1
|
Adhab AH, Altalbawy FMA, Mahdi MS, Baldaniya L, Omar TM, Ganesan S, Juneja B, Pathak PK, Mansoor AS, Radi UK, Abd NS, Kadhim M. NADPH Oxidases in Cancer Therapy-Induced Cardiotoxicity: Mechanisms and Therapeutic Approaches. Cardiovasc Toxicol 2025; 25:631-649. [PMID: 39966326 DOI: 10.1007/s12012-025-09976-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/13/2025] [Indexed: 02/20/2025]
Abstract
Cancer therapy-induced cardiotoxicity remains a significant clinical challenge, limiting the efficacy of cancer treatments and impacting long-term survival and quality of life. NADPH oxidases, a family of enzymes that are able to generate reactive oxygen species (ROS), have emerged as key players in the pathogenesis of cardiotoxicity associated with various cancer therapies. This review comprehensively examines the role of NADPH oxidases in cancer therapy-induced cardiotoxicity, elucidating the underlying mechanisms and exploring potential therapeutic approaches. We discuss the structure and function of NADPH oxidases in the cardiovascular system and their involvement in cardiotoxicity induced by anthracyclines and ionizing radiation. The molecular mechanisms by which NADPH oxidase-derived ROS contribute to cardiac injury are explored, including direct oxidative damage, activation of pro-apoptotic pathways, mitochondrial dysfunction, vascular damage, inflammation, fibrosis, and others. Furthermore, we evaluate therapeutic strategies targeting NADPH oxidases, such as specific inhibitors, antioxidant therapies, natural products, and other cardioprotectors. The review also addresses current challenges in the field, including the need for isoform-specific targeting and the identification of reliable biomarkers. Finally, we highlight future research directions aimed at mitigating NADPH oxidase-mediated cardiotoxicity and alleviating cardiovascular side effects in cancer survivors. By synthesizing current knowledge and identifying knowledge gaps, this review provides a rationale for future studies and the development of novel cardioprotective strategies in cancer therapy.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | | | - Lalji Baldaniya
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Thabit Moath Omar
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Nineveh, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Bhanu Juneja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | | | - Usama Kadem Radi
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Nasr Saadoun Abd
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Munther Kadhim
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
2
|
Gupta J, Almulla AF, Jalil AT, Jasim NY, Aminov Z, Alsaikhan F, Ramaiah P, Chinnasamy L, Jawhar ZH. Melatonin in Chemo/Radiation Therapy; Implications for Normal Tissues Sparing and Tumor Suppression: An Updated Review. Curr Med Chem 2025; 32:511-538. [PMID: 37916636 DOI: 10.2174/0109298673262122231011172100] [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: 05/12/2023] [Revised: 07/20/2023] [Accepted: 09/01/2023] [Indexed: 11/03/2023]
Abstract
Resistance to therapy and the toxicity of normal tissue are the major problems for efficacy associated with chemotherapy and radiotherapy. Drug resistance is responsible for most cases of mortality associated with cancer. Furthermore, their side effects can decrease the quality of life for surviving patients. An enhancement in the tumor response to therapy and alleviation of toxic effects remain unsolved challenges. One of the interesting topics is the administration of agents with low toxicity to protect normal tissues and/or sensitize cancers to chemo/radiotherapy. Melatonin is a natural body hormone that is known as a multitasking molecule. Although it has antioxidant properties, a large number of experiments have uncovered interesting effects of melatonin that can increase the therapeutic efficacy of chemo/radiation therapy. Melatonin can enhance anticancer therapy efficacy through various mechanisms, cells such as the immune system, and modulation of cell cycle and death pathways, tumor suppressor genes, and also through suppression of some drug resistance mediators. However, melatonin may protect normal tissues through the suppression of inflammation, fibrosis, and massive oxidative stress in normal cells and tissues. In this review, we will discuss the distinct effects of melatonin on both tumors and normal tissues. We review how melatonin may enhance radio/chemosensitivity of tumors while protecting normal tissues such as the lung, heart, gastrointestinal system, reproductive system, brain, liver, and kidney.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India
| | - Abbas F Almulla
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | | | | | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
3
|
Li YL, Wang G, Wang BW, Li YH, Ma YX, Huang Y, Yan WT, Xie P. The potential treatment of N-acetylcysteine as an antioxidant in the radiation-induced heart disease. Cardiovasc Diagn Ther 2024; 14:509-524. [PMID: 39263473 PMCID: PMC11384455 DOI: 10.21037/cdt-24-19] [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: 01/09/2024] [Accepted: 07/11/2024] [Indexed: 09/13/2024]
Abstract
Background Radiation-induced heart disease (RIHD) is a serious complication of thoracic tumor radiotherapy that substantially affects the quality of life of cancer patients. Oxidative stress plays a pivotal role in the occurrence and progression of RIHD, which prompted our investigation of an innovative approach for treating RIHD using antioxidant therapy. Methods We used 8-week-old male Sprague-Dawley (SD) rats as experimental animals and H9C2 cells as experimental cells. N-acetylcysteine (NAC) was used as an antioxidant to treat H9C2 cells after X-ray irradiation in this study. In the present study, the extent of cardiomyocyte damage caused by X-ray exposure was determined, alterations in oxidation/antioxidation levels were assessed, and changes in the expression of genes related to mitochondria were examined. The degree of myocardial tissue and cell injury was also determined. Dihydroethidium (DHE) staining, reactive oxygen species (ROS) assays, and glutathione (GSH) and manganese superoxide dismutase (Mn-SOD) assays were used to assess cell oxidation/antioxidation. Flow cytometry was used to determine the mitochondrial membrane potential and mitochondrial permeability transition pore (mPTP) opening. High-throughput transcriptome sequencing and bioinformatics analysis were used to elucidate the expression of mitochondria-related genes in myocardial tissue induced by X-ray exposure. Polymerase chain reaction (PCR) was used to verify the expression of differentially expressed genes. Results X-ray irradiation damaged myocardial tissue and cells, resulting in an imbalance of oxidative and antioxidant substances and mitochondrial damage. NAC treatment increased cell counting kit-8 (CCK-8) levels (P=0.02) and decreased lactate dehydrogenase (LDH) release (P=0.02) in cardiomyocytes. It also reduced the level of ROS (P=0.002) and increased the levels of GSH (P=0.04) and Mn-SOD (P=0.01). The mitochondrial membrane potential was restored (P<0.001), and mPTP opening was inhibited (P<0.001). Transcriptome sequencing and subsequent validation analyses revealed a decrease in the expression of mitochondria-related genes in myocardial tissue induced by X-ray exposure, but antioxidant therapy did not reverse the related DNA damage. Conclusions Antioxidants mitigated radiation-induced myocardial damage to a certain degree, but these agents did not reverse the associated DNA damage. These findings provide a new direction for future investigations by our research group, including exploring the treatment of RIHD-related DNA damage.
Collapse
Affiliation(s)
- Yan-Ling Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Department of Cardiovascular Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Gang Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Bo-Wen Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Department of Cardiovascular Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yong-Hong Li
- The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yong-Xia Ma
- Department of Cardiovascular Medicine, The Second People's Hospital of Lanzhou City, Lanzhou, China
| | - Yuan Huang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Wen-Ting Yan
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ping Xie
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Department of Cardiovascular Medicine, Gansu Provincial Hospital, Lanzhou, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Li YS, Xia J, Chen CY, Ren SH, He MR. Upregulated dual oxidase 1-induced oxidative stress and caspase-1-dependent pyroptosis reflect the etiologies of heart failure. BMC Mol Cell Biol 2024; 25:16. [PMID: 38750444 PMCID: PMC11094974 DOI: 10.1186/s12860-024-00506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 03/08/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Oxidative stress is implicated in the pathogenesis of heart failure. Dual oxidase 1 (DUOX1) might be important in heart failure development through its mediating role in oxidative stress. This study was designed to evaluate the potential role of DUOX1 in heart failure. MATERIALS AND METHODS AC16 cells were treated with 2 µmol/L of doxorubicin (DOX) for 12, 24, and 48 h to construct a heart failure model. DUOX1 overexpression and silencing in AC16 cell were established. DUOX1 expression was detected by Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Pyroptosis and reactive oxygen species (ROS) production were measured by flow cytometry. RESULTS Increased DUOX1 expression levels were observed after DOX treatment for 24 h in AC16 cells. DUOX1 silencing inhibited DOX-induced pyroptosis and ROS production. The release of IL-1β, IL-18, and lactate dehydrogenase (LDH), and expression levels of pyroptosis-related proteins were also decreased. DUOX1 overexpression increased pyroptosis, ROS production, IL-1β, IL-18, and LDH release, and pyroptosis-related protein expression. N-acetyl-cysteine (NAC) significantly reversed DUOX1-induced pyroptosis, ROS, and related factors. CONCLUSION These results suggest that DUOX1-derived genotoxicity could promote heart failure development. In the process, oxidative stress and pyroptosis may be involved in the regulation of DUOX1 in heart failure.
