1
|
Portillo EGD, Olivares-Hernández A, Gudino LC, Félix LC, Hernández LB, Domínguez LP, Jiménez DL, Sarmiento RG, Morillo EDB, Sánchez EF, Miramontes-Gonzáleze JP. Evaluation of the effect of metformin as a radiosensitiser in solid tumours: A systematic review. Clin Transl Radiat Oncol 2025; 52:100930. [PMID: 40028423 PMCID: PMC11871473 DOI: 10.1016/j.ctro.2025.100930] [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: 11/08/2024] [Revised: 01/13/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Background Metformin is an antidiabetic drug that has shown its benefit in increasing the effect of radiotherapy in the treatment of solid tumors in preclinical studies. The objective of this systematic review is to study the effect of metformin as a radiosensitizer in studies carried out in clinical practice. Methods Systematic review carried out according to PRISMA criteria of clinical trials, systematic reviews and observational studies focused on the influence of metformin as a radiosensitizer in solid tumors. The studies were published between the years 2010 and 2022. The results of the studies have been analyzed in terms of survival (OS, PFS, DFS, DMFS) and response (ORR) between patients treated with metformin and without it. Results A total of 16 studies have been found in the literature (the most frequent tumor was prostate cancer, 5 studies). External radiotherapy was administered in all the studies and in two of them to greater brachytherapy. The use of metformin with radiotherapy showed a consistent benefit in terms of survival and response in tumors of prostate, hepatic and gynecological origin. The benefit in the rest of the tumors analyzed (lung, rectal, and head and neck cancer) is doubtful, and the results are contradictory. The greatest benefits were observed in prostate tumors both in OS and SLE. Conclusions The use of metformin in combination with radiotherapy in solid tumors is one of the most promising treatments under development in oncology. The benefit observed in real-life studies makes it necessary to develop clinical trials that specifically evaluate its use in clinical practice in the future.
Collapse
Affiliation(s)
| | - Alejandro Olivares-Hernández
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
| | - Luis Corral Gudino
- Internal Medicine Unit, Río Hortega Hospital, Valladolid, Spain
- Medicine Department, University of Valladolid Medical School, Spain
| | - Laura Corvo Félix
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
| | - Lorena Bellido Hernández
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
- Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Luis Posado Domínguez
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
| | | | - Rogelio González Sarmiento
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Internal Medicine Unit, Río Hortega Hospital, Valladolid, Spain
| | - Edel del Barco Morillo
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
- Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Emilio Fonseca Sánchez
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
- Internal Medicine Unit, Río Hortega Hospital, Valladolid, Spain
| | - José Pablo Miramontes-Gonzáleze
- Internal Medicine Unit, Río Hortega Hospital, Valladolid, Spain
- Medicine Department, University of Valladolid Medical School, Spain
| |
Collapse
|
2
|
Khiabani NA, Doustvandi MA, Story D, Nobari SA, Hajizadeh M, Petersen R, Dunbar G, Rossignol J. Glioblastoma therapy: State of the field and future prospects. Life Sci 2024; 359:123227. [PMID: 39537100 DOI: 10.1016/j.lfs.2024.123227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GB) is a cancerous brain tumor that originates from glial cells and leads to thousands of deaths each year and a five-year survival of only 6.8 %. Treatments for GB include surgery, chemotherapy, radiation, and immunotherapy. GB is an incurable fatal disease, necessitating the development of innovative strategies to find a developing effective therapy. Genetic therapies may be crucial in treating GB by identifying the mutations and amplifications of multiple genes, which drive its proliferation and spread. Use of small interfering RNAs (siRNAs) provides a novel technology used to suppress the genes associated with disease, which forms a basis for targeted therapy in GB and its stem cell population, which are recognized for their ability to develop resistance to chemotherapy and tumorigenic capabilities. This review examines the use of siRNAs in GB, emphasizing their effectiveness in suppressing key oncogenes and signaling pathways associated with tumor development, invasion, stemness, and resistance to standard treatments. siRNA-based gene silencing is a promising approach for developing targeted therapeutics against GB and associated stem cell populations, potentially enhancing patient outcomes and survival rates in this devastating disease.
Collapse
Affiliation(s)
- Nadia Allahyarzadeh Khiabani
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | | | - Darren Story
- Department of Psychology, Saginaw Valley State University, University Center, MI 48710, USA
| | | | | | - Robert Petersen
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Gary Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA.
| |
Collapse
|
3
|
Chi MS, Hsieh PH, Huang SH, Hsu HC, Chi KH. Chronic radiation proctitis refractory to steroid enema was successfully treated by metformin and sodium butyrate: a case report. J Med Case Rep 2024; 18:239. [PMID: 38725071 PMCID: PMC11083804 DOI: 10.1186/s13256-024-04551-x] [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: 02/15/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Radiation proctitis (RP) is a significant complication of pelvic radiation. Effective treatments for chronic RP are currently lacking. We report a case where chronic RP was successfully managed by metformin and butyrate (M-B) enema and suppository therapy. CASE PRESENTATION A 70-year-old Asian male was diagnosed with prostate cancer of bilateral lobes, underwent definitive radiotherapy to the prostate of 76 Gy in 38 fractions and six months of androgen deprivation therapy. Despite a stable PSA nadir of 0.2 ng/mL for 10 months post-radiotherapy, he developed intermittent rectal bleeding, and was diagnosed as chronic RP. Symptoms persisted despite two months of oral mesalamine, mesalamine enema and hydrocortisone enema treatment. Transition to daily 2% metformin and butyrate (M-B) enema for one week led to significant improvement, followed by maintenance therapy with daily 2.0% M-B suppository for three weeks, resulting in continued reduction of rectal bleeding. Endoscopic examination and biopsy demonstrated a good therapeutic effect. CONCLUSIONS M-B enema and suppository may be an effective treatment for chronic RP.
Collapse
Affiliation(s)
- Mau-Shin Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Ping-Hsun Hsieh
- Division of Gastroenterology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Shu-Han Huang
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ho-Chi Hsu
- Department of General Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.
| |
Collapse
|
4
|
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
|
5
|
Khodamoradi E, Rahmani N, Rashidi K, Najafi M, Shahsavari S, Mohammadi M. Exploring the Potential of Metformin in Mitigating Radiation-induced Gastrointestinal and Hematopoietic System Injury in Rats After Whole-body X-ray Radiation: An Experimental Study. Curr Radiopharm 2024; 17:200-208. [PMID: 38231059 DOI: 10.2174/0118744710261673231115062547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/09/2023] [Accepted: 10/02/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND The modern world faces a growing concern about the possibility of accidental radiation events. The Hematopoietic system is particularly vulnerable to radiationinduced apoptosis, which can lead to death. Metformin, a drug used to treat diabetes, has been shown to protect normal cells and tissues from the toxic effects of radiation. This study aimed to evaluate the effectiveness of metformin in mitigating radiation injury to the gastrointestinal and hematological systems of rats. MATERIALS AND METHODS The study involved 73 male rats. After total body irradiation with 7.5 Gy of X-rays, rats were treated with metformin. Seven days later, the rats were sacrificed and blood samples were taken for evaluation. RESULTS The study found that metformin was not effective in mitigating radiation injury. The histopathological assessment showed no significant changes in goblet cell injury, villi shortening, inflammation, or mucous layer thickness. In terms of biochemical evaluation, metformin did not significantly affect oxidative stress markers, but irradiation increased the mean MDA level in the radiation group. The complete blood count revealed a significant decrease in WBC and platelet, counts in the radiation group compared to the control group, but no significant difference was found between the radiation and radiation + metformin groups. CONCLUSION In conclusion, metformin may not be a good option for reducing radiation toxicity after accidental exposure. Despite treatment, there was no improvement in platelet, white blood cell, and lymphocyte counts, nor was there any decrease in oxidative stress. Further research is needed to explore other potential treatments for radiation injury.
Collapse
Affiliation(s)
- Ehsan Khodamoradi
- Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nafiseh Rahmani
- Student Research Committee, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Khodabakhsh Rashidi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Najafi
- Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soodeh Shahsavari
- Department of Health Information Technology, Faculty of Allied Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Mohammadi
- Student Research Committee, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
6
|
Mohammadgholi M, Hosseinimehr SJ. Crosstalk between Oxidative Stress and Inflammation Induced by Ionizing Radiation in Healthy and Cancerous Cells. Curr Med Chem 2024; 31:2751-2769. [PMID: 37026495 DOI: 10.2174/0929867330666230407104208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/18/2023] [Accepted: 02/24/2023] [Indexed: 04/08/2023]
Abstract
Radiotherapy (RT) is a unique modality in cancer treatment with no replacement in many cases and uses a tumoricidal dose of various ionizing radiation (IR) types to kill cancer cells. It causes oxidative stress through reactive oxygen species (ROS) production or the destruction of antioxidant systems. On the other hand, RT stimulates the immune system both directly and indirectly by releasing danger signals from stress-exposed and dying cells. Oxidative stress and inflammation are two reciprocal and closely related mechanisms, one induced and involved by the other. ROS regulates the intracellular signal transduction pathways, which participate in the activation and expression of pro-inflammatory genes. Reciprocally, inflammatory cells release ROS and immune system mediators during the inflammation process, which drive the induction of oxidative stress. Oxidative stress or inflammation-induced damages can result in cell death (CD) or survival mechanisms that may be destructive for normal cells or beneficial for cancerous cells. The present study has focused on the radioprotection of those agents with binary effects of antioxidant and anti-inflammatory mechanisms IR-induced CD.
