1
|
Delgado-Waldo I, Dokudovskaya S, Loissell-Baltazar YA, Pérez-Arteaga E, Coronel-Hernández J, Martínez-Vázquez M, Pérez-Yépez EA, Lopez-Saavedra A, Jacobo-Herrera N, Pérez Plasencia C. Laherradurin Inhibits Colorectal Cancer Cell Growth by Induction of Mitochondrial Dysfunction and Autophagy Induction. Cells 2024; 13:1649. [PMID: 39404412 PMCID: PMC11475353 DOI: 10.3390/cells13191649] [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: 09/09/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
LAH, an acetogenin from the Annonaceae family, has demonstrated antitumor activity in several cancer cell lines and in vivo models, where it reduced the tumor size and induced programmed cell death. We focused on the effects of LAH on mitochondrial dynamics, mTOR signaling, autophagy, and apoptosis in colorectal cancer (CRC) cells to explore its anticancer potential. METHODS CRC cells were treated with LAH, and its effects on mitochondrial respiration and glycolysis were measured using Seahorse XF technology. The changes in mitochondrial dynamics were observed through fluorescent imaging, while Western blot analysis was used to examine key autophagy and apoptosis markers. RESULTS LAH significantly inhibited mitochondrial complex I activity, inducing ATP depletion and a compensatory increase in glycolysis. This disruption caused mitochondrial fragmentation, a trigger for autophagy, as shown by increased LC3-II expression and mTOR suppression. Apoptosis was also confirmed through the cleavage of caspase-3, contributing to reduced cancer cell viability. CONCLUSIONS LAH's anticancer effects in CRC cells are driven by its disruption of mitochondrial function, triggering both autophagy and apoptosis. These findings highlight its potential as a therapeutic compound for further exploration in cancer treatment.
Collapse
Affiliation(s)
- Izamary Delgado-Waldo
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico
| | - Svetlana Dokudovskaya
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (S.D.); (Y.A.L.-B.)
| | - Yahir A. Loissell-Baltazar
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (S.D.); (Y.A.L.-B.)
| | - Eduardo Pérez-Arteaga
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
| | - Jossimar Coronel-Hernández
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
| | - Mariano Martínez-Vázquez
- Instituto de Química, Universidad Nacional Autónoma de México, C. Exterior, C. Universitaria, Coyoacán, Ciudad de México 04510, Mexico;
| | - Eloy Andrés Pérez-Yépez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
| | - Alejandro Lopez-Saavedra
- Advanced Microscopy Applications Unit (ADMIRA), Instituto Nacional de Cancerología, San Fernando 22. Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey Ciudad de Mexico, C. Puente #222, Coapa, Arboledas del Sur, Tlalpan, Ciudad de Mexico 14380, Mexico
| | - Nadia Jacobo-Herrera
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
| | - Carlos Pérez Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México 54090, Mexico
| |
Collapse
|
2
|
Figueira MI, Carvalho TMA, Macário-Monteiro J, Cardoso HJ, Correia S, Vaz CV, Duarte AP, Socorro S. The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens. Biomedicines 2024; 12:1636. [PMID: 39200101 PMCID: PMC11351860 DOI: 10.3390/biomedicines12081636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
The role of estrogens in prostate cancer (PCa) is shrouded in mystery, with its actions going from angelic to devilish. The findings by Huggins and Hodges establishing PCa as a hormone-sensitive cancer have provided the basis for using estrogens in therapy. However, despite the clinical efficacy in suppressing tumor growth and the panoply of experimental evidence describing its anticarcinogenic effects, estrogens were abolished from PCa treatment because of the adverse secondary effects. Notwithstanding, research work over the years has continued investigating the effects of estrogens, reporting their pros and cons in prostate carcinogenesis. In contrast with the beneficial therapeutic effects, many reports have implicated estrogens in the disruption of prostate cell fate and tissue homeostasis. On the other hand, epidemiological data demonstrating the lower incidence of PCa in Eastern countries associated with a higher consumption of phytoestrogens support the beneficial role of estrogens in counteracting cancer development. Many studies have investigated the effects of phytoestrogens and the underlying mechanisms of action, which may contribute to developing safe estrogen-based anti-PCa therapies. This review compiles the existing data on the anti- and protumorigenic actions of estrogens and summarizes the anticancer effects of several phytoestrogens, highlighting their promising features in PCa treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sílvia Socorro
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (M.I.F.)
| |
Collapse
|
3
|
Zhao X, Luo T, Qiu Y, Yang Z, Wang D, Wang Z, Zeng J, Bi Z. Mechanisms of traditional Chinese medicine overcoming of radiotherapy resistance in breast cancer. Front Oncol 2024; 14:1388750. [PMID: 38993643 PMCID: PMC11237312 DOI: 10.3389/fonc.2024.1388750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/30/2024] [Indexed: 07/13/2024] Open
Abstract
Breast cancer stands as the most prevalent malignancy among women, with radiotherapy serving as a primary treatment modality. Despite radiotherapy, a subset of breast cancer patients experiences local recurrence, attributed to the intrinsic resistance of tumors to radiation. Therefore, there is a compelling need to explore novel approaches that can enhance cytotoxic effects through alternative mechanisms. Traditional Chinese Medicine (TCM) and its active constituents exhibit diverse pharmacological actions, including anti-tumor effects, offering extensive possibilities to identify effective components capable of overcoming radiotherapy resistance. This review delineates the mechanisms underlying radiotherapy resistance in breast cancer, along with potential candidate Chinese herbal medicines that may sensitize breast cancer cells to radiotherapy. The exploration of such herbal interventions holds promise for improving therapeutic outcomes in the context of breast cancer radiotherapy resistance.
