1
|
Wei Y, Ni W, Zhao L, Gao Y, Zhou B, Feng Q, Ma Y, Wang L. Phillygenin Inhibits PI3K-Akt-mTOR Signalling Pathway to Prevent bleomycin-Induced Idiopathic Pulmonary Fibrosis in Mice. Clin Exp Pharmacol Physiol 2025; 52:e70017. [PMID: 39746665 DOI: 10.1111/1440-1681.70017] [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: 08/15/2024] [Revised: 11/15/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease characterised by irreversible lung structure and function. Phillygenin (PHI) is a lignan extracted from Forsythiae fructus with the activities of anti-inflammatory and antioxidant. This study aimed to explore the protective effect of PHI on IPF. The mouse model of IPF was established by bleomycin (BLM), and then treated with PHI. After 15 days of administration, the lung index was calculated. H&E staining, Masson staining and immunohistochemical methods were used to detect the effect of PHI on pulmonary fibrosis. MDA and SOD were tested to evaluate the effect of PHI on lung tissue oxidative stress. Western blot was used to detect the effect of PHI on the expressions of α-SMA, p-smad2, TGF- β1, Nrf2, HO-1 and NQO-1. Network pharmacology was used to identify the key signalling pathways for PHI to improve IPF, and Western blot was used to validate the result. The results showed that PHI prevented mice from BLM-induced IPF, manifested by reducing lung index, improving lung tissue pathological damage, inhibiting collagen deposition and expression of fibrosis markers including α-SMA, collagen1, p-smad2 and TGF-β1. PHI inhibited oxidative stress by upregulating the expressions of Nrf2, HO-1 and NQO-1. Network pharmacology revealed that PI3K-Akt-mTOR signalling pathway was the underlying target of PHI for IPF. Molecular docking indicated strong binding of PHI with PIK3CA, AKT1 and RELA. Western blot validated that PHI downregulated the PI3K-Akt-mTOR signalling pathway and stimulated autophagy. This study indicated that PHI prevented BLM-induced pulmonary fibrosis by inhibiting PI3K-Akt-mTOR signalling pathway.
Collapse
Affiliation(s)
- Yongjia Wei
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Wenting Ni
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lizhi Zhao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Yanhong Gao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Bing Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Qun Feng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Yun Ma
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Linyi, China
| | - Limin Wang
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
2
|
Moosavi F, Firoozi R, Tavakkoli M, Nazari S, Alipour A, Firuzi O. Combination of chemotherapy and c-MET inhibitors has synergistic effects in c-MET overexpressing pancreatic cancer cells. Biochimie 2024:S0300-9084(24)00299-2. [PMID: 39675659 DOI: 10.1016/j.biochi.2024.12.006] [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: 06/21/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains as one of the most lethal malignancies. c-MET is an important oncogenic kinase involved in PDAC progression. We determined the anticancer effect of c-MET inhibitors, crizotinib and cabozantinib, combined with chemotherapeutic agents, doxorubicin, oxaliplatin and gemcitabine, against different PDAC and a lung adenocarcinoma cell line expressing different levels of c-MET. MTT assay was performed to assess cell growth inhibition. Synergistic combinations were evaluated in spheroid cultures, while apoptosis was determined through Hoechst33258 staining. The effect of drug combinations on cell cycle and apoptosis induction was examined by RNase/PI flow cytometric assay. We also evaluated reactive oxygen species (ROS) levels using 2',7'-dichlorofluorescein-diacetate (DCFH-DA) assay to explore the possible mechanisms contributing to synergism. Combination of crizotinib or cabozantinib with doxorubicin exhibited synergistic effects in c-MET overexpressing cells. Conversely, combinations of c-MET inhibitors with other agents were additive or even antagonistic. Combination index (CI) values calculated with Calcusyn software were 0.631-0.730 for crizotinib and 0.542-0.746 for cabozantinib co-administered with doxorubicin. These synergistic combinations showed significant spheroid growth inhibition and apoptosis induction in Suit-2, c-MET dependent PDAC cells. These combinations also significantly increased the number of cells in both apoptotic sub-G1 phase and the G2/M phase compared to single-drug treatment. Increased ROS production seemed to be a possible mechanism underlying synergism. In conclusion, c-MET inhibitors synergize with DNA damaging agent, doxorubicin, in cancer cells with c-MET overexpression, indicating that these combination therapies may be a promising cancer therapeutic strategy.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roya Firoozi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marjan Tavakkoli
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Alipour