Collapse
Affiliation(s)
- Mohsen Mohammadgholi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
7
|
Gupta J, Jalil AT, Abd Alzahraa ZH, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Najafi M. The Metformin Immunoregulatory Actions in Tumor Suppression and Normal Tissues Protection. Curr Med Chem 2024; 31:5370-5396. [PMID: 37403391 DOI: 10.2174/0929867331666230703143907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023]
Abstract
The immune system is the key player in a wide range of responses in normal tissues and tumors to anticancer therapy. Inflammatory and fibrotic responses in normal tissues are the main limitations of chemotherapy, radiotherapy, and also some newer anticancer drugs such as immune checkpoint inhibitors (ICIs). Immune system responses within solid tumors including anti-tumor and tumor-promoting responses can suppress or help tumor growth. Thus, modulation of immune cells and their secretions such as cytokines, growth factors and epigenetic modulators, pro-apoptosis molecules, and some other molecules can be suggested to alleviate side effects in normal tissues and drug-resistance mechanisms in the tumor. Metformin as an anti-diabetes drug has shown intriguing properties such as anti-inflammation, anti-fibrosis, and anticancer effects. Some investigations have uncovered that metformin can ameliorate radiation/chemotherapy toxicity in normal cells and tissues through the modulation of several targets in cells and tissues. These effects of metformin may ameliorate severe inflammatory responses and fibrosis after exposure to ionizing radiation or following treatment with highly toxic chemotherapy drugs. Metformin can suppress the activity of immunosuppressive cells in the tumor through the phosphorylation of AMP-activated protein kinase (AMPK). In addition, metformin may stimulate antigen presentation and maturation of anticancer immune cells, which lead to the induction of anticancer immunity in the tumor. This review aims to explain the detailed mechanisms of normal tissue sparing and tumor suppression during cancer therapy using adjuvant metformin with an emphasis on immune system responses.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 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
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, 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
| | | | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
8
|
Qin L, Wu J. Targeting anticancer immunity in oral cancer: Drugs, products, and nanoparticles. ENVIRONMENTAL RESEARCH 2023; 239:116751. [PMID: 37507044 DOI: 10.1016/j.envres.2023.116751] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
Oral cavity carcinomas are the most frequent malignancies among head and neck malignancies. Oral tumors include not only oral cancer cells with different potency and stemness but also consist of diverse cells, containing anticancer immune cells, stromal and also immunosuppressive cells that influence the immune system reactions. The infiltrated T and natural killer (NK) cells are the substantial tumor-suppressive immune compartments in the tumor. The infiltration of these cells has substantial impacts on the response of tumors to immunotherapy, chemotherapy, and radiotherapy. Nevertheless, cancer cells, stromal cells, and some other compartments like regulatory T cells (Tregs), macrophages, and myeloid-derived suppressor cells (MDSCs) can repress the immune responses against malignant cells. Boosting anticancer immunity by inducing the immune system or repressing the tumor-promoting cells is one of the intriguing approaches for the eradication of malignant cells such as oral cancers. This review aims to concentrate on the secretions and interactions in the oral tumor immune microenvironment. We review targeting tumor stroma, immune system and immunosuppressive interactions in oral tumors. This review will also focus on therapeutic targets and therapeutic agents such as nanoparticles and products with anti-tumor potency that can boost anticancer immunity in oral tumors. We also explain possible future perspectives including delivery of various cells, natural products and drugs by nanoparticles for boosting anticancer immunity in oral tumors.
Collapse
Affiliation(s)
- Liling Qin
- Gezhouba Central Hospital of the Third Clinical Medical College of Three Gorges University, Yichang, Hubei, 443002, China
| | - Jianan Wu
- Experimental and Practical Teaching Center, Hubei College of Chinese Medicine, Jingzhou, Hubei, 434000, China.
| |
Collapse
|
9
|
Cheki M, Mostafaei S, Hanafi MG, Farasat M, Talaiezadeh A, Ghasemi MS, Modava M, Abdollahi H. Radioproteomics modeling of metformin-enhanced radiosensitivity: an animal study. Jpn J Radiol 2023; 41:1265-1274. [PMID: 37204669 DOI: 10.1007/s11604-023-01445-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
PURPOSE Metformin is considered as radiation modulator in both tumors and healthy tissues. Radiomics has the potential to decode biological mechanisms of radiotherapy response. The aim of this study was to apply radiomics analysis in metformin-induced radiosensitivity and finding radioproteomics associations of computed tomography (CT) imaging features and proteins involved in metformin radiosensitivity signaling pathways. MATERIALS AND METHODS A total of 32 female BALB/c mice were used in this study and were subjected to injection of breast cancer cells. When tumors reached a mean volume of 150 mm3, mice were randomly divided into the four groups including Control, Metformin, Radiation, and Radiation + Metformin. Western blot analysis was performed after treatment to measure expression of proteins including AMPK-alpha, phospho-AMPK-alpha (Thr172), mTOR, phospho-mTOR (Ser2448), phospho-4EBP1 (Thr37/46), phospho-ACC (Ser79), and β-actin. CT imaging was performed before treatment and at the end of treatment in all groups. Radiomics features extracted from segmented tumors were selected using Elastic-net regression and were assessed in terms of correlation with expression of the proteins. RESULTS It was observed that proteins including phospho-mTOR, phospho-4EBP1, and mTOR had positive correlations with changes in tumor volumes in days 28, 24, 20, 16, and 12, while tumor volume changes at these days had negative correlations with AMPK-alpha, phospho-AMPK-alpha, and phospho-ACC proteins. Furthermore, median feature had a positive correlation with AMPK-alpha, phospho-ACC, and phospho-AMPK-alpha proteins. Also, Cluster shade feature had positive correlations with mTOR and p-mTOR. On the other hand, LGLZE feature had negative correlations with AMPK-alpha and phospho-AMPK-alpha. CONCLUSION Radiomics features can decode proteins that involved in response to metformin and radiation, although further studies are warranted to investigate the optimal way to integrate radiomics into biological experiments.
Collapse
Affiliation(s)
- Mohsen Cheki
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Medical Imaging and Radiation Sciences, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Shayan Mostafaei
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Maryam Farasat
- Department of Radiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | | | - Mohammad Modava
- Department of Electrical Engineering, Faculty of Engineering, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Hamid Abdollahi
- Department of Radiology Technology, Faculty of Allied Medical Sciences, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
10
|
Yu Z, Xu C, Song B, Zhang S, Chen C, Li C, Zhang S. Tissue fibrosis induced by radiotherapy: current understanding of the molecular mechanisms, diagnosis and therapeutic advances. J Transl Med 2023; 21:708. [PMID: 37814303 PMCID: PMC10563272 DOI: 10.1186/s12967-023-04554-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/22/2023] [Indexed: 10/11/2023] Open
Abstract
Cancer remains the leading cause of death around the world. In cancer treatment, over 50% of cancer patients receive radiotherapy alone or in multimodal combinations with other therapies. One of the adverse consequences after radiation exposure is the occurrence of radiation-induced tissue fibrosis (RIF), which is characterized by the abnormal activation of myofibroblasts and the excessive accumulation of extracellular matrix. This phenotype can manifest in multiple organs, such as lung, skin, liver and kidney. In-depth studies on the mechanisms of radiation-induced fibrosis have shown that a variety of extracellular signals such as immune cells and abnormal release of cytokines, and intracellular signals such as cGAS/STING, oxidative stress response, metabolic reprogramming and proteasome pathway activation are involved in the activation of myofibroblasts. Tissue fibrosis is extremely harmful to patients' health and requires early diagnosis. In addition to traditional serum markers, histologic and imaging tests, the diagnostic potential of nuclear medicine techniques is emerging. Anti-inflammatory and antioxidant therapies are the traditional treatments for radiation-induced fibrosis. Recently, some promising therapeutic strategies have emerged, such as stem cell therapy and targeted therapies. However, incomplete knowledge of the mechanisms hinders the treatment of this disease. Here, we also highlight the potential mechanistic, diagnostic and therapeutic directions of radiation-induced fibrosis.
Collapse
Affiliation(s)
- Zuxiang Yu
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chaoyu Xu
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Bin Song
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, 621099, China
| | - Shihao Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chong Chen
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221200, China
| | - Changlong Li
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- Department of Molecular Biology and Biochemistry, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China.
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, 621099, China.
| |
Collapse
|
11
|
Karmanova EE, Chernikov AV, Popova NR, Sharapov MG, Ivanov VE, Bruskov VI. Metformin mitigates radiation toxicity exerting antioxidant and genoprotective properties. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2449-2460. [PMID: 36961549 PMCID: PMC10036983 DOI: 10.1007/s00210-023-02466-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/14/2023] [Indexed: 03/25/2023]
Abstract
The antidiabetic drug metformin (MF) exhibits redox-modulating effects in various pathologies associated with oxidative stress and mitigates ionizing radiation-induced toxicity, but the underlying mechanisms remain to be elucidated. Thus, we studied some radiomitigatory effects of MF and explored the possible mechanisms behind them. Highly sensitive luminescence methods and non-competitive enzyme-linked immunosorbent assay (ELISA) were used in in vitro studies, and in vivo the damage to bone marrow cells and its repair were assessed by the micronucleus test. In a solution, MF at concentrations exceeding 0.1 µM effectively intercepts •OH upon X-ray-irradiation, but does not react directly with H2O2. MF accelerates the decomposition of H2O2 catalyzed by copper ions. MF does not affect the radiation-induced formation of H2O2 in the solution of bovine gamma-globulin (BGG), but has a modulating effect on the generation of H2O2 in the solution of bovine serum albumin (BSA). MF at 0.05-1 mM decreases the radiation-induced formation of 8-oxoguanine in a DNA solution depending on the concentration of MF with a maximum at 0.25 mM. MF at doses of 3 mg/kg body weight (bw) and 30 mg/kg bw administered to mice after irradiation, but not before irradiation, reduces the frequency of micronucleus formation in polychromatophilic erythrocytes of mouse red bone marrow. Our work has shown that the radiomitigatory properties of MF are mediated by antioxidant mechanisms of action, possibly including its ability to chelate polyvalent metal ions.
Collapse
Affiliation(s)
- Ekaterina E Karmanova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 3 Institutskaya St., Pushchino, Moscow Region, 142290, Russia
- Institute of Cell Biophysics, Pushchino Scientific Center for Biological Research, Federal Research Center of the Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Anatoly V Chernikov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 3 Institutskaya St., Pushchino, Moscow Region, 142290, Russia.
| | - Nelli R Popova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 3 Institutskaya St., Pushchino, Moscow Region, 142290, Russia
| | - Mars G Sharapov
- Institute of Cell Biophysics, Pushchino Scientific Center for Biological Research, Federal Research Center of the Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Vladimir E Ivanov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 3 Institutskaya St., Pushchino, Moscow Region, 142290, Russia
| | - Vadim I Bruskov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 3 Institutskaya St., Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
12
|
Malaekeh-Nikouei A, Shokri-Naei S, Karbasforoushan S, Bahari H, Baradaran Rahimi V, Heidari R, Askari VR. Metformin beyond an anti-diabetic agent: A comprehensive and mechanistic review on its effects against natural and chemical toxins. Biomed Pharmacother 2023; 165:115263. [PMID: 37541178 DOI: 10.1016/j.biopha.2023.115263] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023] Open
Abstract
In addition to the anti-diabetic effect of metformin, a growing number of studies have shown that metformin has some exciting properties, such as anti-oxidative capabilities, anticancer, genomic stability, anti-inflammation, and anti-fibrosis, which have potent, that can treat other disorders other than diabetes mellitus. We aimed to describe and review the protective and antidotal efficacy of metformin against biologicals, chemicals, natural, medications, pesticides, and radiation-induced toxicities. A comprehensive search has been performed from Scopus, Web of Science, PubMed, and Google Scholar databases from inception to March 8, 2023. All in vitro, in vivo, and clinical studies were considered. Many studies suggest that metformin affects diseases other than diabetes. It is a radioprotective and chemoprotective drug that also affects viral and bacterial diseases. It can be used against inflammation-related and apoptosis-related abnormalities and against toxins to lower their effects. Besides lowering blood sugar, metformin can attenuate the effects of toxins on body weight, inflammation, apoptosis, necrosis, caspase-3 activation, cell viability and survival rate, reactive oxygen species (ROS), NF-κB, TNF-α, many interleukins, lipid profile, and many enzymes activity such as catalase and superoxide dismutase. It also can reduce the histopathological damages induced by many toxins on the kidneys, liver, and colon. However, clinical trials and human studies are needed before using metformin as a therapeutic agent against other diseases.