Collapse
Affiliation(s)
- Xiaohui Zhao
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Oncology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-Sen University, Shanwei, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ting Luo
- Department of Oncology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-Sen University, Shanwei, China
| | - Yuting Qiu
- Department of Oncology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-Sen University, Shanwei, China
| | - Zhiwei Yang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Danni Wang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zairui Wang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiale Zeng
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuofei Bi
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Kim TW. Paeoniflorin Induces ER Stress-Mediated Apoptotic Cell Death by Generating Nox4-Derived ROS under Radiation in Gastric Cancer. Nutrients 2023; 15:5092. [PMID: 38140352 PMCID: PMC10745742 DOI: 10.3390/nu15245092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Gastric cancer is one of the most prevalent cancer types worldwide, and its resistance to cancer therapies, such as chemotherapy and radiotherapy, has made treating it a major challenge. Paeoniflorin (PF) is one potential pharmacological treatment derived from paeony root. However, in cancer, the molecular mechanisms and biological functions of PF are still unclear. In the present study, we found that PF exerts anti-tumor effects in vivo and in vitro and induces apoptotic cell death through ER stress, calcium (Ca2+), and reactive oxygen species (ROS) release in gastric cancer cells. However, ROS inhibition by DPI and NAC blocks cell death and the PERK signaling pathway via the reduction of Nox4. Moreover, PF triggers a synergistic inhibitory effect of the epithelial-mesenchymal transition (EMT) process under radiation exposure in radiation-resistant gastric cancer cells. These findings indicate that PF-induced Ca2+ and ROS release overcomes radioresistance via ER stress and induces cell death under radiation in gastric cancer cells. Therefore, PF, in combination with radiation, may be a powerful strategy for gastric cancer therapy.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, Gyeongju 38066, Republic of Korea
| |
Collapse
|
5
|
Wu SX, Xiong RG, Huang SY, Zhou DD, Saimaiti A, Zhao CN, Shang A, Zhang YJ, Gan RY, Li HB. Effects and mechanisms of resveratrol for prevention and management of cancers: An updated review. Crit Rev Food Sci Nutr 2023; 63:12422-12440. [PMID: 35852215 DOI: 10.1080/10408398.2022.2101428] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer is a severe public health problem. Resveratrol is a famous natural compound that has various bioactivities, such as antioxidant, anti-inflammatory, antidiabetic and antiaging activities. Especially, resveratrol could prevent and treat various cancers, such as oral, thyroid, breast, lung, liver, pancreatic, gastric, colorectal, bladder, prostate and ovarian cancers. The underlying mechanisms have been widely studied, such as inhibiting cell proliferation, suppressing metastasis, inducing apoptosis, stimulating autophagy, modulating immune system, attenuating inflammation, regulating gut microbiota and enhancing effects of other anticancer drugs. In this review, we summarize effects and mechanisms of resveratrol on different cancers. This paper is helpful to develop resveratrol, crude extract containing resveratrol, or foods containing resveratrol into functional food, dietary supplements or auxiliary agents for prevention and management of cancers.
Collapse
Affiliation(s)
- Si-Xia Wu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Ruo-Gu Xiong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Si-Yu Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Dan-Dan Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Adila Saimaiti
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Cai-Ning Zhao
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ao Shang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yun-Jian Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ren-You Gan
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
6
|
Komorowska D, Radzik T, Kalenik S, Rodacka A. Natural Radiosensitizers in Radiotherapy: Cancer Treatment by Combining Ionizing Radiation with Resveratrol. Int J Mol Sci 2022; 23:ijms231810627. [PMID: 36142554 PMCID: PMC9501384 DOI: 10.3390/ijms231810627] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Conventional cancer treatment is mainly based on the surgical removal of the tumor followed by radiotherapy and/or chemotherapy. When surgical removal is not possible, radiotherapy and, less often, chemotherapy is the only way to treat patients. However, despite significant progress in understanding the molecular mechanisms of carcinogenesis and developments in modern radiotherapy techniques, radiotherapy (alone or in combination) does not always guarantee treatment success. One of the main causes is the radioresistance of cancer cells. Increasing the radiosensitivity of cancer cells improves the processes leading to their elimination during radiotherapy and prolonging the survival of cancer patients. In order to enhance the effect of radiotherapy in the treatment of radioresistant neoplasms, radiosensitizers are used. In clinical practice, synthetic radiosensitizers are commonly applied, but scientists have recently focused on using natural products (phytocompounds) as adjuvants in radiotherapy. In this review article, we only discuss naturally occurring radiosensitizers currently in clinical trials (paclitaxel, curcumin, genistein, and papaverine) and those whose radiation sensitizing effects, such as resveratrol, have been repeatedly confirmed by many independent studies.
Collapse
Affiliation(s)
- Dominika Komorowska
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
| | - Tomasz Radzik
- MARINEX International, 4 Placowa St., 93-446 Lodz, Poland
| | - Sebastian Kalenik
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
| | - Aleksandra Rodacka
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
- Correspondence: ; Fax: +48-426354473
| |
Collapse
|
7
|
Nisar S, Masoodi T, Prabhu KS, Kuttikrishnan S, Zarif L, Khatoon S, Ali S, Uddin S, Akil AAS, Singh M, Macha MA, Bhat AA. Natural products as chemo-radiation therapy sensitizers in cancers. Biomed Pharmacother 2022; 154:113610. [PMID: 36030591 DOI: 10.1016/j.biopha.2022.113610] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/02/2022] Open
Abstract
Cancer is a devastating disease and is the second leading cause of death worldwide. Surgery, chemotherapy (CT), and/or radiation therapy (RT) are the treatment of choice for most advanced tumors. Unfortunately, treatment failure due to intrinsic and acquired resistance to the current CT and RT is a significant challenge associated with poor patient prognosis. There is an urgent need to develop and identify agents that can sensitize tumor cells to chemo-radiation therapy (CRT) with minimal cytotoxicity to the healthy tissues. While many recent studies have identified the underlying molecular mechanisms and therapeutic targets for CRT failure, using small molecule inhibitors to chemo/radio sensitize tumors is associated with high toxicity and increased morbidity. Natural products have long been used as chemopreventive agents in many cancers. Combining many of these compounds with the standard chemotherapeutic agents or with RT has shown synergistic effects on cancer cell death and overall improvement in patient survival. Based on the available data, there is strong evidence that natural products have a robust therapeutic potential along with CRT and their well-known chemopreventive effects in many solid tumors. This review article reports updated literature on different natural products used as CT or RT sensitizers in many solid tumors. This is the first review discussing CT and RT sensitizers together in cancer.
Collapse
Affiliation(s)
- Sabah Nisar
- Depertment of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Tariq Masoodi
- Laboratory of Cancer immunology and genetics, Sidra Medicine, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Qatar
| | - Lubna Zarif
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Qatar
| | - Summaiya Khatoon
- Depertment of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Shahid Ali
- International Potato Center (CIP), Shillong, Meghalaya, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Ammira Al-Shabeeb Akil
- Depertment of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Mayank Singh
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, AIIMS, New Delhi, India.
