debabrata Mukhopadhyay
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Prasanna Vibhute
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Zbigniew K Wszolek
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Abba C Zubair
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Alfredo Quiñones-Hinojosa
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
- Department of Neurosurgery, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
| |
Collapse
|
6
|
Pazdrowski J, Gornowicz-Porowska J, Kaźmierska J, Krajka-Kuźniak V, Polanska A, Masternak M, Szewczyk M, Golusiński W, Danczak-Pazdrowska A. Radiation-induced skin injury in the head and neck region: pathogenesis, clinics, prevention, treatment considerations and proposal for management algorithm. Rep Pract Oncol Radiother 2024; 29:373-390. [PMID: 39144266 PMCID: PMC11321788 DOI: 10.5603/rpor.100775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/16/2024] [Indexed: 08/16/2024] Open
Abstract
Worldwide increase of head and neck cancers ranks these malignancies among top causes of cancer in human population. Radiation induced skin injury (RISI) is one of the major side effects of radiotherapy (RT). Skin of the neck is exposed to radiation due to necessity of therapeutic or prophylactic (elective) irradiation of neck lymph nodes and target organs, including the larynx and hypopharynx. The location of the neck exposes these regions of the skin to various additional exposomes such as ultraviolet radiation (UVR), pollution and cigarette smoke. There are many controversies or inconsistencies regarding RISI, from molecular aspects and therapy to terminology. There is lack of high-quality and large-sample studies in both forms of RISI: acute (aRISI) and chronic (cRISI). Finally, no gold standards in the management of aRISI and cRISI have been established yet. In this article, the authors discuss the pathogenesis, clinical picture, prevention and clinical interventions and present a proposed treatment algorithm.
Collapse
Affiliation(s)
- Jakub Pazdrowski
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan, Poland
| | - Justyna Gornowicz-Porowska
- Department and Division of Practical Cosmetology and Skin Diseases Prophylaxis, Poznan University of Medical Sciences, Poznan, Poland
| | - Joanna Kaźmierska
- Department of Radiotherapy, Poznan University of Medical Sciences, Poznan, Poland
- Radiotherapy and Oncology, Greater Poland Cancer Centre, Poznan, Poland
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznan, Poland
| | - Adriana Polanska
- Department of Dermatology and Venereology Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States
| | - Mateusz Szewczyk
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan, Poland
| | - Wojciech Golusiński
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan, Poland
| | | |
Collapse
|
7
|
Kura B, Pavelkova P, Kalocayova B, Pobijakova M, Slezak J. MicroRNAs as Regulators of Radiation-Induced Oxidative Stress. Curr Issues Mol Biol 2024; 46:7097-7113. [PMID: 39057064 PMCID: PMC11276491 DOI: 10.3390/cimb46070423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
microRNAs (miRNAs) represent small RNA molecules involved in the regulation of gene expression. They are implicated in the regulation of diverse cellular processes ranging from cellular homeostasis to stress responses. Unintended irradiation of the cells and tissues, e.g., during medical uses, induces various pathological conditions, including oxidative stress. miRNAs may regulate the expression of transcription factors (e.g., nuclear factor erythroid 2 related factor 2 (Nrf2), nuclear factor kappa B (NF-κB), tumor suppressor protein p53) and other redox-sensitive genes (e.g., mitogen-activated protein kinase (MAPKs), sirtuins (SIRTs)), which trigger and modulate cellular redox signaling. During irradiation, miRNAs mainly act with reactive oxygen species (ROS) to regulate the cell fate. Depending on the pathway involved and the extent of oxidative stress, this may lead to cell survival or cell death. In the context of radiation-induced oxidative stress, miRNA-21 and miRNA-34a are among the best-studied miRNAs. miRNA-21 has been shown to directly target superoxide dismutase (SOD), or NF-κB, whereas miRNA-34a is a direct regulator of NADPH oxidase (NOX), SIRT1, or p53. Understanding the mechanisms underlying radiation-induced injury including the involvement of redox-responsive miRNAs may help to develop novel approaches for modulating the cellular response to radiation exposure.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| | - Patricia Pavelkova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia
| | - Margita Pobijakova
- Department of Radiation Oncology, Bory Hospital–Penta Hospitals, 841 03 Bratislava, Slovakia;
- Radiological Science, Faculty of Nursing and Medical Professional Studies, Slovak Medical University, 831 01 Bratislava, Slovakia
| | - Jan Slezak
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| |
Collapse
|
8
|
Holmes-Hampton GP, Kumar VP, Valenzia K, Ghosh SP. FSL-1: A Synthetic Peptide Increases Survival in a Murine Model of Hematopoietic Acute Radiation Syndrome. Radiat Res 2024; 201:449-459. [PMID: 38373011 DOI: 10.1667/rade-23-00142.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/05/2023] [Indexed: 02/20/2024]
Abstract
In the current geopolitical climate there is an unmet need to identify and develop prophylactic radiation countermeasures, particularly to ensure the well-being of warfighters and first responders that may be required to perform on radiation-contaminated fields for operational or rescue missions. Currently, no countermeasures have been approved by the U.S. FDA for prophylactic administration. Here we report on the efficacious nature of FSL-1 (toll-like receptor 2/6 agonist) and the protection from acute radiation syndrome (ARS) in a murine total-body irradiation (TBI) model. A single dose of FSL-1 was administered subcutaneously in mice. The safety of the compound was assessed in non-irradiated animals, the efficacy of the compound was assessed in animals exposed to TBI in the AFRRI Co-60 facility, the dose of FSL-1 was optimized, and common hematological parameters [complete blood cell (CBC), cytokines, and bone marrow progenitor cells] were assessed. Animals were monitored up to 60 days after exposure and radiation-induced damage was evaluated. FSL-1 was shown to be non-toxic when administered to non-irradiated mice at doses up to 3 mg/kg. The window of efficacy was determined to be 24 h prior to 24 h after TBI. FSL-1 administration resulted in significantly increased survival when administered either 24 h prior to or 24 h after exposure to supralethal doses of TBI. The optimal dose of FSL-1 administration was determined to be 1.5 mg/kg when administered prior to irradiation. Finally, FSL-1 protected the hematopoietic system (recovery of CBC and bone marrow CFU). Taken together, the effects of increased survival and accelerated recovery of hematological parameters suggests that FSL-1 should be developed as a novel radiation countermeasure for soldiers and civilians, which can be used either before or after irradiation in the aftermath of a radiological or nuclear event.
Collapse
Affiliation(s)
- Gregory P Holmes-Hampton
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Vidya P Kumar
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Kaylee Valenzia
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Sanchita P Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| |
Collapse
|
9
|
Kumar R, Kushawaha PK. Interferon inducible guanylate-binding protein 1 modulates the lipopolysaccharide-induced cytokines/chemokines and mitogen-activated protein kinases in macrophages. Microbiol Immunol 2024; 68:185-195. [PMID: 38462687 DOI: 10.1111/1348-0421.13123] [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: 09/11/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024]
Abstract
Guanylate-binding proteins (GBPs) are a family of interferon (IFN)-inducible GTPases and play a pivotal role in the host immune response to microbial infections. These are upregulated in immune cells after recognizing the lipopolysaccharides (LPS), the major membrane component of Gram-negative bacteria. In the present study, the expression pattern of GBP1-7 was initially mapped in phorbol 12-myristate 13-acetate-differentiated human monocytes THP-1 and mouse macrophages RAW 264.7 cell lines stimulated with LPS. A time-dependent significant expression of GBP1-7 was observed in these cells. Moreover, among the various GBPs, GBP1 has emerged as a central player in regulating innate immunity and inflammation. Therefore, to study the specific role of GBP1 in LPS-induced inflammation, knockdown of the Gbp1 gene was carried out in both cells using small interfering RNA interference. Altered levels of different cytokines (interleukin [IL]-4, IL-10, IL-12β, IFN-γ, tumor necrosis factor-α), inducible nitric oxide synthase, histocompatibility 2, class II antigen A, protein kinase R, and chemokines (chemokine (C-X-C motif) ligand 9 [CXCL9], CXCL10, and CXCL11) in GBP1 knockdown cells were reported compared to control cells. Interestingly, the extracellular-signal-regulated kinase 1/2 mitogen-activated protein (MAP) kinases and signal transducer and activator of transcription 1 (STAT1) transcription factor levels were considerably induced in knockdown cells compared to the control cells. However, no change in the level of phosphorylated nuclear factor-kB, c-Jun, and p38 transcription factors was observed in GBP1 knockdown cells compared to the control cells. This study concludes that GBP1 may alter the expression of cytokines, chemokines, and effector molecules mediated by MAP kinases and STAT1 transcription factors.