Collapse
Affiliation(s)
- Amirhossein Malaekeh-Nikouei
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Shokri-Naei
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sobhan Karbasforoushan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Bahari
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Heidari
- Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran; Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
| | - Vahid Reza Askari
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Buckley CE, O’Brien RM, Nugent TS, Donlon NE, O’Connell F, Reynolds JV, Hafeez A, O’Ríordáin DS, Hannon RA, Neary P, Kalbassi R, Mehigan BJ, McCormick PH, Dunne C, Kelly ME, Larkin JO, O’Sullivan J, Lynam-Lennon N. Metformin is a metabolic modulator and radiosensitiser in rectal cancer. Front Oncol 2023; 13:1216911. [PMID: 37601689 PMCID: PMC10435980 DOI: 10.3389/fonc.2023.1216911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Resistance to neoadjuvant chemoradiation therapy, is a major challenge in the management of rectal cancer. Increasing evidence supports a role for altered energy metabolism in the resistance of tumours to anti-cancer therapy, suggesting that targeting tumour metabolism may have potential as a novel therapeutic strategy to boost treatment response. In this study, the impact of metformin on the radiosensitivity of colorectal cancer cells, and the potential mechanisms of action of metformin-mediated radiosensitisation were investigated. Metformin treatment was demonstrated to significantly radiosensitise both radiosensitive and radioresistant colorectal cancer cells in vitro. Transcriptomic and functional analysis demonstrated metformin-mediated alterations to energy metabolism, mitochondrial function, cell cycle distribution and progression, cell death and antioxidant levels in colorectal cancer cells. Using ex vivo models, metformin treatment significantly inhibited oxidative phosphorylation and glycolysis in treatment naïve rectal cancer biopsies, without affecting the real-time metabolic profile of non-cancer rectal tissue. Importantly, metformin treatment differentially altered the protein secretome of rectal cancer tissue when compared to non-cancer rectal tissue. Together these data highlight the potential utility of metformin as an anti-metabolic radiosensitiser in rectal cancer.
Collapse
Affiliation(s)
- Croí E. Buckley
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
| | - Rebecca M. O’Brien
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
| | - Timothy S. Nugent
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
- Department of Surgery, Beacon Hospital, Dublin, Ireland
| | - Noel E. Donlon
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
- Department of Surgery, Beacon Hospital, Dublin, Ireland
- Gastrointestinal Medicine and Surgery (GEMS) Directorate, St. James’s Hospital, Dublin, Ireland
| | - Fiona O’Connell
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
| | - John V. Reynolds
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
| | - Adnan Hafeez
- Department of Surgery, Beacon Hospital, Dublin, Ireland
| | | | | | - Paul Neary
- Department of Surgery, Beacon Hospital, Dublin, Ireland
| | - Reza Kalbassi
- Department of Surgery, Beacon Hospital, Dublin, Ireland
| | - Brian J. Mehigan
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
- Gastrointestinal Medicine and Surgery (GEMS) Directorate, St. James’s Hospital, Dublin, Ireland
| | - Paul H. McCormick
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
- Gastrointestinal Medicine and Surgery (GEMS) Directorate, St. James’s Hospital, Dublin, Ireland
| | - Cara Dunne
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
- Gastrointestinal Medicine and Surgery (GEMS) Directorate, St. James’s Hospital, Dublin, Ireland
| | - Michael E. Kelly
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
- Gastrointestinal Medicine and Surgery (GEMS) Directorate, St. James’s Hospital, Dublin, Ireland
| | - John O. Larkin
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
- Gastrointestinal Medicine and Surgery (GEMS) Directorate, St. James’s Hospital, Dublin, Ireland
| | - Jacintha O’Sullivan
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
14
|
Men J, Zhang L, Peng R, Li Y, Li M, Wang H, Zhao L, Zhang J, Wang H, Xu X, Dong J, Wang J, Yao B, Guo J. Metformin Ameliorates 2.856 GHz Microwave- Radiation-Induced Reproductive Impairments in Male Rats via Inhibition of Oxidative Stress and Apoptosis. Int J Mol Sci 2023; 24:12250. [PMID: 37569626 PMCID: PMC10418945 DOI: 10.3390/ijms241512250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
The reproductive system has been increasingly implicated as a sensitive target of microwave radiation. Oxidative stress plays a critical role in microwave radiation -induced reproductive damage, though precise mechanisms are obscure. Metformin, a widely used antidiabetic drug, has emerged as an efficient antioxidant against a variety of oxidative injuries. In the present study, we hypothesized that metformin can function as an antioxidant and protect the reproductive system from microwave radiation. To test this hypothesis, rats were exposed to 2.856 GHz microwave radiation for 6 weeks to simulate real-life exposure to high-frequency microwave radiation. Our results showed that exposure to 2.856 GHz microwave radiation elicited serum hormone disorder, decreased sperm motility, and depleted sperm energy, and it induced abnormalities of testicular structure as well as mitochondrial impairment. Metformin was found to effectively protect the reproductive system against structural and functional impairments caused by microwave radiation. In particular, metformin can ameliorate microwave-radiation-induced oxidative injury and mitigate apoptosis in the testis, as determined by glutathione/-oxidized glutathione (GSH/GSSG), lipid peroxidation, and protein expression of heme oxygenase-1 (HO-1). These findings demonstrated that exposure to 2.856 GHz microwave radiation induces obvious structural and functional impairments of the male reproductive system, and suggested that metformin can function as a promising antioxidant to inhibit microwave-radiation-induced harmful effects by inhibiting oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Junqi Men
- PLA Center for Disease Control and Prevention, Beijing 100071, China; (J.M.); (L.Z.)
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
- School of Public Health, China Medical University, Shenyang 110122, China
| | - Li Zhang
- PLA Center for Disease Control and Prevention, Beijing 100071, China; (J.M.); (L.Z.)
| | - Ruiyun Peng
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Yanyang Li
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Meng Li
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Hui Wang
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Li Zhao
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Jing Zhang
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Haoyu Wang
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Xinping Xu
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Ji Dong
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Juan Wang
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Binwei Yao
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing 100850, China; (R.P.); (Y.L.); (M.L.); (H.W.); (L.Z.); (J.Z.); (H.W.); (X.X.); (J.D.); (J.W.)
| | - Jiabin Guo
- PLA Center for Disease Control and Prevention, Beijing 100071, China; (J.M.); (L.Z.)
- School of Public Health, China Medical University, Shenyang 110122, China
| |
Collapse
|
15
|
Sekar P, Hsiao G, Hsu SH, Huang DY, Lin WW, Chan CM. Metformin inhibits methylglyoxal-induced retinal pigment epithelial cell death and retinopathy via AMPK-dependent mechanisms: Reversing mitochondrial dysfunction and upregulating glyoxalase 1. Redox Biol 2023; 64:102786. [PMID: 37348156 PMCID: PMC10363482 DOI: 10.1016/j.redox.2023.102786] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 06/24/2023] Open
Abstract
Diabetic retinopathy (DR) is a major cause of blindness in adult, and the accumulation of advanced glycation end products (AGEs) is a major pathologic event in DR. Methylglyoxal (MGO), a highly reactive dicarbonyl compound, is a precursor of AGEs. Although the therapeutic potential of metformin for retinopathy disorders has recently been elucidated, possibly through AMPK activation, it remains unknown how metformin directly affects the MGO-induced stress response in retinal pigment epithelial cells. Therefore, in this study, we compared the effects of metformin and the AMPK activator A769662 on MGO-induced DR in mice, as well as evaluated cytotoxicity, mitochondrial dynamic changes and dysfunction in ARPE-19 cells. We found MGO can induce mitochondrial ROS production and mitochondrial membrane potential loss, but reduce cytosolic ROS level in ARPE-19 cells. Although these effects of MGO can be reversed by both metformin and A769662, we demonstrated that reduction of mitochondrial ROS production rather than restoration of cytosolic ROS level contributes to cell protective effects of metformin and A769662. Moreover, MGO inhibits AMPK activity, reduces LC3II accumulation, and suppresses protein and gene expressions of MFN1, PGC-1α and TFAM, leading to mitochondrial fission, inhibition of mitochondrial biogenesis and autophagy. In contrast, these events of MGO were reversed by metformin in an AMPK-dependent manner as evidenced by the effects of compound C and AMPK silencing. In addition, we observed an AMPK-dependent upregulation of glyoxalase 1, a ubiquitous cellular enzyme that participates in the detoxification of MGO. In intravitreal drug-treated mice, we found that AMPK activators can reverse the MGO-induced cotton wool spots, macular edema and retinal damage. Functional, histological and optical coherence tomography analysis support the protective actions of both agents against MGO-elicited retinal damage. Metformin and A769662 via AMPK activation exert a strong protection against MGO-induced retinal pigment epithelial cell death and retinopathy. Therefore, metformin and AMPK activator can be therapeutic agents for DR.