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu & Kashmir, India.
| | - Ajaz A Bhat
- Depertment of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
8
|
Anjaly K, Tiku AB. Caffeic acid phenethyl ester induces radiosensitization via inhibition of DNA damage repair in androgen-independent prostate cancer cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:995-1006. [PMID: 35006630 DOI: 10.1002/tox.23459] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
In the present study, we evaluated the radiomodulatory potential of caffeic acid phenethyl ester (CAPE), an active component of traditional herbal medicine propolis. CAPE has been identified as a potent anticancer agent in multiple cancer types and is reported to have the dual role of radioprotection and radiosensitization. However, the radiomodulatory potential of CAPE in prostate cancer (PCa), which eventually becomes radioresistant is not known. Therefore, we studied the effect of co-treatment of CAPE and gamma radiation on androgen-independent DU145 and PC3 cells. The combination treatment sensitized PCa cells to radiation in a dose-dependent manner. The radiosensitizing effect of CAPE was observed in both cell lines. CAPE enhanced the level of ionizing radiation (IR)-induced gamma H2AX foci and cell death by apoptosis. The combination treatment also decreased the migration potential of PCa cells. This was confirmed by increased expression of E-cadherin and decrease in vimentin expression. CAPE sensitized PCa cells to radiation in vitro and induced apoptosis, augmented phosphorylation of Akt/mTOR, and hampered cell migration. At the mechanistic level, co-treatment of CAPE and IR inhibited cell growth by decreasing RAD50 and RAD51 proteins involved in DNA repair. This resulted in enhanced DNA damage and cell death. CAPE might represent a promising new adjuvant for the treatment of hormone-refractory radioresistant PCa.
Collapse
Affiliation(s)
- Km Anjaly
- Radiation and Cancer Therapeutics Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Ashu Bhan Tiku
- Radiation and Cancer Therapeutics Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
9
|
Akter R, Najda A, Rahman MH, Shah M, Wesołowska S, Hassan SSU, Mubin S, Bibi P, Saeeda S. Potential Role of Natural Products to Combat Radiotherapy and Their Future Perspectives. Molecules 2021; 26:5997. [PMID: 34641542 PMCID: PMC8512367 DOI: 10.3390/molecules26195997] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is the second leading cause of death in the world. Chemotherapy and radiotherapy (RT) are the common cancer treatments. In addition to these limitations, the development of adverse effects from chemotherapy and RT reduces the quality of life for cancer patients. Cellular radiosensitivity, or the ability to resist and overcome cell damage caused by ionizing radiation (IR), is directly related to cancer cells' response to RT. Therefore, radiobiological research is emphasizing chemical compounds 'radiosensitization of cancer cells so that they are more reactive in the IR spectrum. Recent years researchers have seen an increase in interest in natural products that have antitumor effects with minimal side effects. Natural products, on the other hand, are easy to recover and therefore less expensive. There have been several scientific studies done based on these compounds that have tested their ability in vitro and in vivo to induce tumor radiosensitization. The role of natural products in RT, as well as their usefulness and potential applications, is the goal of this current review.
Collapse
Affiliation(s)
- Rokeya Akter
- Department of Pharmacy, Jagannath University, Dhaka 1100, Bangladesh;
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Gangwon-do, Wonju 26426, Korea
| | - Agnieszka Najda
- Department of Vegetable and Herbal Crops, University of Life Sciences in Lublin, 50A Doświadczalna Street, 20-280 Lublin, Poland
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Gangwon-do, Wonju 26426, Korea
- Department of Pharmacy, Southeast University, Banani Street, Dhaka 1213, Bangladesh
| | - Muddaser Shah
- Department of Botany, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (P.B.); (S.S.)
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Oman
| | - Sylwia Wesołowska
- Institute of Soil Science and Environment Shaping, University of Life Sciences in Lublin, 7 Leszczyńskiego Street, 20-069 Lublin, Poland;
| | - Syed Shams ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai JiaoTong University, Shanghai 200240, China;
| | - Sidra Mubin
- Department of Botany, Hazara University Mansehra, Mansehra 21310, Pakistan;
| | - Parveen Bibi
- Department of Botany, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (P.B.); (S.S.)
| | - Saeeda Saeeda
- Department of Botany, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; (P.B.); (S.S.)
| |
Collapse
|
10
|
Lele W, Lei L, Liting Q. Resveratrol sensitizes A549 cells to irradiation damage via suppression of store-operated calcium entry with Orai1 and STIM1 downregulation. Exp Ther Med 2021; 21:587. [PMID: 33850559 PMCID: PMC8027717 DOI: 10.3892/etm.2021.10019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 11/26/2020] [Indexed: 01/04/2023] Open
Abstract
Resveratrol is a natural polyphenol with multiple positive biofunctions and was found to have potential as a radiosensitizer with an intricate molecular mechanism. Store-operated calcium entry (SOCE) is a novel intracellular calcium regulatory pattern that is mainly mediated by iron channels, such as by the stromal interaction molecule (STIM) and calcium release-activated calcium channel protein (Orai) families. SOCE was recently reported to be suppressed via the downregulation of STIM or Orai families for the promotion of tumor cell death induced by resveratrol. In the present study, resveratrol combined with irradiation treatment were found to induce more evident cell damage compared with irradiation treatment alone, as shown with Cell Counting Kit-8 assay and mitochondrial membrane potential detection with rhodamine 123. Additionally, resveratrol combined with irradiation treatment decreased the expression of STIM1 and Orai1, while it had no effects on STIM2, Orai2 and Orai3. Moreover, resveratrol combined with irradiation treatment lead to alleviated thapsigargin-induced SOCE. In addition, overexpression of STIM1 and Orai1 reversed resveratrol-induced SOCE inhibition and reduced death in A549 cells under irradiation. In summary, the present results revealed that resveratrol can significantly enhance the effect of irradiation damage on lung adenocarcinoma A549 cells, and this effect may be mediated by suppression of SOCE with reduced expression of both STIM1 and Orai1.