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
He C, Li Q, Wu W, Liu K, Li X, Zheng H, Lai Y. Ferroptosis-associated genes and compounds in renal cell carcinoma. Front Immunol 2024; 15:1473203. [PMID: 39399506 PMCID: PMC11466770 DOI: 10.3389/fimmu.2024.1473203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
As the main type of renal cell carcinoma (RCC), clear cell RCC (ccRCC) is often associated with the deletion or mutation of the von Hippel Lindau (VHL) gene, enhancement of glucose and lipid metabolism, and heterogeneity of the tumor microenvironment. VHL alterations in RCC cells lead to the activation of hypoxia-inducible factors and their downstream target vascular endothelial growth factor, and to the reprogramming of multiple cell death pathways and metabolic weakness, including ferroptosis, which are associated with targeted therapy or immunotherapy. The changes in biological metabolites (e.g., iron and lipids) support ferroptosis as a potential therapeutic strategy for RCC, while iron metabolism and ferroptosis regulation have been examined as anti-RCC agents in numerous studies, and various ferroptosis-related molecules have been shown to be related to the metastasis and prognosis of ccRCC. For example, glutathione peroxidase 4 and glutaminase inhibitors can inhibit pyrimidine synthesis and increase reactive oxygen species levels in VHL-deficient RCC cells. In addition, the release of damage-associated molecular patterns by tumor cells undergoing ferroptosis also mediates antitumor immunity, and immune therapy can synergize with targeted therapy or radiotherapy through ferroptosis. However, Inducing ferroptosis not only suppresses cancer, but also promotes cancer development due to its potential negative effects on anti-cancer immunity. Therefore, ferroptosis and various tumor microenviroment-related molecules may co-occur during the development and treatment of RCC, and further understanding of the interactions, core targets, and related drugs of ferroptosis may provide new combination drug strategies for RCC treatment. Here we summarize the key genes and compounds on ferroptosis and RCC in order to envision future treatment strategies and to provide sufficient information for overcoming RCC resistance through ferroptosis.
Collapse
Affiliation(s)
- Chengwu He
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Qingyi Li
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Weijia Wu
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ke Liu
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xingwen Li
- Tibet Future Biomedicine Company Limited, Golmud, Qinghai, China
| | - Hanxiong Zheng
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yongchang Lai
- Department of Pharmaceutical Management, School of Medical Business, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Zhao T, Fang R, Ding J, Liu Y, Cheng M, Zhou F, Liu F, Li W, Li S, Jiang K, Shi X, Liu M, Xu B, Zou X, Zhu H, Zhou L. Melatonin ameliorates multiorgan injuries induced by severe acute pancreatitis in mice by regulating the Nrf2 signaling pathway. Eur J Pharmacol 2024; 975:176646. [PMID: 38762157 DOI: 10.1016/j.ejphar.2024.176646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
Severe acute pancreatitis (SAP) is a complicated inflammatory reaction that impacts the pancreas, often resulting in damage to numerous organs. This disorder encompasses a range of processes such as inflammation, oxidative stress, and pancreatitis. The hormone melatonin (MT) is primarily secreted by the pineal gland and plays a crucial role in mitigating inflammation, countering the harmful effects of free radicals, and regulating oxidative stress. The aim of this research was to investigate the potential protective impact and the underlying mechanism of melatonin in mice afflicted with SAP. The biochemical and histological assessments unequivocally demonstrated that melatonin effectively inhibited necrosis, infiltration, edema and cell death in pancreatic tissues, thereby suppressing acute pancreatitis. Notably, melatonin also alleviated the consequent harm to distant organs, notably the lungs, liver, and kidneys. Furthermore, both preventive and therapeutic administration of melatonin prompted nuclear factor E2-related factor 2 (Nrf2) activation followed by Nrf2 target gene expression. Nrf2 initiates the activation of antioxidant genes, thereby providing defense against oxidative stress. Conversely, Nrf2 reduction may contribute to impaired antioxidant protection in SAP. The beneficial impact of Nrf2 on antioxidants was absent in Nrf2-knockout mice, leading to the accumulation of LDH and exacerbation of cell death. This deterioration in both pancreatitis and injuries in distant organs intensified significantly. The results indicate that melatonin has an enhanced ability to protect against multiorgan damage caused by SAP, which is accomplished through the increase in Nrf2 expression. Additionally, Nrf2 initiates the activation of antioxidant genes that offer defense against cell death.