Collapse
Affiliation(s)
- Ponarulselvam Sekar
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - George Hsiao
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Shu-Hao Hsu
- Medical Research Center, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan; Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan.
| | - Chi-Ming Chan
- Department of Ophthalmology, Cardinal Tien Hospital, New Taipei City, Taiwan; School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
16
|
Mortezaee K, Majidpoor J, Kharazinejad E. The impact of hypoxia on tumor-mediated bypassing anti-PD-(L)1 therapy. Biomed Pharmacother 2023; 162:114646. [PMID: 37011483 DOI: 10.1016/j.biopha.2023.114646] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
Extending the durability of response is the current focus in cancer immunotherapy with immune checkpoint inhibitors (ICIs). However, factors like non-immunogenic tumor microenvironment (TME) along with aberrant angiogenesis and dysregulated metabolic systems are negative contributors. Hypoxia is a key TME condition and a critical promoter of tumor hallmarks. It acts on immune and non-immune cells within TME in order for promoting immune evasion and therapy resistance. Extreme hypoxia is a major promoter of resistance to the programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitor therapy. Hypoxia inducible factor-1 (HIF-1) acts as a key mediator of hypoxia and a critical promoter of resistance to the anti-PD-(L)1. Targeting hypoxia or HIF-1 can thus be an effective strategy for reinvigoration of cellular immunity against cancer. Among various strategies presented so far, the key focus is over vascular normalization, which is an approach highly effective for reducing the rate of hypoxia, increasing drug delivery into the tumor area, and boosting the efficacy of anti-PD-(L)1.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Islamic Republic of Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Islamic Republic of Iran
| | - Ebrahim Kharazinejad
- Department of Anatomy, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Islamic Republic of Iran.
| |
Collapse
|
17
|
Karmanova E, Chernikov A, Usacheva A, Ivanov V, Bruskov V. Metformin counters oxidative stress and mitigates adverse effects of radiation exposure: An overview. Fundam Clin Pharmacol 2023. [PMID: 36852652 DOI: 10.1111/fcp.12884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/19/2023] [Accepted: 02/15/2023] [Indexed: 03/01/2023]
Abstract
Metformin (1,1-dimethylbiguanidine hydrochloride) (MF) is a drug that has long been in use for the treatment of type 2 diabetes mellitus and recently is coming into use in the radiation therapy of cancer and other conditions. Exposure to ionizing radiation disturbs the redox homeostasis of cells and causes damage to proteins, membranes, and mitochondria, destroying a number of biological processes. After irradiation, MF activates cellular antioxidant and repair systems by signaling to eliminate the harmful consequences of disruption of redox homeostasis. The use of MF in the treatment of the negative effects of irradiation has great potential in medical patients after radiotherapy and in victims of nuclear accidents or radiologic terrorism.
Collapse
Affiliation(s)
- Ekaterina Karmanova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.,Institute of Cell Biophysics, Pushchino Scientific Center for Biological Research, Federal Research Center of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Anatoly Chernikov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Anna Usacheva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vladimir Ivanov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vadim Bruskov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
18
|
Wu B, Shi X, Jiang M, Liu H. Cross-talk between cancer stem cells and immune cells: potential therapeutic targets in the tumor immune microenvironment. Mol Cancer 2023; 22:38. [PMID: 36810098 PMCID: PMC9942413 DOI: 10.1186/s12943-023-01748-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Ongoing research has revealed that the existence of cancer stem cells (CSCs) is one of the biggest obstacles in the current cancer therapy. CSCs make an influential function in tumor progression, recurrence and chemoresistance due to their typical stemness characteristics. CSCs are preferentially distributed in niches, and those niche sites exhibit characteristics typical of the tumor microenvironment (TME). The complex interactions between CSCs and TME illustrate these synergistic effects. The phenotypic heterogeneity within CSCs and the spatial interactions with the surrounding tumor microenvironment led to increased therapeutic challenges. CSCs interact with immune cells to protect themselves against immune clearance by exploiting the immunosuppressive function of multiple immune checkpoint molecules. CSCs also can protect themselves against immune surveillance by excreting extracellular vesicles (EVs), growth factors, metabolites and cytokines into the TME, thereby modulating the composition of the TME. Therefore, these interactions are also being considered for the therapeutic development of anti-tumor agents. We discuss here the immune molecular mechanisms of CSCs and comprehensively review the interplay between CSCs and the immune system. Thus, studies on this topic seem to provide novel ideas for reinvigorating therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Bo Wu
- grid.459742.90000 0004 1798 5889Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Xiang Shi
- grid.459742.90000 0004 1798 5889Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Meixi Jiang
- grid.412644.10000 0004 5909 0696Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032 China
| | - Hongxu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
19
|
Zhang Y, Zhou F, Guan J, Zhou L, Chen B. Action Mechanism of Metformin and Its Application in Hematological Malignancy Treatments: A Review. Biomolecules 2023; 13:250. [PMID: 36830619 PMCID: PMC9953052 DOI: 10.3390/biom13020250] [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: 11/03/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
Hematologic malignancies (HMs) mainly include acute and chronic leukemia, lymphoma, myeloma and other heterogeneous tumors that seriously threaten human life and health. The common effective treatments are radiotherapy, chemotherapy and hematopoietic stem cell transplantation (HSCT), which have limited options and are prone to tumor recurrence and (or) drug resistance. Metformin is the first-line drug for the treatment of type 2 diabetes (T2DM). Recently, studies identified the potential anti-cancer ability of metformin in both T2DM patients and patients that are non-diabetic. The latest epidemiological and preclinical studies suggested a potential benefit of metformin in the prevention and treatment of patients with HM. The mechanism may involve the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway by metformin as well as other AMPK-independent pathways to exert anti-cancer properties. In addition, combining current conventional anti-cancer drugs with metformin may improve the efficacy and reduce adverse drug reactions. Therefore, metformin can also be used as an adjuvant therapeutic agent for HM. This paper highlights the anti-hyperglycemic effects and potential anti-cancer effects of metformin, and also compiles the in vitro and clinical trials of metformin as an anti-cancer and chemosensitizing agent for the treatment of HM. The need for future research on the use of metformin in the treatment of HM is indicated.
Collapse
Affiliation(s)
| | | | | | | | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
20
|
Rouzbahani E, Majidpoor J, Najafi S, Mortezaee K. Cancer stem cells in immunoregulation and bypassing anti-checkpoint therapy. Biomed Pharmacother 2022; 156:113906. [DOI: 10.1016/j.biopha.2022.113906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/26/2022] Open
|
21
|
Kim JM, Kim H, Oh SH, Jang WI, Lee SB, Park M, Kim S, Park S, Shim S, Jang H. Combined Administration of Pravastatin and Metformin Attenuates Acute Radiation-Induced Intestinal Injury in Mouse and Minipig Models. Int J Mol Sci 2022; 23:ijms232314827. [PMID: 36499155 PMCID: PMC9739896 DOI: 10.3390/ijms232314827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
Radiation-induced gastrointestinal (GI) damage is one of the critical factors that serve as basis for the lethality of nuclear accidents or terrorism. Further, there are no Food and Drug Administration-approved agents available to mitigate radiation-induced intestinal injury. Although pravastatin (PS) has been shown to exhibit anti-inflammatory and epithelial reconstructive effects following radiation exposure using mouse and minipig models, the treatment failed to improve the survival rate of high-dose irradiated intestinal injury. Moreover, we previously found that metformin (MF), a common drug used for treating type 2 diabetes mellitus, has a mitigating effect on radiation-induced enteropathy by promoting stem cell properties. In this study, we investigated whether the combined administration of PS and MF could achieve therapeutic effects on acute radiation-induced intestinal injury in mouse and minipig models. We found that the combined treatment markedly increased the survival rate and attenuated histological damage in a radiation-induced intestinal injury mouse model, in addition to epithelial barrier recovery, anti-inflammatory effects, and improved epithelial proliferation with stem cell properties. Furthermore, in minipig models, combined treatment with PS and MF ameliorates gross pathological damage in abdominal organs and attenuated radiation-induced intestinal histological damage. Therefore, the combination of PS and MF effectively alleviated radiation-induced intestinal injury in the mouse and minipig models. We believe that the combined use of PS and MF is a promising therapeutic approach for treating radiation-induced intestinal injury.
Collapse
Affiliation(s)
- Jung Moon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Su Hyun Oh
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Won Il Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Mineon Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Soyeon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| | - Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| |
Collapse
|
22
|
Weiss M, Nikisher B, Haran H, Tefft K, Adams J, Edwards JG. High throughput screen of small molecules as potential countermeasures to galactic cosmic radiation induced cellular dysfunction. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:76-87. [PMID: 36336373 DOI: 10.1016/j.lssr.2022.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/23/2022] [Accepted: 06/16/2022] [Indexed: 06/16/2023]
Abstract
Space travel increases galactic cosmic ray exposure to flight crews and this is significantly elevated once travel moves beyond low Earth orbit. This includes combinations of high energy protons and heavy ions such as 56Fe or 16O. There are distinct differences in the biological response to low-energy transfer (x-rays) or high-energy transfer (High-LET). However, given the relatively low fluence rate of exposure during flight operations, it might be possible to manage these deleterious effects using small molecules currently available. Virtually all reports to date examining small molecule management of radiation exposure are based on low-LET challenges. To that end an FDA approved drug library (725 drugs) was used to perform a high throughput screen of cultured cells following exposure to galactic cosmic radiation. The H9c2 myoblasts, ES-D3 pluripotent cells, and Hy926 endothelial cell lines were exposed to a single exposure (75 cGy) using the 5-ion GCRsim protocol developed at the NASA Space Radiation Laboratory (NSRL). Following GCR exposure cells were maintained for up to two weeks. For each drug (@10µM), a hierarchical cumulative score was developed incorporating measures of mitochondrial and cellular function, oxidant stress and cell senescence. The top 160 scores were retested following a similar protocol using 1µM of each drug. Within the 160 drugs, 33 are considered to have an anti-inflammatory capacity, while others also indirectly suppressed pro-inflammatory pathways or had noted antioxidant capacity. Lead candidates came from different drug classes that included angiotensin converting enzyme inhibitors or AT1 antagonists, COX2 inhibitors, as well as drugs mediated by histamine receptors. Surprisingly, different classes of anti-diabetic medications were observed to be useful including sulfonylureas and metformin. Using a hierarchical decision structure, we have identified several lead candidates. That no one drug or even drug class was completely successful across all parameters tested suggests the complexity of managing the consequences of galactic cosmic radiation exposure.
Collapse
Affiliation(s)
- M Weiss
- Department of Physiology, New York Medical College, Valhalla, New York
| | - B Nikisher
- Department of Physiology, New York Medical College, Valhalla, New York
| | - H Haran
- Department of Physiology, New York Medical College, Valhalla, New York
| | - K Tefft
- Department of Physiology, New York Medical College, Valhalla, New York
| | - J Adams
- Department of Physiology, New York Medical College, Valhalla, New York
| | - J G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York.
| |
Collapse
|
23
|
Epithelial-mesenchymal transition in cancer stemness and heterogeneity: updated. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:193. [PMID: 36071302 DOI: 10.1007/s12032-022-01801-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/15/2022] [Indexed: 10/14/2022]
Abstract
Epithelial-mesenchymal transition (EMT) as a trans-differentiation program and a key process in tumor progression is linked positively with increased expansion of cancer stem cells and cells with stem-like properties. This is mediated through modulation of critical tumorigenic events and is positively correlated with hypoxic conditions in tumor microenvironment. The presence of cells eliciting diverse phenotypical states inside tumor is representative of heterogeneity and higher tumor resistance to therapy. In this review, we aimed to discuss about the current understanding toward EMT, stemness, and heterogeneity in tumors of solid organs, their contribution to the key tumorigenic events along with major signaling pathway involved, and, finally, to suggest some strategies to target these critical events.