Collapse
Affiliation(s)
- Wu Lele
- Department of General Medicine, First People's Hospital of Yuhang, Hangzhou, Zhejiang 311100, P.R. China.,Department of Radiotherapy, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China
| | - Lv Lei
- Department of Radiotherapy, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China.,Epigenetic Laboratory, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China
| | - Qian Liting
- Department of Radiotherapy, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China.,Epigenetic Laboratory, Anhui Provincial Hospital, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
11
|
CYP1B1 as a therapeutic target in cardio-oncology. Clin Sci (Lond) 2021; 134:2897-2927. [PMID: 33185690 PMCID: PMC7672255 DOI: 10.1042/cs20200310] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/12/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular complications have been frequently reported in cancer patients and survivors, mainly because of various cardiotoxic cancer treatments. Despite the known cardiovascular toxic effects of these treatments, they are still clinically used because of their effectiveness as anti-cancer agents. In this review, we discuss the growing body of evidence suggesting that inhibition of the cytochrome P450 1B1 enzyme (CYP1B1) can be a promising therapeutic strategy that has the potential to prevent cancer treatment-induced cardiovascular complications without reducing their anti-cancer effects. CYP1B1 is an extrahepatic enzyme that is expressed in cardiovascular tissues and overexpressed in different types of cancers. A growing body of evidence is demonstrating a detrimental role of CYP1B1 in both cardiovascular diseases and cancer, via perturbed metabolism of endogenous compounds, production of carcinogenic metabolites, DNA adduct formation, and generation of reactive oxygen species (ROS). Several chemotherapeutic agents have been shown to induce CYP1B1 in cardiovascular and cancer cells, possibly via activating the Aryl hydrocarbon Receptor (AhR), ROS generation, and inflammatory cytokines. Induction of CYP1B1 is detrimental in many ways. First, it can induce or exacerbate cancer treatment-induced cardiovascular complications. Second, it may lead to significant chemo/radio-resistance, undermining both the safety and effectiveness of cancer treatments. Therefore, numerous preclinical studies demonstrate that inhibition of CYP1B1 protects against chemotherapy-induced cardiotoxicity and prevents chemo- and radio-resistance. Most of these studies have utilized phytochemicals to inhibit CYP1B1. Since phytochemicals have multiple targets, future studies are needed to discern the specific contribution of CYP1B1 to the cardioprotective and chemo/radio-sensitizing effects of these phytochemicals.
Collapse
|
12
|
Yi J, Zhu J, Zhao C, Kang Q, Zhang X, Suo K, Cao N, Hao L, Lu J. Potential of natural products as radioprotectors and radiosensitizers: opportunities and challenges. Food Funct 2021; 12:5204-5218. [PMID: 34018510 DOI: 10.1039/d1fo00525a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Natural products can be used as natural radiosensitizers and radioprotectors, showing promising effects in cancer treatments in combination with radiotherapy, while reducing ionizing radiation (IR) damage to normal cells/tissues. The different effects of natural products on irradiated normal and tumor cells/tissues have attracted more and more researchers' interest. Nonetheless, the clinical applications of natural products in radiotherapy are few, which may be related to their low bioavailability in the human body. Here, we displayed the radiation protection and radiation sensitization of major natural products, highlighted the related molecular mechanisms of these bioactive substances combined with radiotherapy to treat cancer, and critically reviewed their deficiency and improved measures. Lastly, several clinical trials were presented to verify the clinical application of natural products as radiosensitizers and radioprotectors. Further clinical evaluation is still needed. This review provides a reference for the utilization of natural products as radiosensitizers and radioprotectors.
Collapse
Affiliation(s)
- Juanjuan Yi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Jiaqing Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Changcheng Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Qiaozhen Kang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiaomiao Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Keke Suo
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Nana Cao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Limin Hao
- Institute of Quartermaster Engineering and Technology, Academy of Military Sciences PLA China, Beijing, 100010, China.
| | - Jike Lu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
13
|
Lagunas-Rangel FA, Bermúdez-Cruz RM. Natural Compounds That Target DNA Repair Pathways and Their Therapeutic Potential to Counteract Cancer Cells. Front Oncol 2020; 10:598174. [PMID: 33330091 PMCID: PMC7710985 DOI: 10.3389/fonc.2020.598174] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/23/2020] [Indexed: 12/19/2022] Open
Abstract
Resistance to current cancer treatments is an important problem that arises through various mechanisms, but one that stands out involves an overexpression of several factors associated with DNA repair. To counteract this type of resistance, different drugs have been developed to affect one or more DNA repair pathways, therefore, to test different compounds of natural origin that have been shown to induce cell death in cancer cells is paramount. Since natural compounds target components of the DNA repair pathways, they have been shown to promote cancer cells to be resensitized to current treatments. For this and other reasons, natural compounds have aroused great curiosity and several research projects are being developed around the world to establish combined treatments between them and radio or chemotherapy. In this work, we summarize the effects of different natural compounds on the DNA repair mechanisms of cancer cells and emphasize their possible application to re-sensitize these cells.
Collapse
Affiliation(s)
- Francisco Alejandro Lagunas-Rangel
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico
| | - Rosa María Bermúdez-Cruz
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
14
|
Chen YW, Huang MZ, Chen CL, Kuo CY, Yang CY, Chiang-Ni C, Chen YYM, Hsieh CM, Wu HY, Kuo ML, Chiu CH, Lai CH. PM 2.5 impairs macrophage functions to exacerbate pneumococcus-induced pulmonary pathogenesis. Part Fibre Toxicol 2020; 17:37. [PMID: 32753046 PMCID: PMC7409448 DOI: 10.1186/s12989-020-00362-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
Background Pneumococcus is one of the most common human airway pathogens that causes life-threatening infections. Ambient fine particulate matter (PM) with aerodynamic diameter ≤ 2.5 μm (PM2.5) is known to significantly contribute to respiratory diseases. PM2.5-induced airway inflammation may decrease innate immune defenses against bacterial infection. However, there is currently limited information available regarding the effect of PM2.5 exposure on molecular interactions between pneumococcus and macrophages. Results PM2.5 exposure hampered macrophage functions, including phagocytosis and proinflammatory cytokine production, in response to pneumococcal infection. In a PM2.5-exposed pneumococcus-infected mouse model, PM2.5 subverted the pulmonary immune response and caused leukocyte infiltration. Further, PM2.5 exposure suppressed the levels of CXCL10 and its receptor, CXCR3, by inhibiting the PI3K/Akt and MAPK pathways. Conclusions The effect of PM2.5 exposure on macrophage activity enhances pneumococcal infectivity and aggravates pulmonary pathogenesis.