Collapse
Affiliation(s)
- Tianming Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China; Department of Gastroenterology, Nanjing Drum Tower Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Jiangsu, Nanjing, 210008, China
| | - Rui Fang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China
| | - Jing Ding
- Department of Hyperbaric Oxygen, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210002, China
| | - Yu Liu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China
| | - Ming Cheng
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China
| | - Fan Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China
| | - Feng Liu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China
| | - Wenting Li
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China
| | - Shupei Li
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210002, China
| | - Kang Jiang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210002, China
| | - Xiaoxiao Shi
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210002, China
| | - Mingdong Liu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China
| | - Bing Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China.
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China.
| | - Hao Zhu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China.
| | - Lin Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, Nanjing, 210008, China.
| |
Collapse
|
5
|
Fakhrioliaei A, Tanhaei S, Pakmehr S, Noori Shakir M, Qasim MT, Hariri M, Nouhi Kararoudi A, Valilo M. Potential Role of Nrf2, HER2, and ALDH in Cancer Stem Cells: A Narrative Review. J Membr Biol 2024; 257:3-16. [PMID: 38356054 DOI: 10.1007/s00232-024-00307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024]
Abstract
Cancer is one of the main causes of death among humans, second only to cardiovascular diseases. In recent years, numerous studies have been conducted on the pathophysiology of cancer, and it has been established that this disease is developed by a group of stem cells known as cancer stem cells (CSCs). Thus, cancer is considered a stem cell disease; however, there is no comprehensive consensus about the characteristics of these cells. Several different signaling pathways including Notch, Hedgehog, transforming growth factor-β (TGF-β), and WNT/β-catenin pathways cause the self-renewal of CSCs. CSCs change their metabolic pathways in order to access easy energy. Therefore, one of the key objectives of researchers in cancer treatment is to destroy CSCs. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays an essential role in the protection of CSCs from reactive oxygen species (ROS) and chemotherapeutic agents by regulating antioxidants and detoxification enzymes. Human epidermal growth factor receptor 2 (HER2) is a member of the tyrosine kinase receptor family, which contributes to the protection of cancer cells against treatment and implicated in the invasion, epithelial-mesenchymal transition (EMT), and tumorigenesis. Aldehyde dehydrogenases (ALDHs) are highly active in CSCs and protect the cells against damage caused by active aldehydes through the regulation of aldehyde metabolism. On the other hand, ALDHs promote the formation and maintenance of tumor cells and lead to drug resistance in tumors through the activation of various signaling pathways, such as the ALDH1A1/HIF-1α/VEGF axis and Wnt/β-catenin, as well as changing the intracellular pH value. Given the growing body of information in this field, in the present narrative review, we attempted to shed light on the function of Nrf2, HER2, and ALDH in CSCs.