Collapse
|
24
|
Mortezaee K, Majidpoor J. Dysregulated metabolism: A friend-to-foe skewer of macrophages. Int Rev Immunol 2022:1-17. [DOI: 10.1080/08830185.2022.2095374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
25
|
Ye J, Li L, Wang M, Ma Q, Tian Y, Zhang Q, Liu J, Li B, Zhang B, Liu H, Sun G. Diabetes Mellitus Promotes the Development of Atherosclerosis: The Role of NLRP3. Front Immunol 2022; 13:900254. [PMID: 35844498 PMCID: PMC9277049 DOI: 10.3389/fimmu.2022.900254] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/01/2022] [Indexed: 11/22/2022] Open
Abstract
Atherosclerosis is one of the main complications of diabetes mellitus, involving a variety of pathogenic factors. Endothelial dysfunction, inflammation, and oxidative stress are hallmarks of diabetes mellitus and atherosclerosis. Although the ability of diabetes to promote atherosclerosis has been demonstrated, a deeper understanding of the underlying biological mechanisms is critical to identifying new targets. NLRP3 plays an important role in both diabetes and atherosclerosis. While the diversity of its activation modes is one of the underlying causes of complex effects in the progression of diabetes and atherosclerosis, it also provides many new insights for targeted interventions in metabolic diseases.
Collapse
Affiliation(s)
- Jingxue Ye
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lanfang Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiuxiao Ma
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Tian
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiushi Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bengang Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haitao Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibo Sun, ; Haitao Liu,
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibo Sun, ; Haitao Liu,
| |
Collapse
|
26
|
NK and cells with NK-like activities in cancer immunotherapy-clinical perspectives. Med Oncol 2022; 39:131. [PMID: 35716327 DOI: 10.1007/s12032-022-01735-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/13/2022] [Indexed: 01/10/2023]
Abstract
Natural killer (NK) cells are lymphoid cells of innate immunity that take important roles in immune surveillance. NK cells are considered as a bridge between innate and adaptive immunity, and their infiltration into tumor area is related positively with prolonged patient survival. They are defined as CD16+ CD56+ CD3- cells in clinic. NK cells promote cytolytic effects on target cells and induce their apoptosis. Loss of NK cell cytotoxic activity and reduction in the number of activating receptors are the current issues for application of such cells in cellular immunotherapy, which resulted in the diminished long-term effects. The focus of this review is to discuss about the activity of NK cells and cells with NK-like activity including natural killer T (NKT), cytokine-induced killer (CIK) and lymphokine-activated killer (LAK) cells in immunotherapy of human solid cancers.
Collapse
|
27
|
Obrador E, Salvador-Palmer R, Villaescusa JI, Gallego E, Pellicer B, Estrela JM, Montoro A. Nuclear and Radiological Emergencies: Biological Effects, Countermeasures and Biodosimetry. Antioxidants (Basel) 2022; 11:1098. [PMID: 35739995 PMCID: PMC9219873 DOI: 10.3390/antiox11061098] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Atomic and radiological crises can be caused by accidents, military activities, terrorist assaults involving atomic installations, the explosion of nuclear devices, or the utilization of concealed radiation exposure devices. Direct damage is caused when radiation interacts directly with cellular components. Indirect effects are mainly caused by the generation of reactive oxygen species due to radiolysis of water molecules. Acute and persistent oxidative stress associates to radiation-induced biological damages. Biological impacts of atomic radiation exposure can be deterministic (in a period range a posteriori of the event and because of destructive tissue/organ harm) or stochastic (irregular, for example cell mutation related pathologies and heritable infections). Potential countermeasures according to a specific scenario require considering basic issues, e.g., the type of radiation, people directly affected and first responders, range of doses received and whether the exposure or contamination has affected the total body or is partial. This review focuses on available medical countermeasures (radioprotectors, radiomitigators, radionuclide scavengers), biodosimetry (biological and biophysical techniques that can be quantitatively correlated with the magnitude of the radiation dose received), and strategies to implement the response to an accidental radiation exposure. In the case of large-scale atomic or radiological events, the most ideal choice for triage, dose assessment and victim classification, is the utilization of global biodosimetry networks, in combination with the automation of strategies based on modular platforms.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Rosario Salvador-Palmer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - Eduardo Gallego
- Energy Engineering Department, School of Industrial Engineering, Polytechnic University of Madrid, 28040 Madrid, Spain;
| | - Blanca Pellicer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
28
|
Wiernsperger N, Al-Salameh A, Cariou B, Lalau JD. Protection by metformin against severe Covid-19: an in-depth mechanistic analysis. DIABETES & METABOLISM 2022; 48:101359. [PMID: 35662580 PMCID: PMC9154087 DOI: 10.1016/j.diabet.2022.101359] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 12/05/2022]
Abstract
Since the outbreak of Covid-19, several observational studies on diabetes and Covid-19 have reported a favourable association between metformin and Covid-19-related outcomes in patients with type 2 diabetes mellitus (T2DM). This is not surprising since metformin affects many of the pathophysiological mechanisms implicated in SARS-CoV-2 immune response, systemic spread and sequelae. A comparison of the multifactorial pathophysiological mechanisms of Covid-19 progression with metformin's well-known pleiotropic properties suggests that the treatment of patients with this drug might be particularly beneficial. Indeed, metformin could alleviate the cytokine storm, diminish virus entry into cells, protect against microvascular damage as well as prevent secondary fibrosis. Although our in-depth analysis covers many potential metformin mechanisms of action, we want to highlight more particularly its unique microcirculatory protective effects since worsening of Covid-19 disease clearly appears as largely due to severe defects in the structure and functioning of microvessels. Overall, these observations confirm that metformin is a unique, pleiotropic drug that targets many of Covid-19′s pathophysiology processes in a diabetes-independent manner.
Collapse
Affiliation(s)
| | - Abdallah Al-Salameh
- Department of Endocrinology, Diabetes Mellitus and Nutrition, Amiens University Hospital, Amiens, France; PériTox/UMR-I 01, University of Picardie Jules Verne, Amiens, France
| | - Bertrand Cariou
- Département d'Endocrinologie, Diabétologie et Nutrition, l'institut du thorax, Inserm, CNRS, UNIV Nantes, CHU Nantes, Hôpital Guillaume et René Laennec, 44093 Nantes Cedex 01, France
| | - Jean-Daniel Lalau
- Department of Endocrinology, Diabetes Mellitus and Nutrition, Amiens University Hospital, Amiens, France; PériTox/UMR-I 01, University of Picardie Jules Verne, Amiens, France.
| |
Collapse
|
29
|
Metformin Protects the Intestinal Barrier by Activating Goblet Cell Maturation and Epithelial Proliferation in Radiation-Induced Enteropathy. Int J Mol Sci 2022; 23:ijms23115929. [PMID: 35682612 PMCID: PMC9180746 DOI: 10.3390/ijms23115929] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Radiotherapy or accidental exposure to high-dose radiation can cause severe damage to healthy organs. The gastrointestinal (GI) tract is a radiation-sensitive organ of the body. The intestinal barrier is the first line of defense in the GI tract, and consists of mucus secreted by goblet cells and a monolayer of epithelium. Intestinal stem cells (ISCs) help in barrier maintenance and intestinal function after injury by regulating efficient regeneration of the epithelium. The Wnt/β-catenin pathway plays a critical role in maintaining the intestinal epithelium and regulates ISC self-renewal. Metformin is the most widely used antidiabetic drug in clinical practice, and its anti-inflammatory, antioxidative, and antiapoptotic effects have also been widely studied. In this study, we investigated whether metformin alleviated radiation-induced enteropathy by focusing on its role in protecting the epithelial barrier. We found that metformin alleviated radiation-induced enteropathy, with increased villi length and crypt numbers, and restored the intestinal barrier function in the irradiated intestine. In a radiation-induced enteropathy mouse model, metformin treatment increased tight-junction expression in the epithelium and inhibited bacterial translocation to mesenteric lymph nodes. Metformin increased the number of ISCs from radiation toxicity and enhanced epithelial repair by activating Wnt/β-catenin signaling. These data suggested that metformin may be a potential therapeutic agent for radiation-induced enteropathy.
Collapse
|
30
|
Roles for macrophage-polarizing interleukins in cancer immunity and immunotherapy. Cell Oncol (Dordr) 2022; 45:333-353. [PMID: 35587857 DOI: 10.1007/s13402-022-00667-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 12/14/2022] Open
Abstract
Macrophages are the most abundant and one of the most critical cells of tumor immunity. They provide a bridge between innate and adaptive immunity through releasing cytokines into the tumor microenvironment (TME). A number of interleukin (IL) cytokine family members is involved in shaping the final phenotype of macrophages toward either a classically-activated pro-inflammatory M1 state with anti-tumor activity or an alternatively-activated anti-inflammatory M2 state with pro-tumor activity. Shaping TME macrophages toward the M1 phenotype or recovering this phenotypic state may offer a promising therapeutic approach in patients with cancer. Here, we focus on the impact of macrophage-polarizing ILs on immune cells and IL-mediated cellular cross-interactions within the TME. The key aim of this review is to define therapeutic schedules for addressing ILs in cancer immunotherapy based on their multi-directional impacts in such a milieu. Gathering more knowledge on this area is also important for defining adverse effects related to cytokine therapy and addressing them for reinforcing the efficacy of immunotherapy against cancer.