Collapse
Affiliation(s)
- Ravindra Kumar
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Pramod Kumar Kushawaha
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
10
|
Nisar H, Sanchidrián González PM, Labonté FM, Schmitz C, Roggan MD, Kronenberg J, Konda B, Chevalier F, Hellweg CE. NF-κB in the Radiation Response of A549 Non-Small Cell Lung Cancer Cells to X-rays and Carbon Ions under Hypoxia. Int J Mol Sci 2024; 25:4495. [PMID: 38674080 PMCID: PMC11050661 DOI: 10.3390/ijms25084495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Cellular hypoxia, detectable in up to 80% of non-small cell lung carcinoma (NSCLC) tumors, is a known cause of radioresistance. High linear energy transfer (LET) particle radiation might be effective in the treatment of hypoxic solid tumors, including NSCLC. Cellular hypoxia can activate nuclear factor κB (NF-κB), which can modulate radioresistance by influencing cancer cell survival. The effect of high-LET radiation on NF-κB activation in hypoxic NSCLC cells is unclear. Therefore, we compared the effect of low (X-rays)- and high (12C)-LET radiation on NF-κB responsive genes' upregulation, as well as its target cytokines' synthesis in normoxic and hypoxic A549 NSCLC cells. The cells were incubated under normoxia (20% O2) or hypoxia (1% O2) for 48 h, followed by irradiation with 8 Gy X-rays or 12C ions, maintaining the oxygen conditions until fixation or lysis. Regulation of NF-κB responsive genes was evaluated by mRNA sequencing. Secretion of NF-κB target cytokines, IL-6 and IL-8, was quantified by ELISA. A greater fold change increase in expression of NF-κB target genes in A549 cells following exposure to 12C ions compared to X-rays was observed, regardless of oxygenation status. These genes regulate cell migration, cell cycle, and cell survival. A greater number of NF-κB target genes was activated under hypoxia, regardless of irradiation status. These genes regulate cell migration, survival, proliferation, and inflammation. X-ray exposure under hypoxia additionally upregulated NF-κB target genes modulating immunosurveillance and epithelial-mesenchymal transition (EMT). Increased IL-6 and IL-8 secretion under hypoxia confirmed NF-κB-mediated expression of pro-inflammatory genes. Therefore, radiotherapy, particularly with X-rays, may increase tumor invasiveness in surviving hypoxic A549 cells.
Collapse
Affiliation(s)
- Hasan Nisar
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (P.M.S.G.); (J.K.); (B.K.)
- Department of Medical Sciences, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad 44000, Pakistan
| | - Paulina Mercedes Sanchidrián González
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (P.M.S.G.); (J.K.); (B.K.)
| | - Frederik M. Labonté
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (P.M.S.G.); (J.K.); (B.K.)
| | - Claudia Schmitz
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (P.M.S.G.); (J.K.); (B.K.)
| | - Marie Denise Roggan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (P.M.S.G.); (J.K.); (B.K.)
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Jessica Kronenberg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (P.M.S.G.); (J.K.); (B.K.)
- Microgravity User Support Center (MUSC), German Aerospace Center (DLR), 51147 Cologne, Germany
| | - Bikash Konda
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (P.M.S.G.); (J.K.); (B.K.)
| | - François Chevalier
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-University of Caen Normandy, 14000 Caen, France;
| | - Christine E. Hellweg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (P.M.S.G.); (J.K.); (B.K.)
| |
Collapse
|
11
|
Zhang B, Zhang M, Tian J, Zhang X, Zhang D, Li J, Yang L. Advances in the regulation of radiation-induced apoptosis by polysaccharides: A review. Int J Biol Macromol 2024; 263:130173. [PMID: 38360238 DOI: 10.1016/j.ijbiomac.2024.130173] [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: 12/02/2023] [Revised: 02/03/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
Polysaccharides are biomolecules composed of monosaccharides that are widely found in animals, plants and microorganisms and are of interest for their various health benefits. Cumulative studies have shown that the modulation of radiation-induced apoptosis by polysaccharides can be effective in preventing and treating a wide range of radiation injuries with safety and few side effects. Therefore, this paper summarizes the monosaccharide compositions, molecular weights, and structure-activity relationships of natural polysaccharides that regulate radiation-induced apoptosis, and also reviews the molecular mechanisms by which these polysaccharides modulate radiation-induced apoptosis, primarily focusing on promoting cancer cell apoptosis to enhance radiotherapy efficacy, reducing radiation damage to normal tissues, and inhibiting apoptosis in normal cells. Additionally, the role of gut microbiota in mediating the interaction between polysaccharides and radiation is discussed, providing innovative ideas for various radiation injuries, including hematopoiesis, immunity, and organ damage. This review will contribute to a better understanding of the value of natural polysaccharides in the field of radiation and provide guidance for the development of natural radioprotective agents and radiosensitizers.
Collapse
Affiliation(s)
- Beibei Zhang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China; Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Mingyu Zhang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China; Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Xi Zhang
- Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Dan Zhang
- Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Jiabao Li
- Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China
| | - Lei Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China; Department of Nutrition, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, China.
| |
Collapse
|
12
|
Ibragimova M, Kussainova A, Aripova A, Bersimbaev R, Bulgakova O. The Molecular Mechanisms in Senescent Cells Induced by Natural Aging and Ionizing Radiation. Cells 2024; 13:550. [PMID: 38534394 DOI: 10.3390/cells13060550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
This review discusses the relationship between cellular senescence and radiation exposure. Given the wide range of ionizing radiation sources encountered by people in professional and medical spheres, as well as the influence of natural background radiation, the question of the effect of radiation on biological processes, particularly on aging processes, remains highly relevant. The parallel relationship between natural and radiation-induced cellular senescence reveals the common aspects underlying these processes. Based on recent scientific data, the key points of the effects of ionizing radiation on cellular processes associated with aging, such as genome instability, mitochondrial dysfunction, altered expression of miRNAs, epigenetic profile, and manifestation of the senescence-associated secretory phenotype (SASP), are discussed. Unraveling the molecular mechanisms of cellular senescence can make a valuable contribution to the understanding of the molecular genetic basis of age-associated diseases in the context of environmental exposure.
Collapse
Affiliation(s)
- Milana Ibragimova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Assiya Kussainova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
- Department of Health Sciences, University of Genova, Via Pastore 1, 16132 Genoa, Italy
| | - Akmaral Aripova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Rakhmetkazhi Bersimbaev
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| | - Olga Bulgakova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan
| |
Collapse
|
13
|
Azmoonfar R, Moslehi M, Shahbazi-Gahrouei D. Radioprotective Effect of Selenium Nanoparticles: A Mini Review. IET Nanobiotechnol 2024; 2024:5538107. [PMID: 38863968 PMCID: PMC11095073 DOI: 10.1049/2024/5538107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 06/13/2024] Open
Abstract
Materials and Methods This study followed the PRISMA reporting guidelines to present the results. A comprehensive search was performed on electronic databases such as PubMed, Scopus, Web of Sciences, and Science Direct. Initially, 413 articles were retrieved. After removing duplicates and applying specific inclusion and exclusion criteria, 10 articles were finally included in this systematic review. Results The reviewed studies showed that selenium nanoparticles had anti-inflammatory and antioxidant properties. They effectively protected the kidneys, liver, and testicles from damage. Furthermore, there was evidence of efficient radioprotection for the organs examined without significant side effects. Conclusions This systematic review emphasizes the potential advantages of using selenium nanoparticles to prevent the negative effects of ionizing radiation. Importantly, these protective effects were achieved without causing noticeable side effects. These findings suggest the potential role of selenium nanoparticles as radioprotective agents, offering possible therapeutic applications to reduce the risks related to ionizing radiation exposure in medical imaging and radiotherapy procedures.
Collapse
Affiliation(s)
- Rasool Azmoonfar
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Masoud Moslehi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Daryoush Shahbazi-Gahrouei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| |
Collapse
|
14
|
Yang M, Li T, Guo S, Song K, Gong C, Huang N, Pang D, Xiao H. CVD phenotyping in oncologic disorders: cardio-miRNAs as a potential target to improve individual outcomes in revers cardio-oncology. J Transl Med 2024; 22:50. [PMID: 38216965 PMCID: PMC10787510 DOI: 10.1186/s12967-023-04680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/28/2023] [Indexed: 01/14/2024] Open
Abstract
With the increase of aging population and prevalence of obesity, the incidence of cardiovascular disease (CVD) and cancer has also presented an increasing tendency. These two different diseases, which share some common risk factors. Relevant studies in the field of reversing Cardio-Oncology have shown that the phenotype of CVD has a significant adverse effect on tumor prognosis, which is mainly manifested by a positive correlation between CVD and malignant progression of concomitant tumors. This distal crosstalk and the link between different diseases makes us aware of the importance of diagnosis, prediction, management and personalized treatment of systemic diseases. The circulatory system bridges the interaction between CVD and cancer, which suggests that we need to fully consider the systemic and holistic characteristics of these two diseases in the process of clinical treatment. The circulating exosome-miRNAs has been intrinsically associated with CVD -related regulation, which has become one of the focuses on clinical and basic research (as biomarker). The changes in the expression profiles of cardiovascular disease-associated miRNAs (Cardio-miRNAs) may adversely affect concomitant tumors. In this article, we sorted and screened CVD and tumor-related miRNA data based on literature, then summarized their commonalities and characteristics (several important pathways), and further discussed the conclusions of Cardio-Oncology related experimental studies. We take a holistic approach to considering CVD as a risk factor for tumor malignancy, which provides an in-depth analysis of the various regulatory mechanisms or pathways involved in the dual attribute miRNAs (Cardio-/Onco-miRNAs). These mechanisms will be key to revealing the systemic effects of CVD on tumors and highlight the holistic nature of different diseases. Therefore, the Cardio-miRNAs should be given great attention from researchers in the field of CVD and tumors, which might become new targets for tumor treatment. Meanwhile, based on the principles of precision medicine (such as the predictive preventive personalized medicine, 3PM) and reverse Cardio-oncology to better improve individual outcomes, we should consider developing personalized medicine and systemic therapy for cancer from the perspective of protecting cardiovascular function.