Collapse
Affiliation(s)
- Yu-Wen Chen
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Mei-Zi Huang
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chyi-Liang Chen
- Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chieh-Ying Kuo
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Yu Yang
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chuan Chiang-Ni
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Yi-Ywan M Chen
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chia-Ming Hsieh
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Yu Wu
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Ling Kuo
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Cheng-Hsun Chiu
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Ho Lai
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan. .,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan. .,Department of Nursing, Asia University, Taichung, Taiwan.
| |
Collapse
|
15
|
Lou BS, Hsieh JH, Chen CM, Hou CW, Wu HY, Chou PY, Lai CH, Lee JW. Helium/Argon-Generated Cold Atmospheric Plasma Facilitates Cutaneous Wound Healing. Front Bioeng Biotechnol 2020; 8:683. [PMID: 32695763 PMCID: PMC7338308 DOI: 10.3389/fbioe.2020.00683] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022] Open
Abstract
Cold atmospheric plasma jet (CAPJ) or non-thermal plasma jet has been employed in various biomedical applications based on their functions in bactericidal activity and wound healing. However, the effect of CAPJ generated by a particular composition of gases on wound closure and the underlying mechanisms that regulate wound healing signals remain elusive. In the present study, we investigated the impact of helium (He)- or a gas mixture of He and argon (He/Ar)-generated CAPJ on cell proliferation, which is a pivotal step during the wound healing process. With careful treatment duration control, He/Ar-CAPJ effectively induced keratinocyte proliferation and migration mediated through the activation of epithelial-to-mesenchymal transition (EMT) and cell cycle progression, which was evidenced by a decrease in E-cadherin levels and increases in N-cadherin, cyclin D1, Ki-67, Cdk2, and p-ERK levels. Rat wound healing studies showed that He/Ar-CAPJ treatment facilitated granulation tissue formation and mitigated inflammation in cutaneous tissue, resulting in accelerated wound closure. These findings highlight the possibility that He/Ar-CAPJ can be developed as a therapeutic agent for enhancing wound healing.
Collapse
Affiliation(s)
- Bih-Show Lou
- Chemistry Division, Center for General Education, Chang Gung University, Taoyuan, Taiwan.,Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jang-Hsing Hsieh
- Center for Plasma and Thin Film Technologies, Ming Chi University of Technology, New Taipei, Taiwan.,Department of Materials Engineering, Ming Chi University of Technology, New Taipei, Taiwan
| | - Chun-Ming Chen
- Center for Plasma and Thin Film Technologies, Ming Chi University of Technology, New Taipei, Taiwan
| | - Chun-Wei Hou
- Chemistry Division, Center for General Education, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Yu Wu
- Department of Microbiology and Immunology, College of Medicine, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Pang-Yun Chou
- Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, College of Medicine, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| | - Jyh-Wei Lee
- Center for Plasma and Thin Film Technologies, Ming Chi University of Technology, New Taipei, Taiwan.,Department of Materials Engineering, Ming Chi University of Technology, New Taipei, Taiwan.,Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Mechanical Engineering, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
16
|
Adhikari K, Lo IW, Chen CL, Wang YL, Lin KH, Zadeh SM, Rattinam R, Li YS, Wu CJ, Li TL. Chemoenzymatic Synthesis and Biological Evaluation for Bioactive Molecules Derived from Bacterial Benzoyl Coenzyme A Ligase and Plant Type III Polyketide Synthase. Biomolecules 2020; 10:biom10050738. [PMID: 32397467 PMCID: PMC7277991 DOI: 10.3390/biom10050738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 11/22/2022] Open
Abstract
Plant type III polyketide synthases produce diverse bioactive molecules with a great medicinal significance to human diseases. Here, we demonstrated versatility of a stilbene synthase (STS) from Pinus Sylvestris, which can accept various non-physiological substrates to form unnatural polyketide products. Three enzymes (4-coumarate CoA ligase, malonyl-CoA synthetase and engineered benzoate CoA ligase) along with synthetic chemistry was practiced to synthesize starter and extender substrates for STS. Of these, the crystal structures of benzoate CoA ligase (BadA) from Rhodopseudomonas palustris in an apo form or in complex with a 2-chloro-1,3-thiazole-5-carboxyl-AMP or 2-methylthiazole-5-carboxyl-AMP intermediate were determined at resolutions of 1.57 Å, 1.7 Å, and 2.13 Å, respectively, which reinforces its capacity in production of unusual CoA starters. STS exhibits broad substrate promiscuity effectively affording structurally diverse polyketide products. Seven novel products showed desired cytotoxicity against a panel of cancer cell lines (A549, HCT116, Cal27). With the treatment of two selected compounds, the cancer cells underwent cell apoptosis in a dose-dependent manner. The precursor-directed biosynthesis alongside structure-guided enzyme engineering greatly expands the pharmaceutical repertoire of lead compounds with promising/enhanced biological activities.
Collapse
Affiliation(s)
- Kamal Adhikari
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica and National Chung Hsing University, Taipei 11529, Taiwan
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan
| | - I-Wen Lo
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
| | - Chun-Liang Chen
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
| | - Yung-Lin Wang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
| | - Kuan-Hung Lin
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
| | - Saeid Malek Zadeh
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Taipei 11529, Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Rajesh Rattinam
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Taipei 11529, Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yi-Shan Li
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
| | - Chang-Jer Wu
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan;
| | - Tsung-Lin Li
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan; (K.A.); (I-W.L.); (C.-L.C.); (Y.-L.W.); (K.-H.L.); (S.M.Z.); (R.R.); (Y.-S.L.)