Collapse
Affiliation(s)
- Yan Song Li
- Department of Cardiovasology, Shanghai Songjiang District Center Hospital, NO.748, Zhongshan Middle Road, Songjiang District, Shanghai, 201600, China.
- Department of Cardiovasology, Shanghai Baoshan District Luodian Hospital, No. 88 Yongshun Road, Baoshan District, Shanghai, 201908, China.
| | - Jingwen Xia
- Department of Cardiovasology, Shanghai Songjiang District Center Hospital, NO.748, Zhongshan Middle Road, Songjiang District, Shanghai, 201600, China
| | - Chang Yuan Chen
- Department of Cardiovasology, Shanghai Songjiang District Center Hospital, NO.748, Zhongshan Middle Road, Songjiang District, Shanghai, 201600, China
| | - Shu Hong Ren
- Department of Cardiovasology, Shanghai Songjiang District Center Hospital, NO.748, Zhongshan Middle Road, Songjiang District, Shanghai, 201600, China
| | - Mao Rong He
- Department of Cardiovasology, Shanghai Songjiang District Center Hospital, NO.748, Zhongshan Middle Road, Songjiang District, Shanghai, 201600, China
| |
Collapse
|
5
|
Li K, Chen W, Ma L, Yan L, Wang B. Approaches for reducing chemo/radiation-induced cardiotoxicity by nanoparticles. ENVIRONMENTAL RESEARCH 2024; 244:117264. [PMID: 37776941 DOI: 10.1016/j.envres.2023.117264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Nanoparticles are fascinating and encouraging carriers for cancer treatment due to their extraordinary properties and potential applications in targeted drug delivery, treatment, and diagnosis. Experimental studies including in vitro and in vivo examinations show that nanoparticles can cause a revolution in different aspects of cancer therapy. Normal tissue toxicity and early and late consequences are the major limitations of cancer therapy by radiotherapy and chemotherapy. However, the delivery of drugs into tumors or reducing the accumulation of drugs in normal tissues can permit a more satisfactory response of malignancies to therapy with more inferior side effects. Cardiac toxicity is one of the major problems for chemotherapy and radiotherapy. Therefore, several experimental studies have been performed to minimize the degenerative impacts of cancer treatment on the heart and also enhance the influences of radiotherapy and chemotherapy agents in cancers. This review article emphasizes the benefits of nanoparticle-based drug delivery techniques, including minimizing the exposure of the heart to anticancer drugs, enhancing the accumulation of drugs in cancers, and expanding the effectiveness of radiotherapy. The article also discusses the challenges and problems accompanied with nanoparticle-based drug delivery techniques such as toxicity, which need to be addressed through further research. Moreover, the article emphasizes the importance of developing safe and effective nanoparticle-based therapies that can be translated into clinical practice.
Collapse
Affiliation(s)
- Ketao Li
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Wan Chen
- Department of Cardiology, Jiulongpo First People's Hospital, Chongqing, 400051, China
| | - Liping Ma
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Laixing Yan
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Bing Wang
- Department of Cardiology, Zouping People's Hospital, Zouping, shandong, 256299, China.
| |
Collapse
|
6
|
Gupta J, Jalil AT, Riyad Muedii ZAH, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Farhood B. The Radiosensitizing Potentials of Silymarin/Silibinin in Cancer: A Systematic Review. Curr Med Chem 2024; 31:6992-7014. [PMID: 37921180 DOI: 10.2174/0109298673248404231006052436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Although radiotherapy is one of the main cancer treatment modalities, exposing healthy organs/tissues to ionizing radiation during treatment and tumor resistance to ionizing radiation are the chief challenges of radiotherapy that can lead to different adverse effects. It was shown that the combined treatment of radiotherapy and natural bioactive compounds (such as silymarin/silibinin) can alleviate the ionizing radiation-induced adverse side effects and induce synergies between these therapeutic modalities. In the present review, the potential radiosensitization effects of silymarin/silibinin during cancer radiation exposure/radiotherapy were studied. METHODS According to the PRISMA guideline, a systematic search was performed for the identification of relevant studies in different electronic databases of Google Scholar, PubMed, Web of Science, and Scopus up to October 2022. We screened 843 articles in accordance with a predefined set of inclusion and exclusion criteria. Seven studies were finally included in this systematic review. RESULTS Compared to the control group, the cell survival/proliferation of cancer cells treated with ionizing radiation was considerably less, and silymarin/silibinin administration synergistically increased ionizing radiation-induced cytotoxicity. Furthermore, there was a decrease in the tumor volume, weight, and growth of ionizing radiation-treated mice as compared to the untreated groups, and these diminutions were predominant in those treated with radiotherapy plus silymarin/ silibinin. Furthermore, the irradiation led to a set of biochemical and histopathological changes in tumoral cells/tissues, and the ionizing radiation-induced alterations were synergized following silymarin/silibinin administration (in most cases). CONCLUSION In most cases, silymarin/silibinin administration could sensitize the cancer cells to ionizing radiation through an increase of free radical formation, induction of DNA damage, increase of apoptosis, inhibition of angiogenesis and metastasis, etc. However, suggesting the use of silymarin/silibinin during radiotherapeutic treatment of cancer patients requires further clinical studies.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, U.P., India
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Andrés Alexis Ramírez-Coronel
- Psychometry and Ethology Laboratory, Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellin, Colombia
- Educational Statistics Research Group (GIEE), National University of Education, Cuenca, Ecuador
| | | | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
7
|
Klunko NS, Achmad H, Abdullah TM, Mohammed S, Saha I, Salim KS, Obaid RF, Romero-Parra RM, Al-Hasnawi SS, Al-Janabi WH, Farhood B. The Anti-hypoxia Potentials of Trans-sodium Crocetinate in Hypoxiarelated Diseases: A Review. Curr Radiopharm 2024; 17:30-37. [PMID: 37877507 DOI: 10.2174/0118744710268127231020083505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023]
Abstract
Crocetin is a kind of apocarotenoid carboxylic acid extracted from saffron (Crocus sativus L.), which is effective in upregulating tissue oxygenation. However, crocetin is difficult to solubilize. It was shown that the trans isomer of crocetin is effective in improving oxygen diffusivity, while its cis isomer appears not to be. Hence, the isolated trans isomer of crocetin or trans-sodium crocetinate (TSC) can be used instead of crocetin. It is shown that TSC can upregulate hypoxic tissue oxygenation and be effective in treating some hypoxia-related diseases. Moreover, experimental and clinical studies have reported no adverse effects following TSC treatment, even at high doses. The current study will discuss the potential role of TSC in hemorrhagic shock, ischemia, brain tumor radiotherapy, and others.