Collapse
Affiliation(s)
- Ming Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Tiepeng Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shujin Guo
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Kangping Song
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chuhui Gong
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ning Huang
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Dejiang Pang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China.
| | - Hengyi Xiao
- The Lab of Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Mohammadgholi M, Hosseinimehr SJ. Crosstalk between Oxidative Stress and Inflammation Induced by Ionizing Radiation in Healthy and Cancerous Cells. Curr Med Chem 2024; 31:2751-2769. [PMID: 37026495 DOI: 10.2174/0929867330666230407104208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/18/2023] [Accepted: 02/24/2023] [Indexed: 04/08/2023]
Abstract
Radiotherapy (RT) is a unique modality in cancer treatment with no replacement in many cases and uses a tumoricidal dose of various ionizing radiation (IR) types to kill cancer cells. It causes oxidative stress through reactive oxygen species (ROS) production or the destruction of antioxidant systems. On the other hand, RT stimulates the immune system both directly and indirectly by releasing danger signals from stress-exposed and dying cells. Oxidative stress and inflammation are two reciprocal and closely related mechanisms, one induced and involved by the other. ROS regulates the intracellular signal transduction pathways, which participate in the activation and expression of pro-inflammatory genes. Reciprocally, inflammatory cells release ROS and immune system mediators during the inflammation process, which drive the induction of oxidative stress. Oxidative stress or inflammation-induced damages can result in cell death (CD) or survival mechanisms that may be destructive for normal cells or beneficial for cancerous cells. The present study has focused on the radioprotection of those agents with binary effects of antioxidant and anti-inflammatory mechanisms IR-induced CD.
Collapse
Affiliation(s)
- Mohsen Mohammadgholi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
16
|
Chukavin NN, Filippova KO, Ermakov AM, Karmanova EE, Popova NR, Anikina VA, Ivanova OS, Ivanov VK, Popov AL. Redox-Active Cerium Fluoride Nanoparticles Selectively Modulate Cellular Response against X-ray Irradiation In Vitro. Biomedicines 2023; 12:11. [PMID: 38275372 PMCID: PMC10813610 DOI: 10.3390/biomedicines12010011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Ionizing radiation-induced damage in cancer and normal cells leads to apoptosis and cell death, through the intracellular oxidative stress, DNA damage and disorders of their metabolism. Irradiation doses that do not lead to the death of tumor cells can result in the emergence of radioresistant clones of these cells due to the rearrangement of metabolism and the emergence of new mutations, including those in the genes responsible for DNA repair. The search for the substances capable of modulating the functioning of the tumor cell repair system is an urgent task. Here we analyzed the effect of cerium(III) fluoride nanoparticles (CeF3 NPs) on normal (human mesenchymal stem cells-hMSC) and cancer (MCF-7 line) human cells after X-ray radiation. CeF3 NPs effectively prevent the formation of hydrogen peroxide and hydroxyl radicals in an irradiated aqueous solution, showing pronounced antioxidant properties. CeF3 NPs are able to protect hMSC from radiation-induced proliferation arrest, increasing their viability and mitochondrial membrane potential, and, conversely, inducing the cell death of MCF-7 cancer cells, causing radiation-induced mitochondrial hyperpolarization. CeF3 NPs provided a significant decrease in the number of double-strand breaks (DSBs) in hMSC, while in MCF-7 cells the number of γ-H2AX foci dramatically increased in the presence of CeF3 4 h after irradiation. In the presence of CeF3 NPs, there was a tendency to modulate the expression of most analyzed genes associated with the development of intracellular oxidative stress, cell redox status and the DNA-repair system after X-ray irradiation. Cerium-containing nanoparticles are capable of providing selective protection of hMSC from radiation-induced injuries and are considered as a platform for the development of promising clinical radioprotectors.
Collapse
Affiliation(s)
- Nikita N. Chukavin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia; (N.N.C.); (K.O.F.); (A.M.E.); (E.E.K.); (N.R.P.); (V.A.A.)
- Scientific and Educational Center, State University of Education, Moscow 105005, Russia
| | - Kristina O. Filippova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia; (N.N.C.); (K.O.F.); (A.M.E.); (E.E.K.); (N.R.P.); (V.A.A.)
| | - Artem M. Ermakov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia; (N.N.C.); (K.O.F.); (A.M.E.); (E.E.K.); (N.R.P.); (V.A.A.)
- Scientific and Educational Center, State University of Education, Moscow 105005, Russia
| | - Ekaterina E. Karmanova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia; (N.N.C.); (K.O.F.); (A.M.E.); (E.E.K.); (N.R.P.); (V.A.A.)
| | - Nelli R. Popova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia; (N.N.C.); (K.O.F.); (A.M.E.); (E.E.K.); (N.R.P.); (V.A.A.)
| | - Viktoriia A. Anikina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia; (N.N.C.); (K.O.F.); (A.M.E.); (E.E.K.); (N.R.P.); (V.A.A.)
| | - Olga S. Ivanova
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, Moscow 119071, Russia;
| | - Vladimir K. Ivanov
- Kurnakov Institute of General and Inorganic Chemistry, Russian Academy of Sciences, Moscow 119991, Russia;
| | - Anton L. Popov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia; (N.N.C.); (K.O.F.); (A.M.E.); (E.E.K.); (N.R.P.); (V.A.A.)
| |
Collapse
|
17
|
Shrum SA, Nukala U, Shrimali S, Pineda EN, Krager KJ, Thakkar S, Jones DE, Pathak R, Breen PJ, Aykin-Burns N, Compadre CM. Tocotrienols Provide Radioprotection to Multiple Organ Systems through Complementary Mechanisms of Antioxidant and Signaling Effects. Antioxidants (Basel) 2023; 12:1987. [PMID: 38001840 PMCID: PMC10668991 DOI: 10.3390/antiox12111987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Tocotrienols have powerful radioprotective properties in multiple organ systems and are promising candidates for development as clinically effective radiation countermeasures. To facilitate their development as clinical radiation countermeasures, it is crucial to understand the mechanisms behind their powerful multi-organ radioprotective properties. In this context, their antioxidant effects are recognized for directly preventing oxidative damage to cellular biomolecules from ionizing radiation. However, there is a growing body of evidence indicating that the radioprotective mechanism of action for tocotrienols extends beyond their antioxidant properties. This raises a new pharmacological paradigm that tocotrienols are uniquely efficacious radioprotectors due to a synergistic combination of antioxidant and other signaling effects. In this review, we have covered the wide range of multi-organ radioprotective effects observed for tocotrienols and the mechanisms underlying it. These radioprotective effects for tocotrienols can be characterized as (1) direct cytoprotective effects, characteristic of the classic antioxidant properties, and (2) other effects that modulate a wide array of critical signaling factors involved in radiation injury.
Collapse
Affiliation(s)
- Stephen A. Shrum
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Tocol Pharmaceuticals, LLC, Little Rock, AR 77205, USA
| | - Ujwani Nukala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Shivangi Shrimali
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Edith Nathalie Pineda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Kimberly J. Krager
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Shraddha Thakkar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Darin E. Jones
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Rupak Pathak
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Philip J. Breen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Nukhet Aykin-Burns
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Cesar M. Compadre
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Tocol Pharmaceuticals, LLC, Little Rock, AR 77205, USA
| |
Collapse
|
18
|
Aleman Oliva C, Aleman Espino E, Demory Beckler M, Kesselman MM. Radiation-Induced Peripheral Artery Disease in a 63-Year-Old Patient. Cureus 2023; 15:e47372. [PMID: 38022038 PMCID: PMC10657155 DOI: 10.7759/cureus.47372] [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: 09/08/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Tobacco use, hypertension, diabetes, and hypercholesterolemia are known risk factors for peripheral artery disease (PAD). However, additional causes of PAD, such as radiation therapy, should be considered for the prevention and diagnosis of this disease. The patient described in this report had 36 radiation therapies directly to the pelvis and bladder area due to bladder cancer. The presence of severe PAD on this patient's right external iliac artery, the same area where he received radiation therapy, raises the question of whether radiation therapy contributed to the development of PAD. In addition, his history of anal intraepithelial neoplasia, obstructive uropathy, and chronic kidney disease further demonstrated that he possibly suffered extensive tissue damage due to radiation to the pelvis. This case report explores the current diagnosis guidelines and treatment options for patients with radiation-induced PAD. Through this case study, we aim to bring awareness to this lesser-known cause of PAD among medical providers and promote research for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Claudia Aleman Oliva
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Erik Aleman Espino
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Michelle Demory Beckler
- Microbiology and Immunology, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Marc M Kesselman
- Rheumatology, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| |
Collapse
|
19
|
Aktas MS, Eren E, Kucukler S, Eroglu MS, Ilgun M, Yanar KE, Aydin O. Investigation of haematological, inflammatory and immunological response in naturally infected cattle with Theileria annulata. Parasite Immunol 2023; 45:e13002. [PMID: 37461131 DOI: 10.1111/pim.13002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 08/15/2023]
Abstract
In this study, we aimed to investigate haematological, pro-inflammatory, inflammatory, anti-inflammatory and immunological responses in naturally Theileria annulata-infected cattle. The study material consisted of 25 Simmental cattle, 2-4 years of age, one of which was a control group consisting of healthy animals (Control group, n = 10), and the other was a Theileria group that include animals positive for Theileria annulata (Theileria group, n = 15). Haematological analysis (red blood cell [RBC], haemoglobin [HGB], haematocrit [HCT]), pro-inflammatory (tumour necrosis factor-α [TNF-α], nuclear factor kappa B [NF-ĸB] and interleukin-1 beta, [IL-1β]), inflammatory (neutrophil-lymphocyte ratio [NLR]), anti-inflammatory (interleukin-10 [IL-10]) and antimicrobial peptide (CAMP) analyses were performed by using ELISA kit from blood samples. It was found that the rectal temperature of the Theileria group was found to be significantly higher (p < .001) than that of the control group. Haematological and biochemical analysis revealed that the RBC and HGB count and HCT percentage decreased (p < .001), while NF-ĸB (p < .001), TNF-α (p = .002), IL-1β (p < .001), IL-10 (p = .012), NLR (p < .001) and CAMP (p = .037) levels increased in Theileria group compared to the control group. There was a strong correlation between NF-ĸB and TNF-α, NF-ĸB and IL-10, NLR and IL-1β, NF-ĸB and CAMP, TNF-α and CAMP and IL-10 and CAMP. As a result of this study, it was revealed that a pro-inflammatory and immunological response also occurs along with the anti-inflammatory response in the inflammatory process.