Collapse
|
31
|
Espinal A, Epperly MW, Mukherjee A, Fisher R, Shields D, Wang H, Huq MS, Hamade DF, Vlad AM, Coffman L, Buckanovich R, Yu J, Leibowitz BJ, van Pijkeren JP, Patel RB, Stolz D, Watkins S, Ejaz A, Greenberger JS. Intestinal Radiation Protection and Mitigation by Second-Generation Probiotic Lactobacillus-reuteri Engineered to Deliver Interleukin-22. Int J Mol Sci 2022; 23:5616. [PMID: 35628427 PMCID: PMC9145862 DOI: 10.3390/ijms23105616] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/13/2022] Open
Abstract
(1) Background: The systemic administration of therapeutic agents to the intestine including cytokines, such as Interleukin-22 (IL-22), is compromised by damage to the microvasculature 24 hrs after total body irradiation (TBI). At that time, there is significant death of intestinal microvascular endothelial cells and destruction of the lamina propria, which limits drug delivery through the circulation, thus reducing the capacity of therapeutics to stabilize the numbers of Lgr5+ intestinal crypt stem cells and their progeny, and improve survival. By its direct action on intestinal stem cells and their villus regeneration capacity, IL-22 is both an ionizing irradiation protector and mitigator. (2) Methods: To improve delivery of IL-22 to the irradiated intestine, we gavaged Lactobacillus-reuteri as a platform for the second-generation probiotic Lactobacillus-reuteri-Interleukin-22 (LR-IL-22). (3) Results: There was effective radiation mitigation by gavage of LR-IL-22 at 24 h after intestinal irradiation. Multiple biomarkers of radiation damage to the intestine, immune system and bone marrow were improved by LR-IL-22 compared to the gavage of control LR or intraperitoneal injection of IL-22 protein. (4) Conclusions: Oral administration of LR-IL-22 is an effective protector and mitigator of intestinal irradiation damage.
Collapse
Affiliation(s)
- Alexis Espinal
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - M. Saiful Huq
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Diala Fatima Hamade
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Anda M. Vlad
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - Lan Coffman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA; (L.C.); (R.B.)
| | - Ronald Buckanovich
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA; (L.C.); (R.B.)
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232, USA; (J.Y.); (B.J.L.)
| | - Brian J. Leibowitz
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232, USA; (J.Y.); (B.J.L.)
| | | | - Ravi B. Patel
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Donna Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15232, USA; (D.S.); (S.W.)
| | - Simon Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15232, USA; (D.S.); (S.W.)
| | - Asim Ejaz
- Department of Plastic and Reconstructive Surgery, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| |
Collapse
|
32
|
Extermann M, Chetty IJ, Brown SL, Al-Jumayli M, Movsas B. Predictors of Toxicity Among Older Adults with Cancer. Semin Radiat Oncol 2022; 32:179-185. [DOI: 10.1016/j.semradonc.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Mortezaee K, Majidpoor J. Checkpoint inhibitor/interleukin-based combination therapy of cancer. Cancer Med 2022; 11:2934-2943. [PMID: 35301813 PMCID: PMC9359865 DOI: 10.1002/cam4.4659] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/12/2022] [Accepted: 02/23/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Immunotherapy using immune checkpoint inhibitors (ICIs) is the current focus in cancer immunotherapy. However, issues are raised in the area, as the recent studies showed that such therapeutic modality suffers from low durability and low or no efficacy for patients with some tumor types including cases with non-inflamed or cold cancers. Therefore, efforts have been made to solve the issue using immune combination therapy, such as the use of immunocytokines. The combination of ICI with interleukins (ILs) and IL-targeting agents is now under consideration in the area of therapy, and the primary results are promising. PURPOSE The focus of this review is to discuss the possibility of using ILs and IL-targeting drugs in combination with ICI in cancer immunotherapy and describing recent advances in the field using PEGylated ILs and fusion proteins. The key focus in this area is to reduce adverse events and to increase the efficacy and durability of such combination therapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
34
|
Mortezaee K, Majidpoor J. (Im)maturity in Tumor Ecosystem. Front Oncol 2022; 11:813897. [PMID: 35145911 PMCID: PMC8821092 DOI: 10.3389/fonc.2021.813897] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023] Open
Abstract
Tumors have special features that make them distinct from their normal counterparts. Immature cells in a tumor mass and their critical contributions to the tumorigenesis will open new windows toward cancer therapy. Incomplete cellular development brings versatile and unique functionality in the cellular tumor ecosystem, such as what is seen for highly potential embryonic cells. There is evidence that maturation of certain types of cells in this ecosystem can recover the sensitivity of the tumor. Therefore, understanding more about the mechanisms that contributed to this immaturity will render new therapeutic approaches in cancer therapy. Targeting such mechanisms can be exploited as a supplementary to the current immunotherapeutic treatment schedules, such as immune checkpoint inhibitor (ICI) therapy. The key focus of this review is to discuss the impact of (im)maturity in cellular tumor ecosystems on cancer progression, focusing mainly on immaturity in the immune cell compartment of the tumor, as well as on the stemness of tumor cells.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
35
|
Majidpoor J, Mortezaee K. Interleukin-6 in SARS-CoV-2 induced disease: Interactions and therapeutic applications. Biomed Pharmacother 2022; 145:112419. [PMID: 34781146 PMCID: PMC8585600 DOI: 10.1016/j.biopha.2021.112419] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/07/2023] Open
Abstract
Interleukin-6 (IL-6) is a multi-tasking cytokine that represents high activity in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and cancer. High concentration of this pleiotropic cytokine accounts for hyperinflammation and cytokine storm, and is related to multi-organ failure in patients with SARS-CoV-2 induced disease. IL-6 promotes lymphopenia and increases C-reactive protein (CRP) in such cases. However, blockade of IL-6 is not a full-proof of complete response. Hypoxia, hypoxemia, aberrant angiogenesis and chronic inflammation are inter-related events occurring as a response to the SARS-CoV-2 stimulatory effect on high IL-6 activity. Taking both pro- and anti-inflammatory activities will make complex targeting IL-6 in patient with SARS-CoV-2 induced disease. The aim of this review was to discuss about interactions occurring within the body of patients with SARS-CoV-2 induced disease who are representing high IL-6 levels, and to determine whether IL-6 inhibition therapy is effective for such patients or not. We also address the interactions and targeted therapies in cancer patients who also have SARS-CoV-2 induced disease.
Collapse
Affiliation(s)
- Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
36
|
Mortezaee K, Majidpoor J. The impact of hypoxia on immune state in cancer. Life Sci 2021; 286:120057. [PMID: 34662552 DOI: 10.1016/j.lfs.2021.120057] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022]
Abstract
Hypoxia is a known feature of solid tumors and a critical promoter of tumor hallmarks. Hypoxia influences tumor immunity in a way favoring immune evasion and resistance. Extreme hypoxia and aberrant hypoxia-inducible factor-1 (HIF-1) activity in tumor microenvironment (TME) is a drawback for effective immunotherapy. Infiltration and activity of CD8+ T cells is reduced in such condition, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) show high activities. Highly hypoxic TME also impairs maturation and activity of dendritic cell (DCs) and natural killer (NK) cells. In addition, the hypoxic TME positively is linked positively with metabolic changes in cells of immune system. These alterations are indicative of a need for hypoxia modulation as a complementary targeting strategy to go with immune checkpoint inhibitor (ICI) therapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
37
|
Yang R, Tan C, Najafi M. Cardiac inflammation and fibrosis following chemo/radiation therapy: mechanisms and therapeutic agents. Inflammopharmacology 2021; 30:73-89. [PMID: 34813027 DOI: 10.1007/s10787-021-00894-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
The incidence of cardiovascular disorders is one of the most concerns among people who underwent cancer therapy. The heart side effects of cancer therapy may occur during treatment to some years after the end of treatment. Some epidemiological studies confirm that heart diseases are one of the most common reasons for mortality among patients that were received treatment for cancer. Experimental studies and also clinical investigations indicate that inflammatory changes such as pericarditis, myocarditis, and also fibrosis are key mechanisms of cardiac diseases following chemotherapy/radiotherapy. It seems that chronic oxidative stress, massive cell death, and chronic overproduction of pro-inflammatory and pro-fibrosis cytokines are the key mechanisms of cardiovascular diseases following cancer therapy. Furthermore, infiltration of inflammatory cells and upregulation of some enzymes such as NADPH Oxidases are a hallmark of heart diseases after cancer therapy. In the current review, we aim to explain how radiation or chemotherapy can induce inflammatory and fibrosis-related diseases in the heart. We will explain the cellular and molecular mechanisms of cardiac inflammation and fibrosis following chemo/radiation therapy, and then review some adjuvants to reduce the risk of inflammation and fibrosis in the heart.
Collapse
Affiliation(s)
- Run Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Road, Changsha, Hunan, People's Republic of China
| | - Changming Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Road, Changsha, Hunan, People's Republic of China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
38
|
Mortezaee K, Majidpoor J. Key promoters of tumor hallmarks. Int J Clin Oncol 2021; 27:45-58. [PMID: 34773527 DOI: 10.1007/s10147-021-02074-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/31/2021] [Indexed: 02/06/2023]
Abstract
Evolution of tumor hallmarks is a result of accommodation of tumor cells with their nearby milieu called tumor microenvironment (TME). Accommodation or adaptive responses is highly important for a cell to survive, without which no cell is allowed to take any further steps in tumorigenesis. Metabolism of cancer cells is largely depended on stroma. Composition and plasticity of cells within the stroma is highly affected from inflammatory setting of TME. Hypoxia which is a common event in many solid cancers, is known as one of the key hallmarks of chronic inflammation and the master regulator of metastasis. Transforming growth factor (TGF)-β is produced in the chronic inflammatory and chronic hypoxic settings, and it is considered as a cardinal factor for induction of all tumor hallmarks. Aging, obesity and smoking are the main predisposing factors of cancer, acting mainly through modulation of TME.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
39
|
Hou CP, Tsui KH, Chang KS, Sung HC, Hsu SY, Lin YH, Yang PS, Chen CL, Feng TH, Juang HH. Caffeic acid phenethyl ester inhibits the growth of bladder carcinoma cells by upregulating growth differentiation factor 15. Biomed J 2021; 45:763-775. [PMID: 34662721 DOI: 10.1016/j.bj.2021.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Caffeic acid phenethyl ester (CAPE), a bioactive component of propolis, has beneficial effects on cancer prevention. Growth differentiation factor 15 (GDF15) is an antitumor gene of bladder cancer. Therefore, this study investigated the anti-cancer effect of CAPE on bladder carcinoma cells and related mechanisms. METHODS The expressions of GDF15, N-myc downstream-regulated gene 1 (NDRG1), and maspin, and the activations of ERK, JNK, p38, and AMPKα1/2 in human bladder cells after gene transfection or knockdown were determined by immunoblot, RT-qPCR, and reporter assays. The assays of 5-ethynyl-2'-deoxyuridine (EdU), CyQUANT cell proliferation, and Matrigel invasion, and the xenograft animal study were used to assess the cell proliferation, invasion, and tumorigenesis. RESULTS GDF15 expression in epithelial cells was negatively correlated with neoplasia in vitro. Also, GDF15 exhibits in bladder fibroblasts and smooth muscle cells. CAPE-induced expressions of NDRG1 and maspin decreased cell proliferation and invasion of bladder carcinoma cells in a GDF15-dependent manner in vitro. The xenograft animal study suggesting CAPE attenuated tumor growth in vivo. CAPE increased phosphorylation of ERK, JNK, p38, and AMPKα1/2 to modulate the GDF15 expressions. Pretreatments with ERK, JNK, or p38 inhibitors partially inhibited the CAPE effects on the inductions of GDF15, NDRG1, or maspin. Knockdown of AMPKα1/2 attenuated the CAPE-induced GDF15 expression and cell proliferation in bladder carcinoma cells. CONCLUSIONS Our findings indicate that CAPE is a promising agent for anti-tumor growth in human bladder carcinoma cells via the upregulation of GDF15.