Collapse
Affiliation(s)
- Natalia S Klunko
- Department of Training of Scientific and Scientific-Pedagogical Personnel, Russian New University, Moscow, Russia
| | - Harun Achmad
- Department of Pediatric Dentistry, Faculty of Dentistry, Hasanuddin University, Makassar, Indonesia
| | | | - Sami Mohammed
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Indranil Saha
- Department of Physics, GLA University, Mathura, Pin- 281406, U.P., India
| | | | - Rasha Fadhel Obaid
- Department of Biomedical Engineering, Al-Mustaqbal University College, Babylon, Iraq
| | | | | | | | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
8
|
Aslam MA, Ahmad H, Malik HS, Uinarni H, Karim YS, Akhmedov YM, Abdelbasset WK, Awadh SA, Abid MK, Mustafa YF, Farhood B, Sahebkar A. Radiotherapy-associated Sensorineural Hearing Loss in Pediatric Oncology Patients. Curr Med Chem 2024; 31:5351-5369. [PMID: 37190814 DOI: 10.2174/0929867330666230515112245] [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: 11/25/2022] [Revised: 03/08/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023]
Abstract
During the radiotherapeutic treatment of pediatric oncology patients, they would be at a latent risk of developing ionizing radiation-induced ototoxicity when the cochlea or auditory nerve is located within the radiation field. Sensorineural hearing loss (SNHL) is an irreversible late complication of radiotherapy, and its incidence depends on various factors such as the patient's hearing sensitivity, total radiation dose to the cochlea, radiotherapy fractionation regimen, age and chemoradiation. Importantly, this complication exhibits serious challenges to adult survivors of childhood cancer, as it has been linked to impairments in academic achievement, psychosocial development, independent living skills, and employment in the survivor population. Therefore, early detection and proper management can alleviate academic, speech, language, social, and psychological morbidity arising from hearing deficits. In the present review, we have addressed issues such as underlying mechanisms of radiation-induced SNHL, audiometric findings of pediatric cancer patients treated with radiotherapy, and management and protection measures against radiation-induced ototoxicity.
Collapse
Affiliation(s)
- Muhammad Ammar Aslam
- Department of Emergency Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Hassaan Ahmad
- Department of Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Hamza Sultan Malik
- Department of Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Herlina Uinarni
- Department of Anatomy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
- Radiologist at Pantai Indah Kapuk Hospital, Jakarta, Indonesia
| | | | - Yusuf Makhmudovich Akhmedov
- Department of Pediatric Surgery, Samarkand State Medical Institute, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, Makhtumkuli Street 103, Tashkent, 100047, Uzbekistan
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Sura A Awadh
- Department of Anesthesia, Al-Mustaqbal University, Babylon, Iraq
| | - Mohammed Kadhem Abid
- Department of Anesthesia, College of Health & medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhosein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Basirat U, Bin Tariq U, Moeen N, Jawhar ZH, Shoja SJ, Kareem AK, Ramírez-Coronel AA, Romero-Parra RM, Zabibah RS, Gupta J, Mustafa YF, Farhood B. A Systematic Review of the Chemo/Radioprotective Effects of Melatonin against Ototoxic Adverse Effects Induced by Chemotherapy and Radiotherapy. Curr Pharm Des 2023; 29:1218-1229. [PMID: 37138418 DOI: 10.2174/1381612829666230503145707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/05/2023] [Accepted: 03/13/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND Although chemotherapy and radiotherapy are effective in cancer treatment, different adverse effects induced by these therapeutic modalities (such as ototoxicity) restrict their clinical use. Co-treatment of melatonin may alleviate the chemotherapy/radiotherapy-induced ototoxicity. OBJECTIVE In the present study, the otoprotective potentials of melatonin against the ototoxicity induced by chemotherapy and radiotherapy were reviewed. METHODS According to the PRISMA guideline, a systematic search was carried out to identify all relevant studies on "the role of melatonin against ototoxic damage associated with chemotherapy and radiotherapy" in the different electronic databases up to September 2022. Sixty-seven articles were screened based on a predefined set of inclusion and exclusion criteria. Seven eligible studies were finally included in this review. RESULTS The in vitro findings showed that cisplatin chemotherapy significantly decreased the auditory cell viability compared to the control group; in contrast, the melatonin co-administration increased the cell viability of cisplatin-treated cells. The results obtained from the distortion product otoacoustic emission (DPOAE) and auditory brainstem response (ABR) tests demonstrated a decreased amplitude of DPOAE and increased values of ABR I-IV interval and ABR threshold in mice/rats receiving radiotherapy and cisplatin; nevertheless, melatonin co-treatment indicated an opposite pattern on these evaluated parameters. It was also found that cisplatin and radiotherapy could significantly induce the histological and biochemical changes in the auditory cells/tissue. However, melatonin co-treatment resulted in alleviating the cisplatin/radiotherapy-induced biochemical and histological changes. CONCLUSION According to the findings, it was shown that melatonin co-treatment alleviates the ototoxic damage induced by chemotherapy and radiotherapy. Mechanically, melatonin may exert its otoprotective effects via its anti-oxidant, anti-apoptotic, and anti-inflammatory activities and other mechanisms.
Collapse
Affiliation(s)
| | | | - Nawal Moeen
- Nawaz Sharif Medical College, Gujrat, Pakistan
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Sarah Jawad Shoja
- College of Health & Medical Technology, Al-Ayen University, Nasiriyah, Iraq
| | - Ali Kamil Kareem
- Biomedical Engineering Department, Al-Mustaqbal University College, Babylon, Iraq
| | | | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
10
|
Yalcin B, Yay AH, Tan FC, Özdamar S, Yildiz OG. Investigation of the anti-oxidative and anti-inflammatory effects of melatonin on experimental liver damage by radiation. Pathol Res Pract 2023; 246:154477. [PMID: 37148837 DOI: 10.1016/j.prp.2023.154477] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/19/2023] [Accepted: 04/20/2023] [Indexed: 05/08/2023]
Abstract
Radiotherapy is one of the inevitable treatment approaches for several types of cancer. We aimed to show the protective and therapeutic effects of daily use of melatonin on liver tissues subjected to a single dose of 10 Gy (gamma-ray) total body radiation. Rats were divided into 6 groups, of which 10 were in each: control, sham, melatonin, radiation, radiation+melatonin, and melatonin+radiation. The rats received 10 Gy of external radiation throughout their entire bodies. The rats were given 10 mg/kg/day of melatonin intraperitoneally before or after radiation treatment, depending on the group. Histological methods, immunohistochemical analysis (Caspase-3, Sirtuin-1, α-SMA, NFΚB-p65), biochemical analysis by ELİSA (SOD, CAT, GSH-PX, MDA, TNF-α, TGF-β, PDGF, PGC-1α) and the Comet assay as a marker of DNA damage were applied to the liver tissues. Histopathological examinations showed structural changes in the liver tissue of the radiation group. Radiation treatment increased the immunoreactivity of Caspase-3, Sirtuin-1 and α-SMA, but these effects were relatively attenuated in the melatonin-treated groups. The melatonin+radiation group had statistically significant results close to those of the control group, in terms of Caspase-3, NFΚB-p65 and Sirtuin-1 immunoreactivity. In melatonin treated groups, hepatic biochemical markers, MDA, SOD, TNF-α, TGF-β levels, and DNA damage parameters were decreased. Administration of melatonin before and after radiation has beneficial effects, but using it before radiation may be more efficient. Accordingly, daily melatonin usage could mitigate ionizing radiation induced damage.