Collapse
Affiliation(s)
- Mustafa Sinan Aktas
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Emre Eren
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Muhammed Sertac Eroglu
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Murat Ilgun
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Kerim Emre Yanar
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Omer Aydin
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
20
|
ShokriShokri F, Mozdarani H, Omrani MD. Rel-A/PACER/miR 7 Axis May Play a Role in Radiotherapy Treatment in Breast Cancer Patients. IRANIAN BIOMEDICAL JOURNAL 2023; 27:173-82. [PMID: 37507347 PMCID: PMC10507291 DOI: 10.61186/ibj.3901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/06/2023] [Indexed: 12/17/2023]
Abstract
Background Radiotherapy has become the standard form of treatment for breast cancer (BC). Radioresistance is an issue that limits the effectiveness of radiotherapy (RT). Therefore, predictive biomarkers are needed to choose the appropriate RT for the patient. Activation of the proinflammatory transcription factor, nuclear factor-kappa B (NF-κB), is a frequently noted pathway in BC. Investigating the relationship between RT and alterations in gene expression involved in the immune pathway can help better control the disease. This research investigated the impact of RT on the expression levels of Rel-A, PACER, and miR-7 within the NF-κB signaling pathway. Methods Blood samples (n = 15) were obtained from BC patients during four different time intervals: 72 hours prior to initiating RT, as well as one, two, and four weeks following RT completion. Samples were also collected from 20 healthy women who had no immune or cancer-related diseases. Blood RNA was extracted, and complementary DNA was synthesized. Gene expression level was determined using R real-time polymerase chain reaction (RT-PCR). Results There was a significant difference in the expression level of Rel-A between patients and normal individual blood samples (p < 0.05). After four weeks of RT, qRT-PCR revealed a significant downregulation of miR-7 and upregulation of Rel-A and PACER in BC patients. Also, there was a significant association between Rel-A expression and monocyte numbers during RT (p < 0.001). Conclusion The expression level of PACER, miR-7 and Rel-A, changed after RT; therefore, these genes could be used as diagnostic and therapeutic RT markers in BC.
Collapse
Affiliation(s)
- Fazlollah ShokriShokri
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran;
| | - Hossein Mozdarani
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran;
| | - Mir Davood Omrani
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
ShokriShokri F, Mozdarani H, Omrani MD. Rel-A/PACER/miR 7 Axis May Play a Role in Radiotherapy Treatment in Breast Cancer Patients. IRANIAN BIOMEDICAL JOURNAL 2023; 27:173-82. [PMID: 37507347 PMCID: PMC10507291 DOI: 10.52547/ibj.3901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/06/2023] [Indexed: 07/30/2023]
Abstract
Background Radiotherapy has become the standard form of treatment for breast cancer (BC). Radioresistance is an issue that limits the effectiveness of radiotherapy (RT). Therefore, predictive biomarkers are needed to choose the appropriate RT for the patient. Activation of the proinflammatory transcription factor, nuclear factor-kappa B (NF-κB), is a frequently noted pathway in BC. Investigating the relationship between RT and alterations in gene expression involved in the immune pathway can help better control the disease. This research investigated the impact of RT on the expression levels of Rel-A, PACER, and miR-7 within the NF-κB signaling pathway. Methods Blood samples (n = 15) were obtained from BC patients during four different time intervals: 72 hours prior to initiating RT, as well as one, two, and four weeks following RT completion. Samples were also collected from 20 healthy women who had no immune or cancer-related diseases. Blood RNA was extracted, and complementary DNA was synthesized. Gene expression level was determined using R real-time polymerase chain reaction (RT-PCR). Results There was a significant difference in the expression level of Rel-A between patients and normal individual blood samples (p < 0.05). After four weeks of RT, qRT-PCR revealed a significant downregulation of miR-7 and upregulation of Rel-A and PACER in BC patients. Also, there was a significant association between Rel-A expression and monocyte numbers during RT (p < 0.001). Conclusion The expression level of PACER, miR-7 and Rel-A, changed after RT; therefore, these genes could be used as diagnostic and therapeutic RT markers in BC.
Collapse
Affiliation(s)
- Fazlollah ShokriShokri
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran;
| | - Hossein Mozdarani
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran;
| | - Mir Davood Omrani
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Kostenko V, Akimov O, Gutnik O, Kostenko H, Kostenko V, Romantseva T, Morhun Y, Nazarenko S, Taran O. Modulation of redox-sensitive transcription factors with polyphenols as pathogenetically grounded approach in therapy of systemic inflammatory response. Heliyon 2023; 9:e15551. [PMID: 37180884 PMCID: PMC10171461 DOI: 10.1016/j.heliyon.2023.e15551] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/09/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
One of the adverse outcomes of acute inflammatory response is progressing to the chronic stage or transforming into an aggressive process, which can develop rapidly and result in the multiple organ dysfunction syndrome. The leading role in this process is played by the Systemic Inflammatory Response that is accompanied by the production of pro- and anti-inflammatory cytokines, acute phase proteins, and reactive oxygen and nitrogen species. The purpose of this review that highlights both the recent reports and the results of the authors' own research is to encourage scientists to develop new approaches to the differentiated therapy of various SIR manifestations (low- and high-grade systemic inflammatory response phenotypes) by modulating redox-sensitive transcription factors with polyphenols and to evaluate the saturation of the pharmaceutical market with appropriate dosage forms tailored for targeted delivery of these compounds. Redox-sensitive transcription factors such as NFκB, STAT3, AP1 and Nrf2 have a leading role in mechanisms of the formation of low- and high-grade systemic inflammatory phenotypes as variants of SIR. These phenotypic variants underlie the pathogenesis of the most dangerous diseases of internal organs, endocrine and nervous systems, surgical pathologies, and post-traumatic disorders. The use of individual chemical compounds of the class of polyphenols, or their combinations can be an effective technology in the therapy of SIR. Administering natural polyphenols in oral dosage forms is very beneficial in the therapy and management of the number of diseases accompanied with low-grade systemic inflammatory phenotype. The therapy of diseases associated with high-grade systemic inflammatory phenotype requires medicinal phenol preparations manufactured for parenteral administration.
Collapse
Affiliation(s)
- Vitalii Kostenko
- Poltava State Medical University, Department of Pathophysiology, Ukraine
| | - Oleh Akimov
- Poltava State Medical University, Department of Pathophysiology, Ukraine
- Corresponding author.
| | - Oleksandr Gutnik
- Poltava State Medical University, Department of Pathophysiology, Ukraine
| | - Heorhii Kostenko
- Poltava State Medical University, Department of Pathophysiology, Ukraine
| | - Viktoriia Kostenko
- Poltava State Medical University, Department of Foreign Languages with Latin and Medical Terminology, Ukraine
| | - Tamara Romantseva
- Poltava State Medical University, Department of Pathophysiology, Ukraine
| | - Yevhen Morhun
- Poltava State Medical University, Department of Pathophysiology, Ukraine
| | - Svitlana Nazarenko
- Poltava State Medical University, Department of Pathophysiology, Ukraine
| | - Olena Taran
- Poltava State Medical University, Department of Pathophysiology, Ukraine
| |
Collapse
|
23
|
MCPIP1 Suppresses the NF-κB Signaling Pathway Through Negative Regulation of K63-Linked Ubiquitylation of TRAF6 in Colorectal Cancer. Cancer Gene Ther 2023; 30:96-107. [PMID: 36076064 DOI: 10.1038/s41417-022-00528-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/10/2022] [Accepted: 08/24/2022] [Indexed: 01/19/2023]
Abstract
The abnormal activation of the nuclear factor-kappa B (NF-κB) signaling pathway is an important precipitating factor for the inception and development of colorectal cancer (CRC), one of the most common tumors worldwide. As a pro-apoptotic transcription factor, monocyte chemotactic protein-induced protein 1 (MCPIP1) has been closely associated with many tumor types. In the present study, the expression of MCPIP1 was firstly discovered reduced in CRC tissues and correlated with poor patient prognosis. The decreased expression was caused by promoter hypermethylation. Overexpressed MCPIP1 was found to inhibit the proliferative and migratory abilities of CRC cells, whereas knockdown of MCPIP1 produced the opposite result. The subsequent investigation demonstrated that MCPIP1 exerted its "anti-cancer" effect by suppression of the NF-κB signaling pathway through negative regulation of K63-linked ubiquitylation of TNF receptor associated factor 6 (TRAF6). Therefore, our results indicate a prognostic marker for CRC and a theoretical basis for MCPIP1 as a treatment.