Collapse
Affiliation(s)
- Chen-Pang Hou
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Shuang Ho Hospital, New Taipei City, Taiwan; Department of Medicine; TMU Research Center of Urology and Kindey, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Pei-Shan Yang
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chien-Lun Chen
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Tsui-Hsia Feng
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; School of Nursing, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
40
|
Wu Z, Zhang C, Najafi M. Targeting of the tumor immune microenvironment by metformin. J Cell Commun Signal 2021; 16:333-348. [PMID: 34611852 DOI: 10.1007/s12079-021-00648-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Stimulating antitumor immunity is an attractive idea for suppressing tumors. CD4 + and CD8 + T cells as well as natural killer cells (NK) are the primary antitumor immune cells in the tumor microenvironment (TME). In contrast to these cells, regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs) release several molecules to suppress antitumor immunity and stimulate cancer cell invasion and proliferation. Adjuvant treatment with certain nontoxic agents is interesting to boost antitumor immunity. Metformin, which is known as an antidiabetes drug, can modulate both antitumor and protumor immune cells within TME. It has the ability to induce the proliferation of CD8 + T lymphocytes and NK cells. On the other hand, metformin attenuates polarization toward TAMs, CAFs, and Tregs. Metformin also may stimulate the antitumor activity of immune system cells, while it interrupts the positive cross-talk and interactions between immunosuppressive cells and cancer cells. The purpose of this review is to explain the basic mechanisms for the interactions and communications between immunosuppressive, anti-tumoral, and cancer cells within TME. Next, we discuss the modulating effects of metformin on various cells and secretions in TME.
Collapse
Affiliation(s)
- Zihong Wu
- Department of Oncology, The NO.3 People's Hospital of Hubei Province, Jianghan University, Wuhan, 430033, Hubei, China
| | - Caidie Zhang
- Emergency Department, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, 430014, Hubei, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
41
|
Yu C, Yang B, Najafi M. Targeting of cancer cell death mechanisms by curcumin: Implications to cancer therapy. Basic Clin Pharmacol Toxicol 2021; 129:397-415. [PMID: 34473898 DOI: 10.1111/bcpt.13648] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/06/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022]
Abstract
Cancer is known as a second major cause of death globally. Nowadays, several modalities have been developed for the treatment of cancer. Radiotherapy and chemotherapy are the most common modalities in most countries. However, newer modalities such as immunotherapy and targeted therapy drugs can kill cancer cells with minimal side effects. All anticancer agents work based on the killing of cancer cells. Numerous studies are ongoing to kill cancer cells more effectively without increasing side effects to normal tissues. The combination modalities with low toxic agents are interesting for this aim. Curcumin is one of the most common herbal agents that has shown several anticancer properties. It can regulate immune system responses against cancer. Furthermore, curcumin has been shown to potentiate cell death signalling pathways and attenuate survival signalling pathways in cancer cells. The knowledge of how curcumin induces cell death in cancers can improve therapeutic efficiency. In this review, the regulatory effects of curcumin on different cell death mechanisms and their signalling pathways will be discussed. Furthermore, we explain how curcumin may potentiate the anticancer effects of other drugs or radiotherapy through modulation of apoptosis, mitotic catastrophe, senescence, autophagy and ferroptosis.
Collapse
Affiliation(s)
- Chong Yu
- School of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin, China
| | - Bo Yang
- School of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin, China
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
42
|
Najafi M, Majidpoor J, Toolee H, Mortezaee K. The current knowledge concerning solid cancer and therapy. J Biochem Mol Toxicol 2021; 35:e22900. [PMID: 34462987 DOI: 10.1002/jbt.22900] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/29/2021] [Accepted: 08/20/2021] [Indexed: 12/25/2022]
Abstract
Solid cancers comprise a large number of new cases and deaths from cancer each year globally. There are a number of strategies for addressing tumors raised from solid organs including surgery, chemotherapy, radiotherapy, targeted therapy, immunotherapy, combinational therapy, and stem cell and extracellular vesicle (EV) therapy. Surgery, radiotherapy, and chemotherapy are the dominant cures, but are not always effective, in which even in a localized tumor there is a possibility of tumor relapse after surgical resection. Over half of the cancer patients will receive radiotherapy as a part of their therapeutic schedule. Radiotherapy can cause an abscopal response for boosting the activity of the immune system outside the local field of radiation, but it may also cause an unwanted bystander effect, predisposing nonradiated cells into carcinogenesis. In the context of immunotherapy, immune checkpoint inhibition is known as the standard-of-care, but the major concern is in regard with cold cancers that show low responses to such therapy. Stem-cell therapy can be used to send prodrugs toward the tumor area; this strategy, however, has its own predicaments, such as unwanted attraction toward the other sites including healthy tissues and its instability. A substitute to such therapy and quite a novel strategy is to use EVs, by virtue of their stability and potential to cross biological barriers and long-term storage of contents. Combination therapy is the current focus. Despite advances in the field, there are still unmet concerns in the area of effective cancer therapy, raising challenges and opportunities for future investigations.
Collapse
Affiliation(s)
- 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
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Heidar Toolee
- Department of Anatomy, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Keywan Mortezaee
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
43
|
Arabzadeh A, Mortezazadeh T, Aryafar T, Gharepapagh E, Majdaeen M, Farhood B. Therapeutic potentials of resveratrol in combination with radiotherapy and chemotherapy during glioblastoma treatment: a mechanistic review. Cancer Cell Int 2021; 21:391. [PMID: 34289841 PMCID: PMC8296583 DOI: 10.1186/s12935-021-02099-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma, WHO grade IV astrocytoma, is the most aggressive type of brain tumors. These cancerous cells have a rapid growth rate, tendency to penetrate vital brain structures, molecular heterogeneity, etc. and this cancer is associated with a poor prognosis and low survival rate. Due to the resistance of glioblastoma cells to conventional therapeutic modalities (such as radiation therapy and chemotherapy) as well as the adverse effects of these modalities, the researchers have attempted to discover an appropriate alternative or adjuvant treatment for glioblastoma. Resveratrol, as an herbal and natural polyphenolic compound, has anti-tumoral property and has shown to be effective in GBM treatment. Resveratrol exerts its anti-tumoral effect through various mechanisms such as regulation of cell cycle progression and cell proliferation, autophagy, oxidant system, apoptosis pathways, and so on. Resveratrol in combination with radiation therapy and chemotherapy has also been used. In the present study, we summarized the current findings on therapeutic potentials of resveratrol in glioblastoma radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- AmirAhmad Arabzadeh
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Tohid Mortezazadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Tayebeh Aryafar
- Department of Radiation Sciences, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Esmaeil Gharepapagh
- Medical Radiation Sciences Research Team , Tabriz University of Medical Science, Tabriz, Iran
| | - Mehrsa Majdaeen
- Department of Radiotherapy and Oncology, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
44
|
Da F, Guo J, Yao L, Gao Q, Jiao S, Miao X, Liu J. Pretreatment with metformin protects mice from whole-body irradiation. JOURNAL OF RADIATION RESEARCH 2021; 62:618-625. [PMID: 33912960 PMCID: PMC8273805 DOI: 10.1093/jrr/rrab012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/22/2020] [Indexed: 05/04/2023]
Abstract
Metformin, a first-line oral drug for type II diabetes mellitus, not only reduces blood glucose levels, but also has many other biological effects. Recent studies have been conducted to determine the protective effect of metformin in irradiation injuries. However, the results are controversial and mainly focus on the time of metformin administration. In this study, we aimed to investigate the protective effect of metformin in BALB/c mice exposed to 6 Gy or 8 Gy of a 60Co source of γ-rays for total body irradiation (TBI). Survival outcomes were assessed following exposure to 8 Gy or 6 Gy TBI, and hematopoietic damage and intestinal injury were assessed after exposure to 6 Gy TBI. Metformin prolonged the survival of mice exposed to 8 Gy TBI and improved the survival rate of mice exposed to 6 Gy TBI only when administered before exposure to irradiation. Moreover, pretreatment with metformin reduced the frequency of micronuclei (MN) in the bone marrow of mice exposed to 6 Gy TBI. Pretreatment of metformin also protected the intestinal morphology of mice, reduced inflammatory response and decreased the number of apoptotic cells in intestine. In conclusion, we demonstrated that pretreatment with metformin could alleviate irradiation injury.
Collapse
Affiliation(s)
- Fei Da
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710032, China
- Pharmaceutical Preparation Section, The No. 967 Hospital of PLA Joint Logistics Support Force, Dalian 116041, China
| | - Juan Guo
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710032, China
| | - Lin Yao
- Department of Pharmaceutical chemistry and Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Qiaohui Gao
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710032, China
| | - Shengyuan Jiao
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710032, China
| | - Xia Miao
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710032, China
| | - Junye Liu
- Department of Radiation Medical Protection, School of Military Preventive Medicine, Air Force Medical University, Xi’an 710032, China
| |
Collapse
|
45
|
Hansel C, Barr S, Schemann AV, Lauber K, Hess J, Unger K, Zitzelsberger H, Jendrossek V, Klein D. Metformin Protects against Radiation-Induced Acute Effects by Limiting Senescence of Bronchial-Epithelial Cells. Int J Mol Sci 2021; 22:7064. [PMID: 34209135 PMCID: PMC8268757 DOI: 10.3390/ijms22137064] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022] Open
Abstract
Radiation-induced damage to normal lung parenchyma remains a dose-limiting factor in thorax-associated radiotherapy (RT). Severe early and late complications with lungs can increase the risk of morbidity in cancer patients after RT. Herein, senescence of lung epithelial cells following RT-induced cellular stress, or more precisely the respective altered secretory profile, the senescence-associated secretory phenotype (SASP), was suggested as a central process for the initiation and progression of pneumonitis and pulmonary fibrosis. We previously reported that abrogation of certain aspects of the secretome of senescent lung cells, in particular, signaling inhibition of the SASP-factor Ccl2/Mcp1 mediated radioprotection especially by limiting endothelial dysfunction. Here, we investigated the therapeutic potential of a combined metformin treatment to protect normal lung tissue from RT-induced senescence and associated lung injury using a preclinical mouse model of radiation-induced pneumopathy. Metformin treatment efficiently limited RT-induced senescence and SASP expression levels, thereby limiting vascular dysfunctions, namely increased vascular permeability associated with increased extravasation of circulating immune and tumor cells early after irradiation (acute effects). Complementary in vitro studies using normal lung epithelial cell lines confirmed the senescence-limiting effect of metformin following RT finally resulting in radioprotection, while fostering RT-induced cellular stress of cultured malignant epithelial cells accounting for radiosensitization. The radioprotective action of metformin for normal lung tissue without simultaneous protection or preferable radiosensitization of tumor tissue might increase tumor control probabilities and survival because higher radiation doses could be used.