Collapse
Affiliation(s)
- Betul Yalcin
- Adıyaman University, Faculty of Medicine, Department of Histology and Embryology, Adıyaman, Turkey.
| | - Arzu Hanım Yay
- Erciyes University, Faculty of Medicine, Department of Histology and Embryology, Kayseri, Turkey; Erciyes University, Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Fazile Cantürk Tan
- Erciyes University, Faculty of Medicine, Department of Biophysics, Kayseri, Turkey
| | - Saim Özdamar
- Pamukkale University, Faculty of Medicine, Department of Histology and Embryology, Kayseri, Turkey
| | - Oğuz Galip Yildiz
- Erciyes University, Faculty of Medicine, Department of Radiation Oncology, Kayseri, Turkey
| |
Collapse
|
11
|
Sarkar S, Karmakar S, Basu M, Ghosh P, Ghosh MK. Neurological damages in COVID-19 patients: Mechanisms and preventive interventions. MedComm (Beijing) 2023; 4:e247. [PMID: 37035134 PMCID: PMC10080216 DOI: 10.1002/mco2.247] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/14/2023] [Accepted: 03/01/2023] [Indexed: 04/11/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus, causes coronavirus disease 2019 (COVID-19) which led to neurological damage and increased mortality worldwide in its second and third waves. It is associated with systemic inflammation, myocardial infarction, neurological illness including ischemic strokes (e.g., cardiac and cerebral ischemia), and even death through multi-organ failure. At the early stage, the virus infects the lung epithelial cells and is slowly transmitted to the other organs including the gastrointestinal tract, blood vessels, kidneys, heart, and brain. The neurological effect of the virus is mainly due to hypoxia-driven reactive oxygen species (ROS) and generated cytokine storm. Internalization of SARS-CoV-2 triggers ROS production and modulation of the immunological cascade which ultimately initiates the hypercoagulable state and vascular thrombosis. Suppression of immunological machinery and inhibition of ROS play an important role in neurological disturbances. So, COVID-19 associated damage to the central nervous system, patients need special care to prevent multi-organ failure at later stages of disease progression. Here in this review, we are selectively discussing these issues and possible antioxidant-based prevention therapies for COVID-19-associated neurological damage that leads to multi-organ failure.
Collapse
Affiliation(s)
- Sibani Sarkar
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| | - Subhajit Karmakar
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| | - Malini Basu
- Department of MicrobiologyDhruba Chand Halder College, University of CalcuttaDakshin BarasatWBIndia
| | - Pratyasha Ghosh
- Department of EconomicsBethune CollegeUniversity of CalcuttaKolkataIndia
| | - Mrinal K Ghosh
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| |
Collapse
|
12
|
Protective Potentials of Alpha-Lipoic Acid against Ionizing Radiation-Induced Brain Damage in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:4999306. [PMID: 36778212 PMCID: PMC9918365 DOI: 10.1155/2023/4999306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 02/05/2023]
Abstract
Background This study was aimed at determining the effects of alpha-lipoic acid on ionizing irradiation-induced oxidative damage and apoptosis in the brain of rats. Methods The animals were exposed to whole-brain X-radiation with a 15 Gy single dose in the absence or presence of alpha-lipoic acid (200 mg/kg body weight) pretreatment for one week. The rats were divided into four groups (5 rats in each group): vehicle control, alpha-lipoic acid alone (ALA), radiation alone (RAD), and radiation plus alpha-lipoic acid (RAD+ALA). In the next stage, malondialdehyde (MDA), nitric oxide, catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) in the brain tissue of the rats were measured. Furthermore, the Western blot analysis technique was performed to assess the NOX2, NOX4, and caspase-3 protein expression levels. Results Twenty-four hours after the irradiation, MDA and nitric oxide levels in the irradiated rats were significantly higher than those in the control group (p < 0.001); however, the pretreatment with alpha-lipoic acid resulted in a significant reduction in these stress oxidative markers (p < 0.05). Moreover, a significant decrease in CAT, SOD, and GPx levels was observed in the radiation group alone compared to the control group (p < 0.01); in contrast, the activities of these antioxidant enzymes significantly increased in the radiation plus alpha-lipoic acid group in comparison to the radiation group alone (p < 0.05). The results of Western blot analysis revealed that NOX2, NOX4, and caspase-3 protein expressions significantly elevated in the irradiated rats compared to the control group (p < 0.001). The pretreatment with alpha-lipoic acid could significantly decrease the expression levels of NOX2, NOX4, and caspase-3 in comparison with the radiation group alone (p < 0.05). Conclusion According to the obtained findings, it can be mentioned that the alpha-lipoic acid pretreatment could mitigate the ionizing irradiation-induced oxidative damage and apoptosis in the brain of the rats.
Collapse
|
13
|
Wang KX, Ye C, Yang X, Ma P, Yan C, Luo L. New Insights into the Understanding of Mechanisms of Radiation-Induced Heart Disease. Curr Treat Options Oncol 2023; 24:12-29. [PMID: 36598620 DOI: 10.1007/s11864-022-01041-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 01/05/2023]
Abstract
OPINION STATEMENT Cancer patients who receive high-dose thoracic radiotherapy may develop radiation-induced heart disease (RIHD). The clinical presentation of RIHD comprises coronary artery atherosclerosis, valvular disease, pericarditis, cardiomyopathy, and conduction defects. These complications have significantly reduced due to the improved radiotherapy techniques. However, such methods still could not avoid heart radiation exposure. Furthermore, people who received relatively low-dose radiation exposures have exhibited significantly elevated RIHD risks in cohort studies of atomic bomb survivors and occupational exposures. The increased potential in exposure to natural and artificial ionizing radiation sources has emphasized the necessity to understand the development of RIHD. The pathological processes of RIHD include endothelial dysfunction, inflammation, fibrosis, and hypertrophy. The underlying mechanisms may involve the changes in oxidative stress, DNA damage response, telomere erosion, mitochondrial dysfunction, epigenetic regulation, circulation factors, protein post-translational modification, and metabolites. This review will discuss the recent advances in the mechanisms of RIHD at cellular and molecular levels.
Collapse
Affiliation(s)
- Kai-Xuan Wang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Cong Ye
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Xu Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Ping Ma
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Chen Yan
- Department of Rheumatology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330006, People's Republic of China.
| | - Lan Luo
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China.
| |
Collapse
|
14
|
Xu C, Shang Z, Najafi M. Lung Pneumonitis and Fibrosis in Cancer Therapy: A Review on Cellular and Molecular Mechanisms. Curr Drug Targets 2022; 23:1505-1525. [PMID: 36082868 DOI: 10.2174/1389450123666220907144131] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/05/2022] [Accepted: 08/02/2022] [Indexed: 01/25/2023]
Abstract
Fibrosis and pneumonitis are the most important side effects of lung tissue following cancer therapy. Radiotherapy and chemotherapy by some drugs, such as bleomycin, can induce pneumonitis and fibrosis. Targeted therapy and immunotherapy also may induce pneumonitis and fibrosis to a lesser extent compared to chemotherapy and radiotherapy. Activation of lymphocytes by immunotherapy or infiltration of inflammatory cells such as macrophages, lymphocytes, neutrophils, and mast cells following chemo/radiation therapy can induce pneumonitis. Furthermore, the polarization of macrophages toward M2 cells and the release of anti-inflammatory cytokines stimulate fibrosis. Lung fibrosis and pneumonitis may also be potentiated by some other changes such as epithelial-mesenchymal transition (EMT), oxidative stress, reduction/oxidation (redox) responses, renin-angiotensin system, and the upregulation of some inflammatory mediators such as a nuclear factor of kappa B (NF-κB), inflammasome, cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS). Damages to the lung vascular system and the induction of hypoxia also can induce pulmonary injury following chemo/radiation therapy. This review explains various mechanisms of the induction of pneumonitis and lung fibrosis following cancer therapy. Furthermore, the targets and promising agents to mitigate lung fibrosis and pneumonitis will be discussed.
Collapse
Affiliation(s)
- Chaofeng Xu
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang, 311800, China
| | - Zhongtu Shang
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang, 311800, China
| | - Masoud Najafi
- Medical Technology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
15
|
Abstract
Cardiac remodelling is characterized by abnormal changes in the function and morphological properties such as diameter, mass, normal diameter of cavities, heart shape, fibrosis, thickening of vessels and heart layers, cardiomyopathy, infiltration of inflammatory cells, and some others. These damages are associated with damage to systolic and diastolic abnormalities, damage to ventricular function, and vascular remodelling, which may lead to heart failure and death. Exposure of the heart to radiation or anti-cancer drugs including chemotherapy drugs such as doxorubicin, receptor tyrosine kinase inhibitors (RTKIs) such as imatinib, and immune checkpoint inhibitors (ICIs) can induce several abnormal changes in the heart structure and function through the induction of inflammation and fibrosis, vascular remodelling, hypertrophy, and some others. This review aims to explain the basic mechanisms behind cardiac remodelling following cancer therapy by different anti-cancer modalities.