Collapse
|
24
|
Xhuti D, Rebalka IA, Minhas M, May L, Murphy K, Nederveen JP, Tarnopolsky MA. The Acute Effect of Multi-Ingredient Antioxidant Supplementation following Ionizing Radiation. Nutrients 2023; 15:nu15010207. [PMID: 36615864 PMCID: PMC9823556 DOI: 10.3390/nu15010207] [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: 12/07/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Radiation exposure is an undeniable health threat encountered in various occupations and procedures. High energy waves in ionizing radiation cause DNA damage and induce reactive oxygen species (ROS) production, which further exacerbate DNA, protein, and lipid damage, increasing risk of mutations. Although endogenous antioxidants such as superoxide dismutase have evolved to upregulate and neutralize ROS, exogenous dietary antioxidants also have the potential to combat ionizing radiation (IR)-induced ROS production. We evaluated a cocktail of ingredients (AOX) purported to have antioxidant and mitochondrial protective properties on the acute effects of IR. We show that IR stimulates DNA damage through phosphorylation of DNA repair proteins in the heart, brain, and liver of mice. AOX showed partial protection in brain and liver, through a lack of significant activation in given repair proteins. In addition, AOX attenuated the IR-induced increase in NF-kβ mRNA and protein expression in brain and liver. Lastly, cytochrome c oxidase complex transcripts were significantly higher in heart and brain following radiation, which was also diminished by prior ingestion of AOX. Together, our findings suggest that a multi-ingredient AOX supplement may attenuate the IR-induced cellular damage response and represents a feasible and cost-effective preventative supplement for at-risk populations of radiation exposure.
Collapse
Affiliation(s)
- Donald Xhuti
- Department of Pediatrics, McMaster University Health Sciences Centre, Hamilton, ON L8N 3Z5, Canada
- Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON L8N 3Z5, Canada
| | - Irena A. Rebalka
- Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON L8N 3Z5, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Mahek Minhas
- Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON L8N 3Z5, Canada
| | - Linda May
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kieran Murphy
- Department of Medical Imaging, University of Toronto, Toronto, ON M5S 2C5, Canada
- Cora Therapeutics, Toronto, ON M5K 1N2, Canada
| | - Joshua P. Nederveen
- Department of Pediatrics, McMaster University Health Sciences Centre, Hamilton, ON L8N 3Z5, Canada
| | - Mark A. Tarnopolsky
- Department of Pediatrics, McMaster University Health Sciences Centre, Hamilton, ON L8N 3Z5, Canada
- Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON L8N 3Z5, Canada
- Correspondence: ; Tel.: +1-905-521-2100 (ext. 76593); Fax: +1-905-577-8380
| |
Collapse
|
25
|
Maines LW, Schrecengost RS, Zhuang Y, Keller SN, Smith RA, Green CL, Smith CD. Opaganib Protects against Radiation Toxicity: Implications for Homeland Security and Antitumor Radiotherapy. Int J Mol Sci 2022; 23:13191. [PMID: 36361977 PMCID: PMC9655569 DOI: 10.3390/ijms232113191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 12/25/2023] Open
Abstract
Exposure to ionizing radiation (IR) is a lingering threat from accidental or terroristic nuclear events, but is also widely used in cancer therapy. In both cases, host inflammatory responses to IR damage normal tissue causing morbidity and possibly mortality to the victim/patient. Opaganib, a first-in-class inhibitor of sphingolipid metabolism, has broad anti-inflammatory and anticancer activity. Opaganib elevates ceramide and reduces sphingosine 1-phosphate (S1P) in cells, conditions that increase the antitumor efficacy of radiation while concomitantly suppressing inflammatory damage to normal tissue. Therefore, opaganib may suppress toxicity from unintended IR exposure and improve patient response to chemoradiation. To test these hypotheses, we first examined the effects of opaganib on the toxicity and antitumor activity of radiation in mice exposed to total body irradiation (TBI) or IR with partial bone marrow shielding. Oral treatment with opaganib 2 h before TBI shifted the LD75 from 9.5 Gy to 11.5 Gy, and provided substantial protection against gastrointestinal damage associated with suppression of radiation-induced elevations of S1P and TNFα in the small intestines. In the partially shielded model, opaganib provided dose-dependent survival advantages when administered 4 h before or 24 h after radiation exposure, and was particularly effective when given both prior to and following radiation. Relevant to cancer radiotherapy, opaganib decreased the sensitivity of IEC6 (non-transformed mouse intestinal epithelial) cells to radiation, while sensitizing PAN02 cells to in vitro radiation. Next, the in vivo effects of opaganib in combination with radiation were examined in a syngeneic tumor model consisting of C57BL/6 mice bearing xenografts of PAN02 pancreatic cancer cells and a cross-species xenograft model consisting of nude mice bearing xenografts of human FaDu cells. Mice were treated with opaganib and/or IR (plus cisplatin in the case of FaDu tumors). In both tumor models, the optimal suppression of tumor growth was attained by the combination of opaganib with IR (± cisplatin). Overall, opaganib substantially protects normal tissue from radiation damage that may occur through unintended exposure or cancer radiotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Charles D. Smith
- Apogee Biotechnology Corporation, 1214 Research Blvd, Suite 2015, Hummelstown, PA 17036, USA
| |
Collapse
|
26
|
Russ E, Davis CM, Slaven JE, Bradfield DT, Selwyn RG, Day RM. Comparison of the Medical Uses and Cellular Effects of High and Low Linear Energy Transfer Radiation. TOXICS 2022; 10:toxics10100628. [PMID: 36287908 PMCID: PMC9609561 DOI: 10.3390/toxics10100628] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 05/14/2023]
Abstract
Exposure to ionizing radiation can occur during medical treatments, from naturally occurring sources in the environment, or as the result of a nuclear accident or thermonuclear war. The severity of cellular damage from ionizing radiation exposure is dependent upon a number of factors including the absorbed radiation dose of the exposure (energy absorbed per unit mass of the exposure), dose rate, area and volume of tissue exposed, type of radiation (e.g., X-rays, high-energy gamma rays, protons, or neutrons) and linear energy transfer. While the dose, the dose rate, and dose distribution in tissue are aspects of a radiation exposure that can be varied experimentally or in medical treatments, the LET and eV are inherent characteristics of the type of radiation. High-LET radiation deposits a higher concentration of energy in a shorter distance when traversing tissue compared with low-LET radiation. The different biological effects of high and low LET with similar energies have been documented in vivo in animal models and in cultured cells. High-LET results in intense macromolecular damage and more cell death. Findings indicate that while both low- and high-LET radiation activate non-homologous end-joining DNA repair activity, efficient repair of high-LET radiation requires the homologous recombination repair pathway. Low- and high-LET radiation activate p53 transcription factor activity in most cells, but high LET activates NF-kB transcription factor at lower radiation doses than low-LET radiation. Here we review the development, uses, and current understanding of the cellular effects of low- and high-LET radiation exposure.
Collapse
Affiliation(s)
- Eric Russ
- Graduate Program of Cellular and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Catherine M. Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - John E. Slaven
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Dmitry T. Bradfield
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Reed G. Selwyn
- Department of Radiology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Correspondence:
| |
Collapse
|
27
|
Lae Lae Phoo N, Sukhamwang A, Dejkriengkraikul P, Yodkeeree S. Diclofenac Sensitizes Signet Ring Cell Gastric Carcinoma Cells to Cisplatin by Activating Autophagy and Inhibition of Survival Signal Pathways. Int J Mol Sci 2022; 23:ijms232012066. [PMID: 36292923 PMCID: PMC9602524 DOI: 10.3390/ijms232012066] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Gastric cancer has one of the highest incidence rates of cancer worldwide while also contributing to increased drug resistance among patients in clinical practice. Herein, we have investigated the role of diclofenac (DCF) on sensitizing cisplatin resistance in signet ring cell gastric carcinoma cells (SRCGC). Non-toxic concentrations of DCF significantly augmented cisplatin-induced cell death in cisplatin-resistant SRCGC cells (KATO/DDP) but not in cisplatin-sensitive SRCGC cells (KATOIII). Consistently, concomitant treatment of DCF and cisplatin significantly enhanced autophagic cell death due to overproduction of intracellular reactive oxygen species (ROS). At the molecular level, the induction of ROS has been associated with a reduction in antioxidant enzymes expression while inhibiting nuclear factor erythroid 2-related factor 2 (Nrf2) activity. Moreover, the combination of DCF and cisplatin also inhibited the expression of survival proteins including Bcl-2, Bcl-xL, cIAP1 and cyclin D1 in KATO/DDP cells when compared with cisplatin alone. This was due, at least in part, to reduce MAPKs, Akt, NF-κB, AP-1 and STAT-3 activation. Taken together, our results suggested that DCF potentiated the anticancer effect of cisplatin in SRCGC via the regeneration of intracellular ROS, which in turn promoted cell death as an autophagy mechanism and potentially modulated the cell survival signal transduction pathway.