Collapse
Affiliation(s)
| | | | | | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Maytham T Qasim
- Department of Anesthesia, College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Maryam Hariri
- Department of Pathobiology, Auburn University, Auburn, AL, 36832, USA
| | - Alireza Nouhi Kararoudi
- Department of Biology, Faculty of Sciences, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Mohammad Valilo
- Dpartment of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
6
|
Rawat L, Balan M, Sasamoto Y, Sabarwal A, Pal S. A novel combination therapy with Cabozantinib and Honokiol effectively inhibits c-Met-Nrf2-induced renal tumor growth through increased oxidative stress. Redox Biol 2023; 68:102945. [PMID: 37898101 PMCID: PMC10628632 DOI: 10.1016/j.redox.2023.102945] [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: 10/11/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023] Open
Abstract
Receptor tyrosine kinase (RTK), c-Met, is overexpressed and hyper active in renal cell carcinoma (RCC). Most of the therapeutic agents mediate cancer cell death through increased oxidative stress. Induction of c-Met in renal cancer cells promotes the activation of redox-sensitive transcription factor Nrf2 and cytoprotective heme oxygenase-1 (HO-1), which can mediate therapeutic resistance against oxidative stress. c-Met/RTK inhibitor, Cabozantinib, has been approved for the treatment of advanced RCC. However, acquired drug resistance is a major hurdle in the clinical use of cabozantinib. Honokiol, a naturally occurring phenolic compound, has a great potential to downregulate c-Met-induced pathways. In this study, we found that a novel combination treatment with cabozantinib + Honokiol inhibits the growth of renal cancer cells in a synergistic manner through increased production of reactive oxygen species (ROS); and it significantly facilitates apoptosis-and autophagy-mediated cancer cell death. Activation of c-Met can induce Rubicon (a negative regulator of autophagy) and p62 (an autophagy adaptor protein), which can stabilize Nrf2. By utilizing OncoDB online database, we found a positive correlation among c-Met, Rubicon, p62 and Nrf2 in renal cancer. Interestingly, the combination treatment significantly downregulated Rubicon, p62 and Nrf2 in RCC cells. In a tumor xenograft model, this combination treatment markedly inhibited renal tumor growth in vivo; and it is associated with decreased expression of Rubicon, p62, HO-1 and vessel density in the tumor tissues. Together, cabozantinib + Honokiol combination can significantly inhibit c-Met-induced and Nrf2-mediated anti-oxidant pathway in renal cancer cells to promote increased oxidative stress and tumor cell death.
Collapse
Affiliation(s)
- Laxminarayan Rawat
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Murugabaskar Balan
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Yuzuru Sasamoto
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA; Division of Genetics, Brigham and Women's Hospital, MA, USA; Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Akash Sabarwal
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Soumitro Pal
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Baumel-Alterzon S, Tse I, Heidery F, Garcia-Ocaña A, Scott DK. NRF2 Dysregulation in Mice Leads to Inadequate Beta-Cell Mass Expansion during Pregnancy and Gestational Diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555207. [PMID: 37693560 PMCID: PMC10491153 DOI: 10.1101/2023.08.28.555207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The late stages of the mammalian pregnancy are accompanied with increased insulin resistance due to the increased glucose demand of the growing fetus. Therefore, as a compensatory response to maintain the maternal normal blood glucose levels, maternal beta-cell mass expands leading to increased insulin release. Defects in beta-cell adaptive expansion during pregnancy can lead to gestational diabetes mellitus (GDM). Although the exact mechanisms that promote GDM are poorly understood, GDM strongly associates with impaired beta-cell proliferation and with increased levels of reactive oxygen species (ROS). Here, we show that NRF2 levels are upregulated in mouse beta-cells at gestation day 15 (GD15) concomitant with increased beta-cell proliferation. Importantly, mice with tamoxifen-induced beta-cell-specific NRF2 deletion display inhibition of beta-cell proliferation, increased beta-cell oxidative stress and elevated levels of beta-cell death at GD15. This results in attenuated beta-cell mass expansion and disturbed glucose homeostasis towards the end of pregnancy. Collectively, these results highlight the importance of NRF2-oxidative stress regulation in beta-cell mass adaptation to pregnancy and suggest NRF2 as a potential therapeutic target for treating GDM.
Collapse
Affiliation(s)
- Sharon Baumel-Alterzon
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Isabelle Tse
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Fatema Heidery
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adolfo Garcia-Ocaña
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute at City of Hope, Duarte, CA
| | - Donald K. Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
8
|
Zhou C, Huang Y, Nie S, Zhou S, Gao X, Chen G. Biological effects and mechanisms of fisetin in cancer: a promising anti-cancer agent. Eur J Med Res 2023; 28:297. [PMID: 37626424 PMCID: PMC10464434 DOI: 10.1186/s40001-023-01271-8] [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: 07/10/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Fisetin, a natural flavonoid, possesses numerous biological activities that have been extensively studied in various diseases. When it comes to cancer, fisetin exhibits a range of biological effects, such as suppressing cell growth, triggering programmed cell death, reducing the formation of new blood vessels, protecting against oxidative stress, and inhibiting cell migration. Moreover, fisetin has the ability to enhance the effectiveness of chemotherapy. The anticancer properties of fisetin can be attributed to a diverse array of molecules and signaling pathways, including vascular endothelial growth factor (VEGF), mitogen-activated protein kinase (MAPK), nuclear factor-kappa B (NF-κB), PI3K/Akt/mTOR, and Nrf2/HO-1. Consequently, fisetin holds promise as a therapeutic agent for anticancer treatment. In this review, we place emphasis on the biological functions and various molecular targets of fisetin in anticancer therapy.