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica and National Chung Hsing University, Taipei 11529, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Taipei 11529, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
- Correspondence: ; Tel.: +886-22787-1235
| |
Collapse
|
17
|
Seifert M, Peitzsch C, Gorodetska I, Börner C, Klink B, Dubrovska A. Network-based analysis of prostate cancer cell lines reveals novel marker gene candidates associated with radioresistance and patient relapse. PLoS Comput Biol 2019; 15:e1007460. [PMID: 31682594 PMCID: PMC6855562 DOI: 10.1371/journal.pcbi.1007460] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 11/14/2019] [Accepted: 10/05/2019] [Indexed: 12/20/2022] Open
Abstract
Radiation therapy is an important and effective treatment option for prostate cancer, but high-risk patients are prone to relapse due to radioresistance of cancer cells. Molecular mechanisms that contribute to radioresistance are not fully understood. Novel computational strategies are needed to identify radioresistance driver genes from hundreds of gene copy number alterations. We developed a network-based approach based on lasso regression in combination with network propagation for the analysis of prostate cancer cell lines with acquired radioresistance to identify clinically relevant marker genes associated with radioresistance in prostate cancer patients. We analyzed established radioresistant cell lines of the prostate cancer cell lines DU145 and LNCaP and compared their gene copy number and expression profiles to their radiosensitive parental cells. We found that radioresistant DU145 showed much more gene copy number alterations than LNCaP and their gene expression profiles were highly cell line specific. We learned a genome-wide prostate cancer-specific gene regulatory network and quantified impacts of differentially expressed genes with directly underlying copy number alterations on known radioresistance marker genes. This revealed several potential driver candidates involved in the regulation of cancer-relevant processes. Importantly, we found that ten driver candidates from DU145 (ADAMTS9, AKR1B10, CXXC5, FST, FOXL1, GRPR, ITGA2, SOX17, STARD4, VGF) and four from LNCaP (FHL5, LYPLAL1, PAK7, TDRD6) were able to distinguish irradiated prostate cancer patients into early and late relapse groups. Moreover, in-depth in vitro validations for VGF (Neurosecretory protein VGF) showed that siRNA-mediated gene silencing increased the radiosensitivity of DU145 and LNCaP cells. Our computational approach enabled to predict novel radioresistance driver gene candidates. Additional preclinical and clinical studies are required to further validate the role of VGF and other candidate genes as potential biomarkers for the prediction of radiotherapy responses and as potential targets for radiosensitization of prostate cancer. Prostate cancer cell lines represent an important model system to characterize molecular alterations that contribute to radioresistance, but irradiation can cause deletions and amplifications of DNA segments that affect hundreds of genes. This in combination with the small number of cell lines that are usually considered does not allow a straight-forward identification of driver genes by standard statistical methods. Therefore, we developed a network-based approach to analyze gene copy number and expression profiles of such cell lines enabling to identify potential driver genes associated with radioresistance of prostate cancer. We used lasso regression in combination with a significance test for lasso to learn a genome-wide prostate cancer-specific gene regulatory network. We used this network for network flow computations to determine impacts of gene copy number alterations on known radioresistance marker genes. Mapping to prostate cancer samples and additional filtering allowed us to identify 14 driver gene candidates that distinguished irradiated prostate cancer patients into early and late relapse groups. In-depth literature analysis and wet-lab validations suggest that our method can predict novel radioresistance driver genes. Additional preclinical and clinical studies are required to further validate these genes for the prediction of radiotherapy responses and as potential targets to radiosensitize prostate cancer.
Collapse
Affiliation(s)
- Michael Seifert
- Institute for Medical Informatics and Biometry (IMB), Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- * E-mail:
| | - Claudia Peitzsch
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Ielizaveta Gorodetska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Caroline Börner
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Barbara Klink
- Institute for Clinical Genetics, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anna Dubrovska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology-OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK) Partner Site Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
18
|
Calvaruso M, Pucci G, Musso R, Bravatà V, Cammarata FP, Russo G, Forte GI, Minafra L. Nutraceutical Compounds as Sensitizers for Cancer Treatment in Radiation Therapy. Int J Mol Sci 2019; 20:ijms20215267. [PMID: 31652849 PMCID: PMC6861933 DOI: 10.3390/ijms20215267] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 02/05/2023] Open
Abstract
The improvement of diagnostic techniques and the efficacy of new therapies in clinical practice have allowed cancer patients to reach a higher chance to be cured together with a better quality of life. However, tumors still represent the second leading cause of death worldwide. On the contrary, chemotherapy and radiotherapy (RT) still lack treatment plans which take into account the biological features of tumors and depend on this for their response to treatment. Tumor cells' response to RT is strictly-connected to their radiosensitivity, namely, their ability to resist and to overcome cell damage induced by ionizing radiation (IR). For this reason, radiobiological research is focusing on the ability of chemical compounds to radiosensitize cancer cells so to make them more responsive to IR. In recent years, the interests of researchers have been focused on natural compounds that show antitumoral effects with limited collateral issues. Moreover, nutraceuticals are easy to recover and are thus less expensive. On these bases, several scientific projects have aimed to test also their ability to induce tumor radiosensitization both in vitro and in vivo. The goal of this review is to describe what is known about the role of nutraceuticals in radiotherapy, their use and their potential application.
Collapse
Affiliation(s)
- Marco Calvaruso
- Istituto di Bioimmagini e Fisiologia Molecolare-Consiglio Nazionale delle Ricerche (IBFM-CNR), 90015 Cefalù (PA), Italy.
| | - Gaia Pucci
- Istituto di Bioimmagini e Fisiologia Molecolare-Consiglio Nazionale delle Ricerche (IBFM-CNR), 90015 Cefalù (PA), Italy.
| | - Rosa Musso
- Istituto di Bioimmagini e Fisiologia Molecolare-Consiglio Nazionale delle Ricerche (IBFM-CNR), 90015 Cefalù (PA), Italy.
| | - Valentina Bravatà
- Istituto di Bioimmagini e Fisiologia Molecolare-Consiglio Nazionale delle Ricerche (IBFM-CNR), 90015 Cefalù (PA), Italy.
| | - Francesco P Cammarata
- Istituto di Bioimmagini e Fisiologia Molecolare-Consiglio Nazionale delle Ricerche (IBFM-CNR), 90015 Cefalù (PA), Italy.
| | - Giorgio Russo
- Istituto di Bioimmagini e Fisiologia Molecolare-Consiglio Nazionale delle Ricerche (IBFM-CNR), 90015 Cefalù (PA), Italy.
| | - Giusi I Forte
- Istituto di Bioimmagini e Fisiologia Molecolare-Consiglio Nazionale delle Ricerche (IBFM-CNR), 90015 Cefalù (PA), Italy.
| | - Luigi Minafra
- Istituto di Bioimmagini e Fisiologia Molecolare-Consiglio Nazionale delle Ricerche (IBFM-CNR), 90015 Cefalù (PA), Italy.
| |
Collapse
|
19
|
Polyphenols: Major regulators of key components of DNA damage response in cancer. DNA Repair (Amst) 2019; 82:102679. [DOI: 10.1016/j.dnarep.2019.102679] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/27/2019] [Accepted: 07/27/2019] [Indexed: 02/06/2023]
|
20
|
Abdelgawad IY, Grant MKO, Zordoky BN. Leveraging the Cardio-Protective and Anticancer Properties of Resveratrol in Cardio-Oncology. Nutrients 2019; 11:nu11030627. [PMID: 30875799 PMCID: PMC6471701 DOI: 10.3390/nu11030627] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 12/25/2022] Open
Abstract
Cardio-oncology is a clinical/scientific discipline which aims to prevent and/or treat cardiovascular diseases in cancer patients. Although a large number of cancer treatments are known to cause cardiovascular toxicity, they are still widely used because they are highly effective. Unfortunately, therapeutic interventions to prevent and/or treat cancer treatment-induced cardiovascular toxicity have not been established yet. A major challenge for such interventions is to protect the cardiovascular system without compromising the therapeutic benefit of anticancer medications. Intriguingly, the polyphenolic natural compound resveratrol and its analogs have been shown in preclinical studies to protect against cancer treatment-induced cardiovascular toxicity. They have also been shown to possess significant anticancer properties on their own, and to enhance the anticancer effect of other cancer treatments. Thus, they hold significant promise to protect the cardiovascular system and fight the cancer at the same time. In this review, we will discuss the current knowledge regarding the cardio-protective and the anticancer properties of resveratrol and its analogs. Thereafter, we will discuss the challenges that face the clinical application of these agents. To conclude, we will highlight important gaps of knowledge and future research directions to accelerate the translation of these exciting preclinical findings to cancer patient care.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Marianne K O Grant
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
21
|
Abstract
Breast cancer is a common malignancy with poor prognosis. Cancer cells are heterogeneous and cancer stem cells (CSCs) are primarily responsible for tumor relapse, treatment-resistance and metastasis, so for breast cancer stem cells (BCSCs). Diets are known to be associated with carcinogenesis. Food-derived polyphenols are able to attenuate the formation and virulence of BCSCs, implying that these compounds and their analogs might be promising agents for preventing breast cancer. In the present review, we summarized the origin and surface markers of BCSCs and possible mechanisms responsible for the inhibitory effects of polyphenols on BCSCs. The suppressive effects of common dietary polyphenols against BCSCs, such as curcumin, epigallocatechin gallate (EGCG) and related polyphenolic compounds were further discussed.