Collapse
Affiliation(s)
- Nang Lae Lae Phoo
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Amonnat Sukhamwang
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pornngarm Dejkriengkraikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Supachai Yodkeeree
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence:
| |
Collapse
|
28
|
King L, Bernaitis N, Christie D, Chess-Williams R, Sellers D, McDermott C, Dare W, Anoopkumar-Dukie S. Drivers of Radioresistance in Prostate Cancer. J Clin Med 2022; 11:jcm11195637. [PMID: 36233505 PMCID: PMC9573022 DOI: 10.3390/jcm11195637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/15/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer (PCa) is the second most commonly diagnosed cancer worldwide. Radiotherapy remains one of the first-line treatments in localised disease and may be used as monotherapy or in combination with other treatments such as androgen deprivation therapy or radical prostatectomy. Despite advancements in delivery methods and techniques, radiotherapy has been unable to totally overcome radioresistance resulting in treatment failure or recurrence of previously treated PCa. Various factors have been linked to the development of tumour radioresistance including abnormal tumour vasculature, oxygen depletion, glucose and energy deprivation, changes in gene expression and proteome alterations. Understanding the biological mechanisms behind radioresistance is essential in the development of therapies that are able to produce both initial and sustained response to radiotherapy. This review will investigate the different biological mechanisms utilised by PCa tumours to drive radioresistance.
Collapse
Affiliation(s)
- Liam King
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4215, Australia or
- Ramsay Pharmacy Group, Melbourne, VIC 3004, Australia
| | - Nijole Bernaitis
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4215, Australia or
| | - David Christie
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4215, Australia or
- GenesisCare, Gold Coast, QLD 4224, Australia
- Faculty of Health Sciences & Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Russ Chess-Williams
- Faculty of Health Sciences & Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Donna Sellers
- Faculty of Health Sciences & Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Catherine McDermott
- Faculty of Health Sciences & Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Wendy Dare
- Ramsay Pharmacy Group, Melbourne, VIC 3004, Australia
| | - Shailendra Anoopkumar-Dukie
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4215, Australia or
- Correspondence: ; Tel.: +61-(0)-7-5552-7725
| |
Collapse
|
29
|
NFκB1 Polymorphisms Are Associated with Severe Influenza A (H1N1) Virus Infection in a Canadian Population. Microorganisms 2022; 10:microorganisms10101886. [PMID: 36296162 PMCID: PMC9606957 DOI: 10.3390/microorganisms10101886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022] Open
Abstract
Background: We examined associations between NFκB1 polymorphisms and influenza A (H1N1) clinical outcomes in Canadian. Methods: A total of thirty-six Caucasian patients admitted to the intensive care unit (ICU) in hospitals in Canada were recruited during the 2009 H1N1 pandemic. Genomic DNA was extracted from the whole blood samples. The NFkB1 gene was targeted for genotyping using next-generation sequencing technology—Roche 454. Results: A total of 136 single nucleotide polymorphisms (SNPs) were discovered within the NFκB1 gene. Among them, 63 SNPs were significantly enriched in patients admitted in the ICU (p < 0.05) compared with the British Caucasian population in the 1000 Genomes study. These enriched SNPs are mainly intron variants, and only two are exon SNPs from the non-transcribing portion of the NFκB1 gene. Conclusions: Genetic variations in the NFκB1 gene could influence clinical outcomes of pandemic H1N1 infections. Our findings showed that sequence variations of the NFκB1 gene might influence patient response to influenza infection.
Collapse
|
30
|
Biological Mechanisms to Reduce Radioresistance and Increase the Efficacy of Radiotherapy: State of the Art. Int J Mol Sci 2022; 23:ijms231810211. [PMID: 36142122 PMCID: PMC9499172 DOI: 10.3390/ijms231810211] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Cancer treatment with ionizing radiation (IR) is a well-established and effective clinical method to fight different types of tumors and is a palliative treatment to cure metastatic stages. Approximately half of all cancer patients undergo radiotherapy (RT) according to clinical protocols that employ two types of ionizing radiation: sparsely IR (i.e., X-rays) and densely IR (i.e., protons). Most cancer cells irradiated with therapeutic doses exhibit radio-induced cytotoxicity in terms of cell proliferation arrest and cell death by apoptosis. Nevertheless, despite the more tailored advances in RT protocols in the last few years, several tumors show a relatively high percentage of RT failure and tumor relapse due to their radioresistance. To counteract this extremely complex phenomenon and improve clinical protocols, several factors associated with radioresistance, of both a molecular and cellular nature, must be considered. Tumor genetics/epigenetics, tumor microenvironment, tumor metabolism, and the presence of non-malignant cells (i.e., fibroblast-associated cancer cells, macrophage-associated cancer cells, tumor-infiltrating lymphocytes, endothelial cells, cancer stem cells) are the main factors important in determining the tumor response to IR. Here, we attempt to provide an overview of how such factors can be taken advantage of in clinical strategies targeting radioresistant tumors.
Collapse
|
31
|
Sharapov MG, Goncharov RG, Parfenyuk SB, Glushkova OV. Effect of Peroxiredoxin 6 on p53 Transcription Factor Level. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:839-849. [PMID: 36171649 DOI: 10.1134/s0006297922080156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 06/16/2023]
Abstract
Peroxiredoxin 6 (Prdx6) is an important antioxidant enzyme with multiple functions in the cell. Prdx6 neutralizes a wide range of hydroperoxides, participates in phospholipid metabolism and cell membrane repair, and in transmission of intracellular and intercellular signals. Disruption of normal Prdx6 expression in the cell leads to the development of pathological conditions. Decrease in the Prdx6 concentration leads to increase in oxidative damage to the cell. At the same time, hyperproduction of Prdx6 is associated with increase in antioxidant status, suppression of apoptosis, and carcinogenesis. Currently, mechanisms of carcinogenic action of peroxiredoxins are poorly understood. In this work we established that the 3-4-fold increase in Prdx6 production in mouse embryonic fibroblast 3T3 cells leads to the 4-5-fold decrease in the level of oncosuppressor p53. At the same time, hyperproduction of Prdx6 leads to the increased expression of RELA and HIF1A, which have oncogenic effects. The 3-4-fold increase in intracellular Prdx6 increases intensity of cell proliferation by 20-30%, promotes increase in antioxidant activity by 30-50%, and increases radioresistance of the transfected 3T3 cells by 30-40%. Increase of the level of intranuclear Prdx6 leads to the decrease in expression of the DNA repair genes in response to radiation, indicating decrease in the genomic DNA damage. This work discusses possible molecular mechanisms of p53 suppression during Prdx6 hyperproduction, which could be used in the development of new approaches in cancer therapy.
Collapse
Affiliation(s)
- Mars G Sharapov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Ruslan G Goncharov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Svetlana B Parfenyuk
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Olga V Glushkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
32
|
Chaudhury D, Sen U, Sahoo BK, Bhat NN, Kumara K S, Karunakara N, Biswas S, Shenoy P S, Bose B. Thorium promotes lung, liver and kidney damage in BALB/c mouse via alterations in antioxidant systems. Chem Biol Interact 2022; 363:109977. [PMID: 35636501 DOI: 10.1016/j.cbi.2022.109977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/20/2022] [Accepted: 05/04/2022] [Indexed: 01/15/2023]
|
33
|
Parthenolide and Its Soluble Analogues: Multitasking Compounds with Antitumor Properties. Biomedicines 2022; 10:biomedicines10020514. [PMID: 35203723 PMCID: PMC8962426 DOI: 10.3390/biomedicines10020514] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 12/23/2022] Open
Abstract
Due to its chemical properties and multiple molecular effects on different tumor cell types, the sesquiterpene lactone parthenolide (PN) can be considered an effective drug with significant potential in cancer therapy. PN has been shown to induce either classic apoptosis or alternative caspase-independent forms of cell death in many tumor models. The therapeutical potential of PN has been increased by chemical design and synthesis of more soluble analogues including dimethylaminoparthenolide (DMAPT). This review focuses on the molecular mechanisms of both PN and analogues action in tumor models, highlighting their effects on gene expression, signal transduction and execution of different types of cell death. Recent findings indicate that these compounds not only inhibit prosurvival transcriptional factors such as NF-κB and STATs but can also determine the activation of specific death pathways, increasing intracellular reactive oxygen species (ROS) production and modifications of Bcl-2 family members. An intriguing property of these compounds is its specific targeting of cancer stem cells. The unusual actions of PN and its analogues make these agents good candidates for molecular targeted cancer therapy.