Collapse
Affiliation(s)
- Chenhui Zhou
- School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China
| | - Yi Huang
- School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China
| | - Sheng Nie
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China
| | - Shengjun Zhou
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China
| | - Xiang Gao
- School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, 315300, China.
| | - Gao Chen
- School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Department of Neurosurgery, School of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Mekapogu AR, Xu Z, Pothula S, Perera C, Pang T, Hosen SMZ, Damalanka V, Janetka J, Goldstein D, Pirola R, Wilson J, Apte M. HGF/c-Met pathway inhibition combined with chemotherapy increases cytotoxic T-cell infiltration and inhibits pancreatic tumour growth and metastasis. Cancer Lett 2023:216286. [PMID: 37354984 DOI: 10.1016/j.canlet.2023.216286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Pancreatic cancer (PC) is a deadly cancer with a high mortality rate. The unique characteristics of PC, including desmoplasia and immunosuppression, have made it difficult to develop effective treatment strategies. Pancreatic stellate cells (PSCs) play a crucial role in the progression of the disease by interacting with cancer cells. One of the key mediators of PSC - cancer cell interactions is the hepatocyte growth factor (HGF)/c-MET pathway. Using an immunocompetent in vivo model of PC as well as in vitro experiments, this study has shown that a combined approach using HGF/c-MET inhibitors to target stromal-tumour interactions and chemotherapy (gemcitabine) to target cancer cells effectively decreases tumour volume, EMT, and stemness, and importantly, eliminates metastasis. Notably, HGF/c-MET inhibition decreases TGF-β secretion by cancer cells, resulting in an increase in cytotoxic T-cell infiltration, thus contributing to cancer cell death in tumours. HGF/c-MET inhibition + chemotherapy was also found to normalise the gut microbiome and improve gut microbial diversity. These findings provide a strong platform for assessment of this triple therapy (HGF/c-MET inhibition + chemotherapy) approach in the clinical setting.
Collapse
Affiliation(s)
- Alpha Raj Mekapogu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Srinivasa Pothula
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia; AbCellera, Beaconsfield, New South Wales, United Kingdom
| | - Chamini Perera
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Tony Pang
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia; Surgical Innovations Unit, Westmead Hospital, Sydney, Australia; Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - S M Zahid Hosen
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Vishnu Damalanka
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, USA
| | - James Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, USA
| | - David Goldstein
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Romano Pirola
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Jeremy Wilson
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Minoti Apte
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia.
| |
Collapse
|
10
|
Wang H, Cheng Q, Bao L, Li M, Chang K, Yi X. Cytoprotective Role of Heme Oxygenase-1 in Cancer Chemoresistance: Focus on Antioxidant, Antiapoptotic, and Pro-Autophagy Properties. Antioxidants (Basel) 2023; 12:1217. [PMID: 37371947 DOI: 10.3390/antiox12061217] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Chemoresistance remains the foremost challenge in cancer therapy. Targeting reactive oxygen species (ROS) manipulation is a promising strategy in cancer treatment since tumor cells present high levels of intracellular ROS, which makes them more vulnerable to further ROS elevation than normal cells. Nevertheless, dynamic redox evolution and adaptation of tumor cells are capable of counteracting therapy-induced oxidative stress, which leads to chemoresistance. Hence, exploring the cytoprotective mechanisms of tumor cells is urgently needed to overcome chemoresistance. Heme oxygenase-1 (HO-1), a rate-limiting enzyme of heme degradation, acts as a crucial antioxidant defense and cytoprotective molecule in response to cellular stress. Recently, emerging evidence indicated that ROS detoxification and oxidative stress tolerance owing to the antioxidant function of HO-1 contribute to chemoresistance in various cancers. Enhanced HO-1 expression or enzymatic activity was revealed to promote apoptosis resistance and activate protective autophagy, which also involved in the development of chemoresistance. Moreover, inhibition of HO-1 in multiple cancers was identified to reversing chemoresistance or improving chemosensitivity. Here, we summarize the most recent advances regarding the antioxidant, antiapoptotic, and pro-autophagy properties of HO-1 in mediating chemoresistance, highlighting HO-1 as a novel target for overcoming chemoresistance and improving the prognosis of cancer patients.