Collapse
|
16
|
Moslehi M, Moazamiyanfar R, Dakkali MS, Rezaei S, Rastegar-Pouyani N, Jafarzadeh E, Mouludi K, Khodamoradi E, Taeb S, Najafi M. Modulation of the immune system by melatonin; implications for cancer therapy. Int Immunopharmacol 2022; 108:108890. [PMID: 35623297 DOI: 10.1016/j.intimp.2022.108890] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/07/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022]
Abstract
Immune system interactions within the tumour have a key role in the resistance or sensitization of cancer cells to anti-cancer agents. On the other hand, activation of the immune system in normal tissues following chemotherapy or radiotherapy is associated with acute and late effects such as inflammation and fibrosis. Some immune responses can reduce the efficiency of anti-cancer therapy and also promote normal tissue toxicity. Modulation of immune responses can boost the efficiency of anti-tumour therapy and alleviate normal tissue toxicity. Melatonin is a natural body agent that has shown promising results for modulating tumour response to therapy and also alleviating normal tissue toxicity. This review tries to focus on the immunomodulatory actions of melatonin in both tumour and normal tissues. We will explain how anti-cancer drugs may cause toxicity for normal tissues and how tumours can adapt themselves to ionizing radiation and anti-cancer drugs. Then, cellular and molecular mechanisms of immunoregulatory effects of melatonin alone or combined with other anti-cancer agents will be discussed.
Collapse
Affiliation(s)
- Masoud Moslehi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Moazamiyanfar
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Sepideh Rezaei
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Fleming Bldg. Rm 112, Houston, TX 77204-5003, USA
| | - Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Emad Jafarzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Kave Mouludi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran; Medical Biotechnology Research Center, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran; Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
17
|
Applying global longitudinal strain in assessing cardiac dysfunction after radiotherapy among breast cancer patients: a systemic review and meta-analysis. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00493-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Nosrati H, Seidi F, Hosseinmirzaei A, Mousazadeh N, Mohammadi A, Ghaffarlou M, Danafar H, Conde J, Sharafi A. Prodrug Polymeric Nanoconjugates Encapsulating Gold Nanoparticles for Enhanced X-Ray Radiation Therapy in Breast Cancer. Adv Healthc Mater 2022; 11:e2102321. [PMID: 34800003 DOI: 10.1002/adhm.202102321] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Indexed: 12/13/2022]
Abstract
An optimal radiosensitizer with improved tumor retention has an important effect on tumor radiation therapy. Herein, gold nanoparticles (Au NPs) and drug-containing, mPEG-conjugated CUR (mPEG-CUR), self-assembled NPs (mPEG-CUR@Au) are developed and evaluated as a drug carrier and radiosensitizer in a breast cancer mice model. As a result, cancer therapy efficacy is improved significantly by applying all-in-one NPs to achieve synchronous chemoradiotherapy, as evidenced by studies evaluating cell viability, proliferation, and ROS production. In vivo anticancer experiments show that the mPEG-CUR@Au system improves the radiation sensitivity of 4T1 mammary carcinoma and completely abrogates breast cancer.
Collapse
Affiliation(s)
- Hamed Nosrati
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing, 210037, China
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Farzad Seidi
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing, 210037, China
| | - Ali Hosseinmirzaei
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Navid Mousazadeh
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Mohammadi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Hossein Danafar
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - João Conde
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, 1150-082, Portugal
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, 1150-082, Portugal
| | - Ali Sharafi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
19
|
Ribeiro AEAS, Ferreira EF, Leal JDS, Barberino RDS, Oliveira HPD, Palheta Junior RC. Involvement of MT2 receptors in protective effects of melatonin against cisplatin-induced gastrointestinal damage in mice. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
|
20
|
Fu X, Li M, Tang C, Huang Z, Najafi M. Targeting of cancer cell death mechanisms by resveratrol: a review. Apoptosis 2021; 26:561-573. [PMID: 34561763 DOI: 10.1007/s10495-021-01689-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2021] [Indexed: 12/11/2022]
Abstract
Cancer cell death is the utmost aim in cancer therapy. Anti-cancer agents can induce apoptosis, mitotic catastrophe, senescence, or autophagy through the production of free radicals and induction of DNA damage. However, cancer cells can acquire some new properties to adapt to anti-cancer agents. An increase in the incidence of apoptosis, mitotic catastrophe, senescence, and necrosis is in favor of overcoming tumor resistance to therapy. Although an increase in the autophagy process may help the survival of cancer cells, some studies indicated that stimulation of autophagy cell death may be useful for cancer therapy. Using some low toxic agents to amplify cancer cell death is interesting for the eradication of clonogenic cancer cells. Resveratrol (a polyphenol agent) may affect various signaling pathways related to cell death. It can induce death signals and also downregulate the expression of anti-apoptotic genes. Resveratrol has also been shown to modulate autophagy and induce mitotic catastrophe and senescence in some cancer cells. This review focuses on the important targets and mechanisms for the modulation of cancer cell death by resveratrol.
Collapse
Affiliation(s)
- Xiao Fu
- College of Basic Medicine, Shaoyang University, Shaoyang, 422000, China
| | - Mu Li
- College of Basic Medicine, Shaoyang University, Shaoyang, 422000, China
| | - Cuilian Tang
- Department of Obstetrics and Gynecology of the Second Affiliated Hospital, Shaoyang University, Shaoyang, 422000, China
| | - Zezhi Huang
- Shaoyang Key Laboratory of Molecular Biology Diagnosis, Shaoyang, 422000, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
21
|
Jafarpour SM, Shekarchi B, Bagheri H, Farhood B. The Radioprotective Effects of Melatonin and Nanoselenium on DNA Double-Strand Breaks in Peripheral Lymphocytes Caused by I-131. Indian J Nucl Med 2021; 36:134-139. [PMID: 34385783 PMCID: PMC8320833 DOI: 10.4103/ijnm.ijnm_179_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/28/2020] [Accepted: 10/03/2020] [Indexed: 12/30/2022] Open
Abstract
Background: One of the treatment modalities for thyroid cancer and hyperthyroidism is radioiodine-131 (I-131) therapy. The use of this therapeutic modality is not completely safe and can lead to oxidative stress, eventually DNA damages. However, these radiation-induced damages can be reduced by antioxidants. This study aimed to investigate the potential radioprotective effects of melatonin and selenium nanoparticles (SeNPs) on DNA double-stranded breaks (DSBs) caused by I-131. Materials and Methods: After obtaining informed consent, 6 ml blood was taken from each volunteer. The samples were divided into two general groups of control (without I-131) and with I-131. Each group was also divided into three subgroups, including without antioxidant, melatonin, and SeNPs. The samples of control group were incubated for 2 h after adding the antioxidants. The samples of I-131 group were first incubated for 1 h with the antioxidants and then the samples re-incubated for another 1 h after adding the I-131. Then, the samples were prepared for γH2AX assay. Results: The findings showed that after 1 h of incubation with 20 μCi I-131/2 mL, the DSB levels increased by 102.9% in comparison with the control group. In the I-131 group, there were significant reductions of the DSB levels after incubation with melatonin (P < 0.001) and SeNPs (P < 0.001) in comparison with the without antioxidant subgroup. Furthermore, the DSB levels at the melatonin + I-131 and the SeNPs + I-131 subgroups decreased to 38% and 30%, respectively, compared to the I-131 subgroup. Conclusion: According to the obtained findings, it can be concluded that the use of melatonin and SeNPs (as radioprotector agents) can reduce the DSB levels induced by I-131 in peripheral lymphocytes.