Collapse
Affiliation(s)
- Hao-Feng Gu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xue-Ying Mao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| |
Collapse
|
22
|
Chen YA, Tzeng DTW, Huang YP, Lin CJ, Lo UG, Wu CL, Lin H, Hsieh JT, Tang CH, Lai CH. Antrocin Sensitizes Prostate Cancer Cells to Radiotherapy through Inhibiting PI3K/AKT and MAPK Signaling Pathways. Cancers (Basel) 2018; 11:cancers11010034. [PMID: 30602706 PMCID: PMC6356781 DOI: 10.3390/cancers11010034] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/20/2018] [Accepted: 12/25/2018] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is one of the most common treatment options for local or regional advanced prostate cancer (PCa). Importantly, PCa is prone to radioresistance and often develops into malignancies after long-term radiotherapy. Antrocin, a sesquiterpene lactone isolated from Antrodia cinnamomea, possesses pharmacological efficacy against various cancer types; however, its therapeutic potential requires comprehensive exploration, particularly in radioresistant PCa cells. In this study, we emphasized the effects of antrocin on radioresistant PCa cells and addressed the molecular mechanism underlying the radiosensitization induced by antrocin. Our results showed that a combination treatment with antrocin and ionizing radiation (IR) synergistically inhibited cell proliferation and induced apoptosis in radioresistant PCa cells. We further demonstrated that antrocin downregulated PI3K/AKT and MAPK signaling pathways as well as suppressed type 1 insulin-like growth factor 1 receptor (IGF-1R)-mediated induction of β-catenin to regulate cell cycle and apoptosis. Using xenograft mouse models, we showed that antrocin effectively enhanced radiotherapy in PCa. Our study demonstrates that antrocin sensitizes PCa to radiation through constitutive suppression of IGF-1R downstream signaling, revealing that it can be developed as a potent therapeutic agent to overcome radioresistant PCa.
Collapse
Affiliation(s)
- Yu-An Chen
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung 40402, Taiwan.
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
- Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - David T W Tzeng
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Yi-Ping Huang
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Chun-Jung Lin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Chia-Lin Wu
- Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Department of Neurology, Chang Gung Memorial Hospital, Linkou 33305, Taiwan.
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung 40402, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40202, Taiwan.
| | - Chih-Ho Lai
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung 40402, Taiwan.
- Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Department of Nursing, Asia University, Taichung 41354, Taiwan.
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou 33305, Taiwan.
| |
Collapse
|
23
|
Wang D, Gao Z, Zhang X. Resveratrol Induces Apoptosis in Murine Prostate Cancer Cells via Hypoxia-Inducible Factor 1-alpha (HIF-1α)/Reactive Oxygen Species (ROS)/P53 Signaling. Med Sci Monit 2018; 24:8970-8976. [PMID: 30531685 PMCID: PMC6298174 DOI: 10.12659/msm.913290] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Resveratrol, a polyphenol found on the surface of red fruits, is able to suppress many kinds of malignancies. Nevertheless, its mechanism of action is not yet clear. Consequently, this study aimed to elucidate its influence and explore the etiology of PCCs (prostate cancer cells). MATERIAL AND METHODS The proliferation of prostate cancer cells was determined by CCK-8 assay. Cell apoptosis was determined by Hoechst staining FC assay. Cell migration was detected by scratch test. The levels of apoptosis-related protein were detected by Western blot analysis. RESULTS It was discovered that resveratrol suppresses cellular survival and migration and enhances cell death. In addition, it was revealed that resveratrol elevated ROS concentration and expression of biomarker of cell death Bax, while inhibiting Bcl2, an anti-apoptotic protein, and reinforcing expression of p53. Moreover, resveratrol remarkably increased the expressions of HIF-1α and p53 in PC cells. Resveratrol suppressed cell survival and promoted cell death, but its effects were reversed after HIF-1α knockdown, suggesting that the effects of resveratrol in PC are mediated via HIF-1α. CONCLUSIONS Our findings indicate that resveratrol induces apoptosis via HIF-1α/ROS/p53 signaling in prostate cancer cells and may be a useful therapeutic agent against prostate cancer.
Collapse
Affiliation(s)
- Delong Wang
- Department of Urology Surgery, The Affiliated Zhongshan Hospital Dalian University, Dalian, Liaoning, China (mainland)
| | - Zefeng Gao
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Xin Zhang
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
24
|
Chen YA, Shih HW, Lin YC, Hsu HY, Wu TF, Tsai CH, Wu CL, Wu HY, Hsieh JT, Tang CH, Lai CH. Simvastatin Sensitizes Radioresistant Prostate Cancer Cells by Compromising DNA Double-Strand Break Repair. Front Pharmacol 2018; 9:600. [PMID: 29950990 PMCID: PMC6008406 DOI: 10.3389/fphar.2018.00600] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/18/2018] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent male cancers in western world. Radiation therapy (RT) is commonly used to treat PCa patients. However, a certain proportion of patients develop radioresistant PCa cells, which results in metastatic disease. Statins, which inhibit 3-hydroxy-3-methyl glutaryl coenzyme A (HMG-CoA) reductase, are commonly used to treat hypercholesterolemia, exhibiting beneficial effects on cardiovascular diseases and on several types of cancers, including PCa. However, the mechanistic details and crosstalk between statins and RT in PCa cells remain unknown. In this study, radioresistant DOC-2/DAB2 interactive protein (DAB2IP)-deficient PCa cells were used to evaluate whether simvastatin could enhance the effect of ionizing radiation (IR). The crucial molecules that associated with simvastatin elevated radiosensitivity in PCa cells were explored. Our results demonstrated that a combination treatment with simvastatin and IR synergistically induced apoptosis of radioresistant PCa cells. In addition, simvastatin appeared to compromise DNA double-strand breaks repair by activating the expressions of histone 2A family member X (γ-H2AX) and phospho-checkpoint kinase 1 (p-CHK1), suggesting an underlying mechanism for this radiosensitization of PCa cells. These findings reveal that simvastatin may be a potent therapeutic agent for co-treatment with radiation to overcome radioresistance in PCa cells.