Collapse
Affiliation(s)
- Huan Wang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Qi Cheng
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Lingjie Bao
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Mingqing Li
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Kaikai Chang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Xiaofang Yi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| |
Collapse
|
11
|
Ben-Eltriki M, Gayle EJ, Walker N, Deb S. Pharmacological Significance of Heme Oxygenase 1 in Prostate Cancer. Curr Issues Mol Biol 2023; 45:4301-4316. [PMID: 37232742 DOI: 10.3390/cimb45050273] [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: 04/13/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Heme oxygenase 1 (HO-1) is a detoxifying antioxidant microsomal enzyme that regulates inflammation, apoptosis, cell proliferation, and angiogenesis in prostate cancer (PCa). This makes HO-1 a promising target for therapeutic prevention and treatment due to its anti-inflammatory properties and ability to control redox homeostasis. Clinical evidence highlights the possible correlation between HO-1 expression and PCa growth, aggressiveness, metastasized tumors, resistance to therapy, and poor clinical outcomes. Interestingly, studies have reported anticancer benefits mediated by both HO-1 induction and inhibition in PCa models. Contrasting evidence exists on the role of HO-1 in PCa progression and possible treatment targets. Herein, we provide an overview of available evidence on the clinical significance of HO-1 signaling in PCa. It appears that the beneficial effects of HO-1 induction or inhibition are dependent on whether it is a normal versus malignant cell as well as the intensity (major vs. minor) of the increase in HO-1 enzymatic activity. The current literature evidence indicates that HO-1 has dual effects in PCa. The amount of cellular iron and reactive oxygen species (ROS) can determine the role of HO-1 in PCa. A major increase in ROS enforces HO-1 to a protective role. HO-1 overexpression may provide cryoprotection to normal cells against oxidative stress via suppressing the expression of proinflammatory genes, and thus offer therapeutic prevention. In contrast, a moderate increase in ROS can lead to the perpetrator role of HO-1, which is associated with PCa progression and metastasis. HO-1 inhibition by xenobiotics in DNA-damaged cells tilts the balance to promote apoptosis and inhibit PCa proliferation and metastasis. Overall, the totality of the evidence revealed that HO-1 may play a dual role in the therapeutic prevention and treatment of PCa.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- Department of Pharmacology and Therapeutics, Clinical Pharmacology Lab, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Erysa J Gayle
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA
| | - Noah Walker
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA
| |
Collapse
|
12
|
Yang X, Dong S, Li C, Li M, Xing C, He J, Peng C, Shao H, Jia Q. Hydroquinone triggers pyroptosis and endoplasmic reticulum stress via AhR-regulated oxidative stress in human lymphocytes. Toxicol Lett 2023; 376:39-50. [PMID: 36646296 DOI: 10.1016/j.toxlet.2023.01.005] [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: 10/10/2022] [Revised: 01/04/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Benzene is a frequent component of environmental pollution and is abundant in petrochemicals, decorative materials, motor vehicle exhaust and cigarette smoke. Benzene is a well-known carcinogen in humans and animals, but the molecular mechanism has not yet been elucidated. Our earlier research indicated that hydroquinone (HQ), one of the main reactive metabolites of benzene, could activate aryl hydrocarbon receptor (AhR), which is essential for HQ-induced toxicity, including apoptosis and DNA damage. Since AhR is an important regulator of the immune system that integrates the environmental stimulus and immune response, we examined whether and how HQ-induced AhR activity could lead to NLRP3 inflammasome-dependent pyroptosis in JHP cells. Our results showed that HQ could cause inflammation process and resultant pyroptosis. In JHP cells, HQ also induced endoplasmic reticulum stress (ERS) by releasing excessive reactive oxygen species (ROS). The activation of pyroptosis induced by HQ treatment was reversed by an antioxidant (NAC) and an ERS inhibitor (4-PBA). Interestingly, the treatment of CH223191, an AhR inhibitor, reversed HQ-induced oxidative stress, ERS and pyroptosis. These data suggested that AhR-mediated HQ-induced ERS, ROS and inflammasome activation may play vital roles in the toxic effects of benzene. This work provides insights and prospective strategies into potential mechanisms for reducing benzene-induced hematotoxicity.