Collapse
Affiliation(s)
- Seyed Masoud Jafarpour
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Babak Shekarchi
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Hamed Bagheri
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedicine, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
22
|
El-Missiry MA, Shabana S, Ghazala SJ, Othman AI, Amer ME. Melatonin exerts a neuroprotective effect against γ-radiation-induced brain injury in the rat through the modulation of neurotransmitters, inflammatory cytokines, oxidative stress, and apoptosis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:31108-31121. [PMID: 33598836 DOI: 10.1007/s11356-021-12951-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/09/2021] [Indexed: 05/11/2023]
Abstract
The current study aimed to investigate the ameliorative effect of melatonin (MLT) against brain injury in rats undergoing whole-body exposure to γ-radiation. Male Wistar rats were whole-body exposed to 4-Gy γ-radiation from a cesium-137 source. MLT (10 mg/kg) was orally administrated 30 minutes before irradiation and continued once daily for 1 and 7 days after exposure. In the irradiated rats, the plasma levels of glutamate were increased, while the gamma-aminobutyric acid (GABA) levels were decreased, and MLT improved the disturbed glutamate and GABA levels. These effects paralleled an increase in pro-inflammatory cytokines (IL-1b, IL-6, and TNF-a) and C-reactive protein as well as a decrease in IL-10 in the plasma of the irradiated rats. MLT treatment markedly reduced these effects, indicating its anti-inflammatory impact. Immunohistochemical studies demonstrated a remarkable upregulation of caspase-3 and P53 expression, indicating the increased apoptosis in the brain of irradiated rats. MLT significantly downregulated the expression of these parameters compared with that in the irradiated rats, indicating its anti-apoptotic effect. Oxidative stress is developed in the brain as evidenced by increased levels of malondialdehyde; decreased activities of superoxide dismutase, catalase, and glutathione peroxidase; and decreased content of glutathione in the brain. MLT remarkably ameliorated the development of oxidative stress in the brain of the irradiated rats indicating its antioxidant impact. The histopathological results were consistent with the biochemical and immunohistochemical results and showed that MLT remarkably protected the histological structure of brain tissue compared with that in the irradiated rats. In conclusion, MLT showed potential neuroprotective properties by increasing the release of neurotransmitters, antioxidants, and anti-inflammatory factors and reducing pro-inflammatory cytokines and apoptosis in the brain of irradiated rats. MLT can be beneficial in clinical and occupational settings requiring radiation exposure; however, additional studies are required to elucidate its neuroprotective effect in humans.
Collapse
Affiliation(s)
| | - Sameh Shabana
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Sara J Ghazala
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Azza I Othman
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Maggie E Amer
- Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
23
|
Sheikholeslami S, Khodaverdian S, Dorri-Giv M, Mohammad Hosseini S, Souri S, Abedi-Firouzjah R, Zamani H, Dastranj L, Farhood B. The radioprotective effects of alpha-lipoic acid on radiotherapy-induced toxicities: A systematic review. Int Immunopharmacol 2021; 96:107741. [PMID: 33989970 DOI: 10.1016/j.intimp.2021.107741] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/16/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Radiation therapy is one of the main cancer treatment modalities applied in 50-70% of cancer patients. Despite the many advantages of this treatment, such as non-invasiveness, organ-preservation, and spatiotemporal flexibility in tumor targeting, it can lead to complications in irradiated healthy cells/tissues. In this regard, the use of radio-protective agents can alleviate radiation-induced complications. This study aimed to review the potential role of alpha-lipoic acid in the prevention/reduction of radiation-induced toxicities on healthy cells/tissues. METHODS A systematic search was performed following PRISMA guidelines to identify relevant literature on the "role of alpha-lipoic acid in the treatment of radiotherapy-induced toxicity" in the electronic databases of Web of Science, Embase, PubMed, and Scopus up to January 2021. Based on the inclusion and exclusion criteria of the present study, 278 articles were screened. Finally, 29 articles were included in this systematic review. RESULTS The obtained results showed that in experimental in vivo models, the radiation-treated groups had decreased survival rate and body weight compared to the control groups. It was also found that radiation can induce mild to severe toxicities on gastrointestinal, circulatory, reproductive, central nervous, respiratory, endocrine, exocrine systems, etc. However, the use of alpha-lipoic acid could alleviate the radiation-induced toxicities in most cases. This radio-protective agent exerts its effects through mechanisms of anti-oxidant, anti-apoptosis, anti-inflammatory, and so on. CONCLUSION According to the obtained results, it can be mentioned that co-treatment of alpha-lipoic acid with radiotherapy ameliorates the radiation-induced toxicities in healthy cells/tissues.
Collapse
Affiliation(s)
- Sahar Sheikholeslami
- Department of Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shaghayegh Khodaverdian
- Department of Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoumeh Dorri-Giv
- Nuclear Medicine Research Center, Department of Nuclear Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyyed Mohammad Hosseini
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shokoufeh Souri
- Department of Medical Physics, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Hamed Zamani
- Department of Medical Physics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Leila Dastranj
- Department of Physics, Hakim Sabzevari Universuty, Sabzevar, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan. Iran.
| |
Collapse
|
24
|
Wang S, Li J, He Y, Ran Y, Lu B, Gao J, Shu C, Li J, Zhao Y, Zhang X, Hao Y. Protective effect of melatonin entrapped PLGA nanoparticles on radiation-induced lung injury through the miR-21/TGF-β1/Smad3 pathway. Int J Pharm 2021; 602:120584. [PMID: 33887395 DOI: 10.1016/j.ijpharm.2021.120584] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/18/2021] [Accepted: 04/04/2021] [Indexed: 01/04/2023]
Abstract
Radiation-induced lung injury (RILI) is a complication commonly found in victims suffering from nuclear accidents and patients treated with chest tumor radiotherapy, and drugs are limited for effective prevention and treatment. Melatonin (MET) has an anti-radiation effect, but its metabolic period in the body is short. In order to prolong the metabolism period of MET, we prepared MET entrapped poly (lactic-co-glycolic acid) nanoparticles (MET/PLGANPS) for the treatment of RILI. As a result, the release rate of MET/PLGANPS in vitro was lower than MET, with stable physical properties, and it caused no changes in histopathology and biochemical indicators. After 2 weeks and 16 weeks of irradiation with the dose of 15 Gy, MET and MET/PLGANPS could reduce the expression of caspase-3 proteins, inflammatory factors, TGF-β1 and Smad3 to alleviate radiation-induced lung injury. MET/PLGANPS showed better therapeutic effect on RILI than MET. In addition, we also found that high expression of miR-21 could increase the expression levels of TGF-β1, and inhibit the protective effect of MET/PLGANPS. In conclusion, MET/PLGANPS may alleviate RILI by inhibiting the miR-21/TGF-β1/Smad3 pathway, which would provide a new target for the treatment of radiation-induced lung injury.
Collapse
Affiliation(s)
- Shuang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Juan Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Yingjuan He
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Yonghong Ran
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Binghui Lu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Jining Gao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Chang Shu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Jie Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Yazhen Zhao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Xin Zhang
- Chongqing Normal University, No.37, Middle University Road, Shapingba District, Chongqing 401331, China
| | - Yuhui Hao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China.
| |
Collapse
|
25
|
Abstract
Reactive oxygen species (ROS) are ubiquitous metabolic products and important cellular signaling molecules that contribute to several biological functions. Pathophysiology arises when ROS are generated either in excess or in cell types or subcellular locations that normally do not produce ROS or when non-physiological types of ROS (e.g., superoxide instead of hydrogen peroxide) are formed. In the latter scenario, antioxidants were considered as the apparent remedy but, clinically, have consistently failed and even sometimes induced harm. The obvious reason for that is the non-selective ROS scavenging effects of antioxidants which interfere with both qualities of ROS, physiological and pathological. Therefore, it is essential to overcome this "antidote or neutralizer" strategy. We here review the most promising alternative approach by identifying the disease-relevant enzymatic sources of ROS, target these selectively, but leave physiological ROS signaling through other sources intact. Among all ROS sources, NADPH oxidases (NOX1-5 and DUOX1-2) stand out as their sole function is to produce ROS, whereas most other enzymatic sources only produce ROS as a by-product or upon biochemical uncoupling or damage. This qualifies NOXs as the main potential drug-target candidates in diseases associated with dysfunction in ROS signaling. As a reflection of this, the development of several NOX inhibitors has taken place. Recently, the WHO approved a new stem, "naxib," which refers to NADPH oxidase inhibitors, and thereby recognized NOX inhibitors as a new therapeutic class. This has been announced while clinical trials with the first-in-class compound, setanaxib (initially known as GKT137831) had been initiated. We also review the differences between the seven NOX family members in terms of structure and function in health and disease and then focus on the most advanced NOX inhibitors with an exclusive focus on clinically relevant validations and applications. Therapeutically relevant NADPH oxidase isoforms type 1, 2, 4, and 5 (NOX1, NOX2, NOX4, NOX5). Of note, NOX5 is not present in mice and rats and thus pre-clinically less studied. NOX2, formerly termed gp91phox, has been correlated with many, too many, diseases and is rather relevant as genetic deficiency in chronic granulomatous disease (CGD), treated by gene therapy. Overproduction of ROS through NOX1, NOX4, and NOX5 leads to the indicated diseases states including atherosclerosis (red), a condition where NOX4 is surprisingly protective.