Collapse
|
34
|
Chargari C, Rassy E, Helissey C, Achkar S, Francois S, Deutsch E. Impact of radiation therapy on healthy tissues. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 376:69-98. [PMID: 36997270 DOI: 10.1016/bs.ircmb.2022.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Radiation therapy has a fundamental role in the management of cancers. However, despite a constant improvement in radiotherapy techniques, the issue of radiation-induced side effects remains clinically relevant. Mechanisms of acute toxicity and late fibrosis are therefore important topics for translational research to improve the quality of life of patients treated with ionizing radiations. Tissue changes observed after radiotherapy are consequences of complex pathophysiology, involving macrophage activation, cytokine cascade, fibrotic changes, vascularization disorders, hypoxia, tissue destruction and subsequent chronic wound healing. Moreover, numerous data show the impact of these changes in the irradiated stroma on the oncogenic process, with interplays between tumor radiation response and pathways involved in the fibrotic process. The mechanisms of radiation-induced normal tissue inflammation are reviewed, with a focus on the impact of the inflammatory process on the onset of treatment-related toxicities and the oncogenic process. Possible targets for pharmacomodulation are also discussed.
Collapse
|
35
|
Mun H, Townley HE. Mechanism of Action of the Sesquiterpene Compound Helenalin in Rhabdomyosarcoma Cells. Pharmaceuticals (Basel) 2021; 14:ph14121258. [PMID: 34959659 PMCID: PMC8703838 DOI: 10.3390/ph14121258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is the most frequent soft tissue sarcoma in paediatric patients. Relapsed or refractory RMS shows very low 5-year survival rates, which urgently necessitates new chemotherapy agents. Herein, the sesquiterpene lactone, helenalin, was investigated as a new potential therapeutic agent against the embryonal RMS (eRMS) and alveolar RMS (aRMS) cells. We have evaluated in vitro antiproliferative efficacy of helenalin on RMS cells by the MTT and wound healing assay, and estimated several cell death pathways by flow cytometry, confocal microscopy and immunoblotting. It was shown that helenalin was able to increase reactive oxygen species levels, decrease mitochondrial membrane potential, trigger endoplasmic reticulum stress and deactivate the NF-κB pathway. Confirmation was obtained through the use of antagonistic compounds which alleviated the effects of helenalin in the corresponding pathways. Our findings demonstrate that oxidative stress is the pivotal mechanism of action of helenalin in promoting RMS cell death in vitro.
Collapse
Affiliation(s)
- Hakmin Mun
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK;
| | - Helen Elizabeth Townley
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK;
- Department of Engineering Science, University of Oxford, Parks Road, Oxford OX1 3PJ, UK
- Correspondence: ; Tel.: +44-01865283792
| |
Collapse
|
36
|
Regulation of Nuclear Factor-KappaB (NF-κB) signaling pathway by non-coding RNAs in cancer: Inhibiting or promoting carcinogenesis? Cancer Lett 2021; 509:63-80. [PMID: 33838282 DOI: 10.1016/j.canlet.2021.03.025] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/18/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022]
Abstract
The nuclear factor-kappaB (NF-κB) signaling pathway is considered as a potential therapeutic target in cancer therapy. It has been well established that transcription factor NF-κB is involved in regulating physiological and pathological events including inflammation, immune response and differentiation. Increasing evidences suggest that deregulated NF-κB signaling can enhance cancer cell proliferation, metastasis and also mediate radio-as well as chemo-resistance. On the contrary, non-coding RNAs (ncRNAs) have been found to modulate NF-κB signaling pathway under different settings. MicroRNAs (miRNAs) can dually inhibit/induce NF-κB signaling thereby affecting the growth and migration of cancer cells. Furthermore, the response of cancer cells to radiotherapy and chemotherapy may also be regulated by miRNAs. Regulation of NF-κB by miRNAs may be mediated via binding to 3/-UTR region. Interestingly, anti-tumor compounds can increase the expression of tumor-suppressor miRNAs in inhibiting NF-κB activation and the progression of cancers. Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) can also effectively modulate NF-κB signaling thus affecting tumorigenesis. It is noteworthy that several studies have demonstrated that lncRNAs and circRNAs can affect miRNAs in targeting NF-κB activation. They can act as competing endogenous RNA (ceRNA) thereby reducing miRNA expression to induce NF-κB activation that can in turn promote cancer progression and malignancy.
Collapse
|
37
|
Kalekhan F, Kudva AK, Raghu SV, Rao S, Hegde SK, Simon P, Baliga MS. Traditionally Used Natural Products in Preventing Ionizing Radiation-Induced Dermatitis: First Review on the Clinical Studies. Anticancer Agents Med Chem 2021; 22:64-82. [PMID: 33820524 DOI: 10.2174/1871520621666210405093236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/16/2020] [Accepted: 01/15/2021] [Indexed: 11/22/2022]
Abstract
In the treatment of cancer, the use of ionizing radiation is an important modality. However, on the downside, radiation, when used for curative purposes, causes acute dermatitis or radiodermatitis at the site of radiation in most individuals. From a clinical viewpoint, severe dermatitis causes a burning and itching sensation is very painful, and severely affects the quality of life of the individual undergoing treatment. In worse situations, acute radiation dermatitis can cause gaps or breaks in the planned treatment and this can adversely affect the treatment objective and outcome. BACKGROUND In various traditional and folk systems of medicine, plants and plant products have been used since time immemorial for treating various skin ailments. Further, many cosmeceutical creams formulated based on knowledge from ethnomedicinal use are marketed and used to treat various ailments. In the current review, an attempt is made at summarizing the beneficial effects of some plants and plant products in mitigating acute radiation dermatitis in humans undergoing curative radiotherapy. Additionally, the emphasis is also placed on the mechanism/s responsible for the beneficial effects. OBJECTIVE The objective of this review is to summarize the clinical observations on the prevention of radiodermatitis by plant products. In this review, the protective effects of Adlay (Coix lachryma-jobi L.) bran extract, Aloe vera, Calendula officinalis, Cucumis sativus, green tea constituent the epigallocatechin-3-gallate, honey, Achillea millefolium, Matricaria chamomilla, olive oil and some polyherbal creams are addressed by also addressing on the mechanism of action for the beneficial effects. METHODS Two authors' data mined for information in Google Scholar, PubMed, Embase and the Cochrane Library for publications in the field from 1901 up to July 2020. The focus was on acute radiation dermatitis, ionizing radiation, curative radiotherapy, human cancer. The articles were collected and analyzed. RESULTS For the first time, this review addresses the usefulness of natural products like adlay bran, Aloe vera, Calendula officinalis, Cucumis sativus, green tea constituent the epigallocatechin-3-gallate, honey, Achillea millefolium, Matricaria chamomilla, olive oil and some experimentally constituted and commercially available polyherbal creams as skincare agents against the deleterious effects of ionizing radiation on the skin. The protective effects are possibly due to the free radical scavenging, antioxidant, anti-inflammatory, wound healing and skin protective effects. CONCLUSION The authors suggest that these plants have been used since antiquity as medicinal agents and require in-depth investigation with both clinical and preclinical validated models of study. The results of these studies will be extremely useful to cancer patients requiring curative radiotherapy, the dermatology fraternity, agro-based and pharmaceutical sectors at large.
Collapse
Affiliation(s)
- Faizan Kalekhan
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Avinash K Kudva
- Department of Biochemistry, Mangalore University, Mangalagangotri, Karnataka. India
| | - Shamprasad V Raghu
- Neurogenetics Laboratory, Department of Applied Zoology, Mangalore University, Mangalagangotri, Karnataka. India
| | - Suresh Rao
- Radiation Oncology, Mangalore Institute of Oncology, Mangalore, Karnataka. India
| | - Sanath K Hegde
- Radiation Oncology, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Paul Simon
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Manjeshwar S Baliga
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| |
Collapse
|
38
|
Sharapov MG, Glushkova OV, Parfenyuk SB, Gudkov SV, Lunin SM, Novoselova EG. The role of TLR4/NF-κB signaling in the radioprotective effects of exogenous Prdx6. Arch Biochem Biophys 2021; 702:108830. [PMID: 33727039 DOI: 10.1016/j.abb.2021.108830] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/26/2021] [Accepted: 03/07/2021] [Indexed: 01/11/2023]
Abstract
Peroxiredoxin 6 (Prdx6) is a bifunctional enzyme with multi-substrate peroxidase and phospholipase activities that is involved in cell redox homeostasis and regulates intracellular processes. Previously, recombinant Prdx6 was shown to exert a radioprotective effect during whole-body exposure to a lethal dose of X-ray radiation. Moreover, a mutant form Prdx6-C47S, which lacks peroxidase activity, also had a radioprotective effect, and this indicates that the mechanism of radioprotection is unknown. The present study was aimed to test the hypothesis that the radioprotective effect of Prdx6 and Prdx6-C47S may be mediated through the TLR4/NF-κB signaling pathway. It was demonstrated that exogenously applied Prdx6 protected 3T3 fibroblast cells against LD50 X-ray radiation in vitro. Pretreatment with Prdx6 increased cell survival, stimulated proliferation, normalized the level of reactive oxygen species in culture, and suppressed apoptosis and necrosis. Wild-type Prdx6 and, to a lesser degree, the Prdx6-C47S mutant proteins promoted a significant increase in NF-κB activation in irradiated cells, which likely contributes to the antiapoptotic effect. Pretreatment with TLR4 inhibitors, especially those directed to the extracellular part of the receptor, significantly reduced the radioprotective effect, and this supports the role of TLR4 signaling in the protective effects of Prdx6. Therefore, the radioprotective effect of Prdx6 was related not only to its antioxidant properties, but also to its ability to trigger cellular defense mechanisms through interaction with the TLR4 receptor and subsequent activation of the NF-κB pathway. Recombinant Prdx6 may be useful for the development of a new class of safe radioprotective compounds that have a combination of antioxidant and immunomodulatory properties.