Collapse
Affiliation(s)
- Yu-An Chen
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hua-Wei Shih
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Chun Lin
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hui-Ying Hsu
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | - Tsu-Fang Wu
- Department of Applied Cosmetology, Hung Kuang University, Taichung, Taiwan
| | - Chen-Han Tsai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hui-Yu Wu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Ho Lai
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
25
|
Rauf A, Imran M, Suleria HAR, Ahmad B, Peters DG, Mubarak MS. A comprehensive review of the health perspectives of resveratrol. Food Funct 2018; 8:4284-4305. [PMID: 29044265 DOI: 10.1039/c7fo01300k] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many natural products present in our diet, including flavonoids, can prevent the progression of cancer and other diseases. Resveratrol, a natural polyphenol present in various fruits and vegetables, plays an important role as a therapeutic and chemopreventive agent used in the treatment of various illnesses. It exhibits effects against different types of cancer through different pathways. It additionally exerts antidiabetic, anti-inflammatory, and anti-oxidant effects in a variety of cell types. Furthermore, the cardiovascular protective capacities of resveratrol are associated with multiple molecular targets and may lead to the development of novel therapeutic strategies for atherosclerosis, ischemia/reperfusion, metabolic syndrome, and heart failure. Accordingly, this article presents an overview of recent developments in the use of resveratrol for the prevention and treatment of different diseases along with various mechanisms. In addition, the present review summarizes the most recent literature pertaining to resveratrol as a chemotherapeutic agent against multiple diseases and provides an assessment of the potential of this natural compound as a complementary or alternative medicine.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar-23561, Khyber Pakhtunkhwa, Pakistan.
| | | | | | | | | | | |
Collapse
|
26
|
Su YW, Chen KM, Hassanshahi M, Tang Q, Howe PR, Xian CJ. Childhood cancer chemotherapy-induced bone damage: pathobiology and protective effects of resveratrol and other nutraceuticals. Ann N Y Acad Sci 2017; 1403:109-117. [PMID: 28662275 DOI: 10.1111/nyas.13380] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 11/30/2022]
Abstract
Intensive cancer chemotherapy causes significant bone loss, for which the mechanisms remain unclear and effective treatments are lacking. This is a significant issue particularly for childhood cancers, as the most common ones have a >75% cure rate following chemotherapy; there is an increasing population of survivors who live with chronic bone defects. Studies suggest that these defects are the result of reduced bone from increased marrow fat formation and increased bone resorption following chemotherapy. These changes probably result from altered expression/activation of regulatory molecules or pathways regulating skeletal cell formation and activity. Treatment with methotrexate, an antimetabolite commonly used in childhood oncology, has been shown to increase levels of proinflammatory/pro-osteoclastogenic cytokines (e.g., enhanced NF-κB activation), leading to increased osteoclast formation and bone resorption, as well as to attenuate Wnt signaling, leading to both decreased bone and increased marrow fat formation. In recent years, understanding the mechanisms of action and potential health benefits of selected nutraceuticals, including resveratrol, genistein, icariin, and inflammatory fatty acids, has led to preclinical studies that, in some cases, indicate efficacy in reducing chemotherapy-induced bone defects. We summarize the supporting evidence.
Collapse
Affiliation(s)
- Yu-Wen Su
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ke-Ming Chen
- Institute of Orthopaedics, Lanzhou General Hospital, Lanzhou Command of People's Liberation Army, Lanzhou, PR China
| | - Mohammadhossein Hassanshahi
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Qian Tang
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Peter R Howe
- Clinical Nutrition Research Centre, University of Newcastle, Callaghan, New South Wales, Australia
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
27
|
Lee AMC, Shandala T, Soo PP, Su YW, King TJ, Chen KM, Howe PR, Xian CJ. Effects of Resveratrol Supplementation on Methotrexate Chemotherapy-Induced Bone Loss. Nutrients 2017; 9:nu9030255. [PMID: 28282956 PMCID: PMC5372918 DOI: 10.3390/nu9030255] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/08/2017] [Indexed: 12/27/2022] Open
Abstract
Intensive cancer chemotherapy is known to cause bone defects, which currently lack treatments. This study investigated the effects of polyphenol resveratrol (RES) in preventing bone defects in rats caused by methotrexate (MTX), a commonly used antimetabolite in childhood oncology. Young rats received five daily MTX injections at 0.75 mg/kg/day. RES was orally gavaged daily for seven days prior to, and during, five-day MTX administration. MTX reduced growth plate thickness, primary spongiosa height, trabecular bone volume, increased marrow adipocyte density, and increased mRNA expression of the osteogenic, adipogenic, and osteoclastogenic factors in the tibial bone. RES at 10 mg/kg was found not to affect bone health in normal rats, but to aggravate the bone damage in MTX-treated rats. However, RES supplementation at 1 mg/kg preserved the growth plate, primary spongiosa, bone volume, and lowered the adipocyte density. It maintained expression of genes involved in osteogenesis and decreased expression of adipogenic and osteoclastogenic factors. RES suppressed osteoclast formation ex vivo of bone marrow cells from the treated rats. These data suggest that MTX can enhance osteoclast and adipocyte formation and cause bone loss, and that RES supplementation at 1 mg/kg may potentially prevent these bone defects.
Collapse
Affiliation(s)
- Alice M C Lee
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Tetyana Shandala
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Pei Pei Soo
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Yu-Wen Su
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Tristan J King
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Ke-Ming Chen
- Institute of Orthopaedics, Lanzhou General Hospital, Lanzhou Command of People's Liberation Army, Lanzhou 730050, China.
| | - Peter R Howe
- Clinical Nutrition Research Centre, University of Newcastle, Callaghan NSW 2308, Australia.
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| |
Collapse
|