Collapse
Affiliation(s)
- Mahmoud H Elbatreek
- Department of Pharmacology and Personalised Medicine, School of MeHNS, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | | | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, School of MeHNS, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
26
|
Helissey C, Cavallero S, Brossard C, Dusaud M, Chargari C, François S. Chronic Inflammation and Radiation-Induced Cystitis: Molecular Background and Therapeutic Perspectives. Cells 2020; 10:E21. [PMID: 33374374 PMCID: PMC7823735 DOI: 10.3390/cells10010021] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Radiation cystitis is a potential complication following the therapeutic irradiation of pelvic cancers. Its clinical management remains unclear, and few preclinical data are available on its underlying pathophysiology. The therapeutic strategy is difficult to establish because few prospective and randomized trials are available. In this review, we report on the clinical presentation and pathophysiology of radiation cystitis. Then we discuss potential therapeutic approaches, with a focus on the immunopathological processes underlying the onset of radiation cystitis, including the fibrotic process. Potential therapeutic avenues for therapeutic modulation will be highlighted, with a focus on the interaction between mesenchymal stromal cells and macrophages for the prevention and treatment of radiation cystitis.
Collapse
Affiliation(s)
- Carole Helissey
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, 91220 Brétigny-sur-Orge, France; (C.H.); (S.C.); (C.C.)
- Clinical Unit Research, HIA Bégin, 94160 Saint-Mandé, France
| | - Sophie Cavallero
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, 91220 Brétigny-sur-Orge, France; (C.H.); (S.C.); (C.C.)
| | - Clément Brossard
- Radiobiology of Medical Exposure Laboratory (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), 92260 Fontenay-aux-Roses, France;
| | - Marie Dusaud
- Department of Urology, HIA Bégin, 94160 Saint-Mand, France;
| | - Cyrus Chargari
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, 91220 Brétigny-sur-Orge, France; (C.H.); (S.C.); (C.C.)
- Gustave Roussy Comprehensive Cancer Center, Department of Radiation Oncology, 94805 Villejuif, France
- French Military Health Academy, Ecole du Val-de-Grâce (EVDG), 75005 Paris, France
| | - Sabine François
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, 91220 Brétigny-sur-Orge, France; (C.H.); (S.C.); (C.C.)
| |
Collapse
|
27
|
Amini P, Ashrafizadeh M, Motevaseli E, Najafi M, Shirazi A. Mitigation of radiation-induced hematopoietic system injury by melatonin. ENVIRONMENTAL TOXICOLOGY 2020; 35:815-821. [PMID: 32125094 DOI: 10.1002/tox.22917] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/05/2020] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Increased risks of exposure to accidental radiation events are a concern in today's world. Radiation terror, nuclear explosion, as well as accidental exposure to radioactive sources in some industries pose a threat to the life of exposed persons. Studies have been conducted using some low-toxic agents to mitigate radiation toxicity and increase survival probability for exposed people. In the current study, we aimed to show the mitigation of radiation-induced mortality and bone marrow toxicity using postirradiation treatment with melatonin. METHOD Mice whole bodies were exposed to 4 or 7 Gy radiation followed by treatment with melatonin after 24 hours. Survival of mice with or without melatonin, the levels of peripheral cells, transforming growth factor (TGF)-β and 8-hydroxy-2' -deoxyguanosine (8-OHdG) in the bone marrow, as well as the expression of NADPH oxidase (NOX)2 and NOX4 in bone marrow cells were evaluated. RESULTS Whole body irradiation led to mortality 30 days after irradiation. However, melatonin treatment reduced mortality. Irradiation also showed severe reduction of lymphocytes, platelets, and red blood cells. The expressions of NOX2 and NOX4, in addition to TGF-β level, were increased after exposure to radiation. Melatonin ameliorated the increased levels of these factors and improved the number of blood cells. CONCLUSIONS Melatonin showed ability to mitigate radiation-induced hematopoietic system toxicity and also increased survival rate. These results suggest that melatonin could be a potential mitigator for accidental radiation events.
Collapse
Affiliation(s)
- Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Veterinary Medicine Faculty, Tabriz University, Tabriz, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Shirazi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Wang B, Wang H, Zhang M, Ji R, Wei J, Xin Y, Jiang X. Radiation-induced myocardial fibrosis: Mechanisms underlying its pathogenesis and therapeutic strategies. J Cell Mol Med 2020; 24:7717-7729. [PMID: 32536032 PMCID: PMC7348163 DOI: 10.1111/jcmm.15479] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/18/2020] [Accepted: 05/24/2020] [Indexed: 12/24/2022] Open
Abstract
Radiation-induced myocardial fibrosis (RIMF) is a potentially lethal clinical complication of chest radiotherapy (RT) and a final stage of radiation-induced heart disease (RIHD). RIMF is characterized by decreased ventricular elasticity and distensibility, which can result in decreased ejection fraction, heart failure and even sudden cardiac death. Together, these conditions impair the long-term health of post-RT survivors and limit the dose and intensity of RT required to effectively kill tumour cells. Although the exact mechanisms involving in RIMF are unclear, increasing evidence indicates that the occurrence of RIMF is related to various cells, regulatory molecules and cytokines. However, accurately diagnosing and identifying patients who may progress to RIMF has been challenging. Despite the urgent need for an effective treatment, there is currently no medical therapy for RIMF approved for routine clinical application. In this review, we investigated the underlying pathophysiology involved in the initiation and progression of RIMF before outlining potential preventative and therapeutic strategies to counter this toxicity.
Collapse
Affiliation(s)
- Bin Wang
- Department of Radiation OncologyThe First Hospital of Jilin UniversityChangchunChina
- Jilin Provincial Key Laboratory of Radiation Oncology & TherapyThe First Hospital of Jilin UniversityChangchunChina
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunChina
| | - Huanhuan Wang
- Department of Radiation OncologyThe First Hospital of Jilin UniversityChangchunChina
- Jilin Provincial Key Laboratory of Radiation Oncology & TherapyThe First Hospital of Jilin UniversityChangchunChina
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunChina
| | - Mengmeng Zhang
- Phase I Clinical Research CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Rui Ji
- Department of BiologyValencia CollegeOrlandoFLUSA
| | - Jinlong Wei
- Department of Radiation OncologyThe First Hospital of Jilin UniversityChangchunChina
| | - Ying Xin
- Key Laboratory of PathobiologyMinistry of EducationJilin UniversityChangchunChina
| | - Xin Jiang
- Department of Radiation OncologyThe First Hospital of Jilin UniversityChangchunChina
- Jilin Provincial Key Laboratory of Radiation Oncology & TherapyThe First Hospital of Jilin UniversityChangchunChina
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunChina
| |
Collapse
|
29
|
Ping Z, Peng Y, Lang H, Xinyong C, Zhiyi Z, Xiaocheng W, Hong Z, Liang S. Oxidative Stress in Radiation-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3579143. [PMID: 32190171 PMCID: PMC7071808 DOI: 10.1155/2020/3579143] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/03/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
Abstract
There is a distinct increase in the risk of heart disease in people exposed to ionizing radiation (IR). Radiation-induced heart disease (RIHD) is one of the adverse side effects when people are exposed to ionizing radiation. IR may come from various forms, such as diagnostic imaging, radiotherapy for cancer treatment, nuclear disasters, and accidents. However, RIHD was mainly observed after radiotherapy for chest malignant tumors, especially left breast cancer. Radiation therapy (RT) has become one of the main ways to treat all kinds of cancer, which is used to reduce the recurrence of cancer and improve the survival rate of patients. The potential cause of radiation-induced cardiotoxicity is unclear, but it may be relevant to oxidative stress. Oxidative stress, an accumulation of reactive oxygen species (ROS), disrupts intracellular homeostasis through chemical modification and damages proteins, lipids, and DNA; therefore, it results in a series of related pathophysiological changes. The purpose of this review was to summarise the studies of oxidative stress in radiotherapy-induced cardiotoxicity and provide prevention and treatment methods to reduce cardiac damage.