Collapse
Affiliation(s)
- Mars G Sharapov
- Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS, Pushchino, Russia.
| | - Olga V Glushkova
- Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS, Pushchino, Russia
| | - Svetlana B Parfenyuk
- Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS, Pushchino, Russia
| | - Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
| | - Sergey M Lunin
- Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS, Pushchino, Russia
| | - Elena G Novoselova
- Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS, Pushchino, Russia
| |
Collapse
|
39
|
Chen J, Wei Z, Wang Y, Long M, Wu W, Kuca K. Fumonisin B 1: Mechanisms of toxicity and biological detoxification progress in animals. Food Chem Toxicol 2021; 149:111977. [PMID: 33428988 DOI: 10.1016/j.fct.2021.111977] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 01/21/2023]
Abstract
Fumonisin B1 (FB1) is a toxic secondary metabolite produced by the Fusarium molds that can contaminate food and feed. It has been found that FB1 can cause systemic toxicity, including neurotoxicity, hepatotoxicity, nephrotoxicity and mammalian cytotoxicity. This review addresses the toxicity studies carried out on FB1 and outlines the probable mechanisms underlying its immunotoxicity, reproductive toxicity, joint toxicity, apoptosis, and autophagy. In the present work, the research progress of FB1 detoxification in recent years is reviewed, which provides reference for controlling and reducing the toxicity of FB1.
Collapse
Affiliation(s)
- Jia Chen
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Zhen Wei
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Yan Wang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Miao Long
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic.
| |
Collapse
|
40
|
Mohamed HA, Said RS. Coenzyme Q10 attenuates inflammation and fibrosis implicated in radiation enteropathy through suppression of NF-kB/TGF-β/MMP-9 pathways. Int Immunopharmacol 2021; 92:107347. [PMID: 33418245 DOI: 10.1016/j.intimp.2020.107347] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 01/01/2023]
Abstract
Radiation enteropathy is one the most common clinical issue for patients receiving radiotherapy for abdominal/pelvic tumors which severely affect the quality of life of cancer patients due to dysplastic lesions (ischemia, ulcer, or fibrosis) that aggravate the radiation damage. Herein, this study demonstrated the prophylactic role of coenzyme Q10 (CoQ10), a powerful antioxidant, against radiotherapy-induced gastrointestinal injury. Male Sprague Dawley rats were divided into four groups: group 1 was defined as control, and group 2 was the irradiated group. Group 3 and 4 were CoQ10 control and radiation plus CoQ10 groups, respectively. CoQ10 (10 mg/kg) was orally administered for 10 days before 10 Gy whole-body radiation and was continued for 4 days post-irradiation. CoQ10 administration protected rats delivered a lethal dose of ϒ-radiation from changes in crypt-villus structures and promoted regeneration of the intestinal epithelium. CoQ10 attenuated radiation-induced oxidative stress by decreasing lipid peroxidation and increasing the antioxidant enzyme catalase activity and reduced glutathione level. CoQ10 also counteracts inflammatory response mediated after radiation exposure through downregulating intestinal NF-ĸB expression which subsequently decreased the level of inflammatory cytokine IL-6 and the expression of COX-2. Radiation-induced intestinal fibrosis confirmed via Masson's trichrome staining occurred through upregulating transforming growth factor (TGF)-β1 and matrix metalloproteinase (MMP)-9 expression, while CoQ10 administration significantly diminishes these effects which further confirmed the anti-fibrotic property of CoQ10. Therefore, CoQ10 is a promising radioprotector that could prevent intestinal complications and enhance the therapeutic ratio of radiotherapy in patients with pelvic tumors through suppressing the NF-kB/TGF-β1/MMP-9 signaling pathway.
Collapse
Affiliation(s)
- Heba A Mohamed
- Department of Drug Radiation Research, National Center for Radiation Research & Technology, Atomic Energy Authority, Cairo, Egypt
| | - Riham S Said
- Department of Drug Radiation Research, National Center for Radiation Research & Technology, Atomic Energy Authority, Cairo, Egypt.
| |
Collapse
|
41
|
Singh VK, Seed TM. Entolimod as a radiation countermeasure for acute radiation syndrome. Drug Discov Today 2020; 26:17-30. [PMID: 33065293 DOI: 10.1016/j.drudis.2020.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/12/2020] [Accepted: 10/05/2020] [Indexed: 01/11/2023]
Abstract
High doses of total-body or partial-body radiation exposure can result in a life-threatening acute radiation syndrome as manifested by severe morbidity. Entolimod (CBLB502) is effective in protecting against, and mitigating the development of, the hematopoietic and gastrointestinal subsyndromes of the acute radiation syndrome in rodents and nonhuman primates. Entolimod treatment reduces radiation-induced apoptosis and accelerates the regeneration of progenitors in radiation-damaged tissues. The drug has been evaluated clinically for its pharmacokinetics (PK), toxicity, and biomarkers. The US Food and Drug Administration (FDA) has granted investigational new drug, fast-track, and orphan drug statuses to entolimod. Its safety, efficacy, and animal-to-human dose conversion data allowed its progression with a pre-emergency use authorization application submission.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Thomas M Seed
- Tech Micro Services, 4417 Maple Avenue, Bethesda, MD 20814, USA
| |
Collapse
|
42
|
Sarsarshahi S, Madjd Z, Bozsaky E, Kowaliuk J, Kuess P, Ghahremani MH, Doerr W. An evaluation of the effect of bortezomib on radiation-induced urinary bladder dysfunction. Strahlenther Onkol 2019; 195:934-939. [PMID: 31363801 DOI: 10.1007/s00066-019-01497-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/02/2019] [Indexed: 01/12/2023]
Abstract
PURPOSE The urinary bladder is one major organ at risk in radiotherapy of pelvic malignancies. The radiation response manifests in early and chronic changes in bladder function. These are based on inflammatory effects and changes in urothelial cell function and proliferation. This study evaluates the effect of bortezomib as an anti-proliferative and anti-inflammatory compound in an established mouse bladder model. The early radiation-induced bladder dysfunction in the mouse occurs in two phases during the first month after irradiation (phase I: day 0-15, phase II: days 16-30). MATERIALS AND METHODS Daily bortezomib injections (0.02 mg/ml, subcutaneously) were administered between days 0-15 or 15-30 in separate groups. Single graded radiation doses were administered in five dose groups. Cystometry was carried out before (individual control) and during the first month after irradiation. When bladder capacity was decreased by ≥50%, mice were considered as responders. Statistical analysis was performed by the SPSS software version 24. RESULTS Daily bortezomib injections between days 0-15 resulted in a significant decrease in responders for phase I. There was no significant effect with daily bortezomib injections between days 16-30. CONCLUSION Two separate waves of acute radiation-induced urinary bladder dysfunction have distinct mechanisms that need further biological studies.
Collapse
Affiliation(s)
- Sina Sarsarshahi
- Department of Molecular Medicine, Iran University of Medical Science, 14496-14530, Tehran, Iran.,ATRAB-Applied and Translational Radiobiology, Department of Radiation Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Zahra Madjd
- Department of Molecular Medicine, Iran University of Medical Science, 14496-14530, Tehran, Iran. .,Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Eva Bozsaky
- ATRAB-Applied and Translational Radiobiology, Department of Radiation Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Jakob Kowaliuk
- ATRAB-Applied and Translational Radiobiology, Department of Radiation Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Peter Kuess
- Christian Doppler Laboratory for Medical Radiation Research for Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Mohammad Hossein Ghahremani
- Department of Pharmacology-Toxicology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran.,Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Wolfgang Doerr
- ATRAB-Applied and Translational Radiobiology, Department of Radiation Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
43
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Shabeeb D, Musa AE. NF‐κB targeting for overcoming tumor resistance and normal tissues toxicity. J Cell Physiol 2019; 234:17187-17204. [DOI: 10.1002/jcp.28504] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy School of Medicine, Kurdistan University of Medical Sciences Sanandaj Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department School of Paramedical Sciences, Kermanshah University of Medical Sciences Kermanshah Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology Faculty of Paramedical Sciences, Kashan University of Medical Sciences Kashan Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center Faculty of Pharmacy, Mazandaran University of Medical Sciences Sari Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology College of Medicine, University of Misan Misan Iraq
| | - Ahmed E. Musa
- Department of Medical Physics Tehran University of Medical Sciences (International Campus) Tehran Iran
| |
Collapse
|
44
|
Singh VK, Garcia M, Wise SY, Seed TM. Medical countermeasures for unwanted CBRN exposures: Part I chemical and biological threats with review of recent countermeasure patents. Expert Opin Ther Pat 2016; 26:1431-1447. [DOI: 10.1080/13543776.2017.1233178] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Vijay K. Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Melissa Garcia
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Stephen Y. Wise
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | |
Collapse
|