Collapse
Affiliation(s)
- Christine Hansel
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| | - Samantha Barr
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| | - Alina V. Schemann
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU München, 80539 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, 80539 Munich, Germany
- Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.H.); (K.U.); (H.Z.)
| | - Julia Hess
- Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.H.); (K.U.); (H.Z.)
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Kristian Unger
- Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.H.); (K.U.); (H.Z.)
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Horst Zitzelsberger
- Clinical Cooperation Group ‘Personalized Radiotherapy in Head and Neck Cancer’ Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany; (J.H.); (K.U.); (H.Z.)
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| | - Diana Klein
- Institute of Cell Biology (Cancer Research), University Hospital, Essen, University of Duisburg-Essen, 45122 Essen, Germany; (C.H.); (S.B.); (A.V.S.); (V.J.)
| |
Collapse
|
46
|
Mortezaee K. Normalization in tumor ecosystem: Opportunities and challenges. Cell Biol Int 2021; 45:2017-2030. [PMID: 34189798 DOI: 10.1002/cbin.11655] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/24/2021] [Accepted: 06/17/2021] [Indexed: 12/15/2022]
Abstract
Current research in cancer therapy aims to exploit efficient strategies to have long-lasting effects on tumors and to reduce or even revoke the chance of recurrence. Within the tumor stroma, O2 and nutrients are abnormally distributed between various cells (preferentially for supplying cancer cells), the immune contexture is abnormally positioned (permissive essentially for cells exhibiting tumor-promoting capacity), the fibroblast and fibrotic content is abnormally distributed (presence of both extracellular matrix [ECM] stiffening and ECM-degrading factors both for tumor-promoting purposes), and the tumor vasculature is abnormally orchestrated (for hindering drug delivery and increasing the chance of tumor metastasis). Resistance is actually an adaptive response to an imbalance in the tumor ecosystem; thus, the key consideration for effective cancer therapy is to bring back the normal status in this ecosystem so as to reach the desired durable outcome. Vascular normalization, metabolic modulation (glucose delivery in particular), balancing cellular dispersion, and balancing the pH rate and O2 delivery within the tumor microenvironment are suggested strategies to reverse abnormality within the tumor stroma.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
47
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Shabeeb D, Musa AE. Resveratrol as an Adjuvant for Normal Tissues Protection and Tumor Sensitization. Curr Cancer Drug Targets 2021; 20:130-145. [PMID: 31738153 DOI: 10.2174/1568009619666191019143539] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/12/2019] [Accepted: 07/22/2019] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most complicated diseases in present-day medical science. Yearly, several studies suggest various strategies for preventing carcinogenesis. Furthermore, experiments for the treatment of cancer with low side effects are ongoing. Chemotherapy, targeted therapy, radiotherapy and immunotherapy are the most common non-invasive strategies for cancer treatment. One of the most challenging issues encountered with these modalities is low effectiveness, as well as normal tissue toxicity for chemo-radiation therapy. The use of some agents as adjuvants has been suggested to improve tumor responses and also alleviate normal tissue toxicity. Resveratrol, a natural flavonoid, has attracted a lot of attention for the management of both tumor and normal tissue responses to various modalities of cancer therapy. As an antioxidant and anti-inflammatory agent, in vitro and in vivo studies show that it is able to mitigate chemo-radiation toxicity in normal tissues. However, clinical studies to confirm the usage of resveratrol as a chemo-radioprotector are lacking. In addition, it can sensitize various types of cancer cells to both chemotherapy drugs and radiation. In recent years, some clinical studies suggested that resveratrol may have an effect on inducing cancer cell killing. Yet, clinical translation of resveratrol has not yielded desirable results for the combination of resveratrol with radiotherapy, targeted therapy or immunotherapy. In this paper, we review the potential role of resveratrol for preserving normal tissues and sensitization of cancer cells in combination with different cancer treatment modalities.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48175-861, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed E Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| |
Collapse
|
48
|
Mortezaee K. Enriched cancer stem cells, dense stroma, and cold immunity: Interrelated events in pancreatic cancer. J Biochem Mol Toxicol 2021; 35:e22708. [PMID: 33491255 DOI: 10.1002/jbt.22708] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/27/2020] [Accepted: 01/08/2021] [Indexed: 12/11/2022]
Abstract
Cold tumors generally show low mutational burden and low infiltration of effector T cells. The pancreas, prostate, ovary, breast, and colon are placed into the category of cold tumors. In such tumors, effector T cells are either excluded from the tumor area or taken away from being in contact with tumor cells. The stromal reaction in the form of desmoplasia is important for the pathogenesis of tumors like the pancreas. Besides acting as a barrier for the penetration of drugs into the tumor area, the dense stroma presumably creates an immunosuppressive tumor microenvironment (TME), which accounts for low responses from tumor to immunotherapy. Cancer stem cells (CSCs) are an important part of the immunosuppressive complex within the TME. The presence of CSCs within the TME is related negatively to the activity of the antitumor immune system. Here, the question is how desmoplastic aggregates can influence the functionality of CSCs for promoting a cold pancreatic tumor immunity? This review is aimed at responding to this question, the disruption of which can be an effective strategy for improving responses from cold tumors to immunotherapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
49
|
Hypoxia-Induced Cancer Cell Responses Driving Radioresistance of Hypoxic Tumors: Approaches to Targeting and Radiosensitizing. Cancers (Basel) 2021; 13:cancers13051102. [PMID: 33806538 PMCID: PMC7961562 DOI: 10.3390/cancers13051102] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/21/2021] [Accepted: 02/25/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Some regions of aggressive malignancies experience hypoxia due to inadequate blood supply. Cancer cells adapting to hypoxic conditions somehow become more resistant to radiation exposure and this decreases the efficacy of radiotherapy toward hypoxic tumors. The present review article helps clarify two intriguing points: why hypoxia-adapted cancer cells turn out radioresistant and how they can be rendered more radiosensitive. The critical molecular targets associated with intratumoral hypoxia and various approaches are here discussed which may be used for sensitizing hypoxic tumors to radiotherapy. Abstract Within aggressive malignancies, there usually are the “hypoxic zones”—poorly vascularized regions where tumor cells undergo oxygen deficiency through inadequate blood supply. Besides, hypoxia may arise in tumors as a result of antiangiogenic therapy or transarterial embolization. Adapting to hypoxia, tumor cells acquire a hypoxia-resistant phenotype with the characteristic alterations in signaling, gene expression and metabolism. Both the lack of oxygen by itself and the hypoxia-responsive phenotypic modulations render tumor cells more radioresistant, so that hypoxic tumors are a serious challenge for radiotherapy. An understanding of causes of the radioresistance of hypoxic tumors would help to develop novel ways for overcoming this challenge. Molecular targets for and various approaches to radiosensitizing hypoxic tumors are considered in the present review. It is here analyzed how the hypoxia-induced cellular responses involving hypoxia-inducible factor-1, heat shock transcription factor 1, heat shock proteins, glucose-regulated proteins, epigenetic regulators, autophagy, energy metabolism reprogramming, epithelial–mesenchymal transition and exosome generation contribute to the radioresistance of hypoxic tumors or may be inhibited for attenuating this radioresistance. The pretreatments with a multitarget inhibition of the cancer cell adaptation to hypoxia seem to be a promising approach to sensitizing hypoxic carcinomas, gliomas, lymphomas, sarcomas to radiotherapy and, also, liver tumors to radioembolization.
Collapse
|
50
|
Najafi M, Cheki M, Hassanzadeh G, Amini P, Shabeeb D, Musa AE. Protection from Radiation-induced Damage in Rat's Ileum and Colon by Combined Regimens of Melatonin and Metformin: A Histopathological Study. Antiinflamm Antiallergy Agents Med Chem 2021; 19:180-189. [PMID: 31438832 PMCID: PMC7475942 DOI: 10.2174/1871523018666190718161928] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/22/2019] [Accepted: 06/28/2019] [Indexed: 01/07/2023]
Abstract
Background: Radiation-induced enteritis and proctitis are common side effects of abdominopelvic cancers among patients that undergo radiotherapy for prostate, colorectal or urinary cancers. Exposure of these tissues to high doses of radiation leads to damage to villous, inflammation, pain, ulcer and bleeding, which may cause malabsorption and gastrointestinal disorders. To date, several procedures such as pharmaceutical treatment have been proposed for protection and mitigation of gastrointestinal toxicity following radiotherapy. Aims: In the current study, we aimed to investigate the possible radioprotection of ileum and colon in rats using a combination of melatonin and metformin. Methods: In this experimental study, 30 male Wistar rats were randomly assigned to six groups: control, melatonin (100 mg/kg) treatment, melatonin (100 mg/kg) plus metformin (100 mg/kg) treatment, radiation (10 Gy to whole body) group, radiation + melatonin (100 mg/kg) treatment, and radiation + melatonin (100 mg/kg) plus metformin (100 mg/kg) treatment. After 3.5 days, rats were sacrificed and their ileum and colon tissues carefully removed. Histopathological evaluations were conducted on these tissue samples. Results: Histological evaluations reported moderate to severe damages to ileum and colon following whole body irradiation. Melatonin administration was able to protect the ileum remarkably, while the combination of melatonin and metformin was less effective. Interestingly, for the colon, melatonin was less effective while its combination with metformin was able to protect against radiation toxicity completely. Conclusion: For the ileum, melatonin was a more effective radioprotector compared to its combination with metformin. However, the combination of melatonin and metformin can be proposed as an ideal radioprotector for the colon.
Collapse
Affiliation(s)
- Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Cheki
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Science, International Campus, Tehran, Iran.,Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed E Musa
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Science, International Campus, Tehran, Iran.,Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|