Collapse
Affiliation(s)
- Zhang Ping
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Yang Peng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Hong Lang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Cai Xinyong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Zeng Zhiyi
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Wu Xiaocheng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Zeng Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Shao Liang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| |
Collapse
|
30
|
Ashrafizadeh M, Tavakol S, Ahmadi Z, Roomiani S, Mohammadinejad R, Samarghandian S. Therapeutic effects of kaempferol affecting autophagy and endoplasmic reticulum stress. Phytother Res 2019; 34:911-923. [PMID: 31829475 DOI: 10.1002/ptr.6577] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/01/2019] [Accepted: 11/14/2019] [Indexed: 01/06/2023]
Abstract
Regulated cell death (RCD) guarantees to preserve organismal homeostasis. Apoptosis and autophagy are two major arms of RCD, while endoplasmic reticulum (ER) as a crucial organelle involved in proteostasis, promotes cells toward autophagy and apoptosis. Alteration in ER stress and autophagy machinery is responsible for a great number of diseases. Therefore, targeting those pathways appears to be beneficial in the treatment of relevant diseases. Meantime, among the traditional herb medicine, kaempferol as a flavonoid seems to be promising to modulate ER stress and autophagy and exhibits protective effects on malfunctioning cells. There are some reports indicating the capability of kaempferol in affecting autophagy and ER stress. In brief, kaempferol modulates autophagy in noncancerous cells to protect cells against malfunction, while it induces cell mortality derived from autophagy through the elevation of p-AMP-activated protein kinase, light chain-3-II, autophagy-related geness, and Beclin-1 in cancer cells. Noteworthy, kaempferol enhances cell survival through C/EBP homologous protein (CHOP) suppression and GRP78 increment in noncancerous cells, while it enhances cell mortality through the induction of unfolding protein response and CHOP increment in cancer cells. In this review, we discuss how kaempferol modulates autophagy and ER stress in noncancer and cancer cells to expand our knowledge of new pharmacological compounds for the treatment of associated diseases.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Ahmadi
- Department of Basic Science, Shoushtar Branch, Islamic Azad University, Shoushtar, Iran
| | - Sahar Roomiani
- Department of Basic Science, Shoushtar Branch, Islamic Azad University, Shoushtar, Iran
| | - Reza Mohammadinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
31
|
Zou B, Schuster JP, Niu K, Huang Q, Rühle A, Huber PE. Radiotherapy-induced heart disease: a review of the literature. PRECISION CLINICAL MEDICINE 2019; 2:270-282. [PMID: 35693876 PMCID: PMC8985808 DOI: 10.1093/pcmedi/pbz025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 11/20/2022] Open
Abstract
Radiotherapy as one of the four pillars of cancer therapy plays a critical role in the multimodal treatment of thoracic cancers. Due to significant improvements in overall cancer survival, radiotherapy-induced heart disease (RIHD) has become an increasingly recognized adverse reaction which contributes to major radiation-associated toxicities including non-malignant death. This is especially relevant for patients suffering from diseases with excellent prognosis such as breast cancer or Hodgkin's lymphoma, since RIHD may occur decades after radiotherapy. Preclinical studies have enriched our knowledge of many potential mechanisms by which thoracic radiotherapy induces heart injury. Epidemiological findings in humans reveal that irradiation might increase the risk of cardiac disease at even lower doses than previously assumed. Recent preclinical studies have identified non-invasive methods for evaluation of RIHD. Furthermore, potential options preventing or at least attenuating RIHD have been developed. Ongoing research may enrich our limited knowledge about biological mechanisms of RIHD, identify non-invasive early detection biomarkers and investigate potential treatment options that might attenuate or prevent these unwanted side effects. Here, we present a comprehensive review about the published literature regarding clinical manifestation and pathological alterations in RIHD. Biological mechanisms and treatment options are outlined, and challenges in RIHD treatment are summarized.
Collapse
Affiliation(s)
- Bingwen Zou
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Julius Philipp Schuster
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Kerun Niu
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Qianyi Huang
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
- Heidelberg Institute for Radiation Oncology (HIRO) and National Center for Radiation Oncology (NCRO), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Peter Ernst Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
- Heidelberg Institute for Radiation Oncology (HIRO) and National Center for Radiation Oncology (NCRO), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| |
Collapse
|
32
|
Radiation-Induced Normal Tissue Damage: Oxidative Stress and Epigenetic Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3010342. [PMID: 31781332 PMCID: PMC6875293 DOI: 10.1155/2019/3010342] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 01/23/2023]
Abstract
Radiotherapy (RT) is currently one of the leading treatments for various cancers; however, it may cause damage to healthy tissue, with both short-term and long-term side effects. Severe radiation-induced normal tissue damage (RINTD) frequently has a significant influence on the progress of RT and the survival and prognosis of patients. The redox system has been shown to play an important role in the early and late effects of RINTD. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are the main sources of RINTD. The free radicals produced by irradiation can upregulate several enzymes including nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase), lipoxygenases (LOXs), nitric oxide synthase (NOS), and cyclooxygenases (COXs). These enzymes are expressed in distinct ways in various cells, tissues, and organs and participate in the RINTD process through different regulatory mechanisms. In recent years, several studies have demonstrated that epigenetic modulators play an important role in the RINTD process. Epigenetic modifications primarily contain noncoding RNA regulation, histone modifications, and DNA methylation. In this article, we will review the role of oxidative stress and epigenetic mechanisms in radiation damage, and explore possible prophylactic and therapeutic strategies for RINTD.
Collapse
|
33
|
Farhood B, Aliasgharzadeh A, Amini P, Rezaeyan A, Tavassoli A, Motevaseli E, Shabeeb D, Musa AE, Najafi M. Mitigation of Radiation-Induced Lung Pneumonitis and Fibrosis Using Metformin and Melatonin: A Histopathological Study. ACTA ACUST UNITED AC 2019; 55:medicina55080417. [PMID: 31366142 PMCID: PMC6722577 DOI: 10.3390/medicina55080417] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/17/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023]
Abstract
Background and objectives: Pneumonitis and fibrosis are the most common consequences of lung exposure to a high dose of ionizing radiation during an accidental radiological or nuclear event, and may lead to death, after some months to years. So far, some anti-inflammatory and antioxidant agents have been used for mitigation of lung injury. In the present study, we aimed to detect possible mitigatory effects of melatonin and metformin on radiation-induced pneumonitis and lung fibrosis. Materials and methods: 40 male mice were divided into 4 groups (10 mice in each). For control group, mice did not receive radiation or drugs. In group 2, mice were irradiated to chest area with 18 Gy gamma rays. In groups 3 and 4, mice were first irradiated similar to group 2. After 24 h, treatment with melatonin as well as metformin began. Mice were sacrificed after 100 days for determination of mitigation of lung pneumonitis and fibrosis by melatonin or metformin. Results: Results showed that both melatonin and metformin are able to mitigate pneumonitis and fibrosis markers such as infiltration of inflammatory cells, edema, vascular and alveolar thickening, as well as collagen deposition. Conclusion: Melatonin and metformin may have some interesting properties for mitigation of radiation pneumonitis and fibrosis after an accidental radiation event.
Collapse
Affiliation(s)
- Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan 8715988141, Iran
| | - Akbar Aliasgharzadeh
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan 8715988141, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - Abolhasan Rezaeyan
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Alireza Tavassoli
- Department of Pathology, Fasa University of Medical Sciences, Fasa 8668874616, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan 62010, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences (International Campus), Tehran 1416753955, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran.
| |
Collapse
|