Collapse
Affiliation(s)
- Xiaohan Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China
| | - Shuangyan Dong
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China
| | - Ming Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China
| | - Caihong Xing
- Key Laboratory of Chemical Safety and Health, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention (CDC), Beijing 100050, China
| | - Jin He
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China
| | - Cheng Peng
- Eusyn Institute of Health Science, Brisbane, QLD 4108, Australia
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China.
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan 250062, China.
| |
Collapse
|
13
|
Hu J, Chen L, Lu Z, Yao H, Hu Y, Feng L, Pang Y, Wu JQ, Yu Z, Chen WH. Design, Synthesis and Antitumor Activity of Novel Selenium-Containing Tepotinib Derivatives as Dual Inhibitors of c-Met and TrxR. Molecules 2023; 28:molecules28031304. [PMID: 36770971 PMCID: PMC9921947 DOI: 10.3390/molecules28031304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Cellular mesenchymal-epithelial transition factor (c-Met), an oncogenic transmembrane receptor tyrosine kinase (RTK), plays an essential role in cell proliferation during embryo development and liver regeneration. Thioredoxin reductase (TrxR) is overexpressed and constitutively active in most tumors closely related to cancer recurrence. Multi-target-directed ligands (MTDLs) strategy provides a logical approach to drug combinations and would adequately address the pathological complexity of cancer. In this work, we designed and synthesized a series of selenium-containing tepotinib derivatives by means of selenium-based bioisosteric modifications and evaluated their antiproliferative activity. Most of these selenium-containing hybrids exhibited potent dual inhibitory activity toward c-Met and TrxR. Among them, compound 8b was the most active, with an IC50 value of 10 nM against MHCC97H cells. Studies on the mechanism of action revealed that compound 8b triggered cell cycle arrest at the G1 phase and caused ROS accumulations by targeting TrxR, and these effects eventually led to cell apoptosis. These findings strongly suggest that compound 8b serves as a dual inhibitor of c-Met and TrxR, warranting further exploitation for cancer therapy.
Collapse
Affiliation(s)
- Jinhui Hu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
- Correspondence: (J.H.); (W.-H.C.)
| | - Li Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Zhonghui Lu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Han Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yunfei Hu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Luanqi Feng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Yanqing Pang
- Department of Phase I Clinical Research Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jia-Qiang Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Zhiling Yu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Wen-Hua Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
- Correspondence: (J.H.); (W.-H.C.)
| |
Collapse
|
14
|
Ibáñez Gaspar V, McMorrow T. The Curcuminoid EF24 in Combination with TRAIL Reduces Human Renal Cancer Cell Migration by Decreasing MMP-2/MMP-9 Activity through a Reduction in H 2O 2. Int J Mol Sci 2023; 24:ijms24021043. [PMID: 36674555 PMCID: PMC9863498 DOI: 10.3390/ijms24021043] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Cancer cells present high levels of oxidative stress, and although an increase in reactive oxygen species (ROS), such as H2O2, can lead to apoptosis, it can also induce cell invasion and metastasis. As the increase in ROS can lead to an increase in the expression of MMP-2 and MMP-9, thus causing the degradation of the extracellular matrix, an increase in the ROS H2O2 might have an impact on MMP-2/MMP-9 activity. The natural compound curcumin has shown some anticancer effects, although its bioavailability hinders its therapeutic potential. However, curcumin and its analogues were shown to resensitize kidney cancer cells to TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. This study shows that the curcuminoid EF24 in combination with TRAIL increases peroxidase activity in the renal adenocarcinoma cell line ACHN, reducing the level of intracellular H2O2 and MMP-2/MMP-9 activity, a mechanism that is also observed after treatment with curcumin and TRAIL.
Collapse
|
15
|
Farhan M, Rizvi A. Understanding the Prooxidant Action of Plant Polyphenols in the Cellular Microenvironment of Malignant Cells: Role of Copper and Therapeutic Implications. Front Pharmacol 2022; 13:929853. [PMID: 35795551 PMCID: PMC9251333 DOI: 10.3389/fphar.2022.929853] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/02/2022] [Indexed: 12/13/2022] Open
Abstract
Plant derived polyphenolic compounds are considered critical components of human nutrition and have shown chemotherapeutic effects against a number of malignancies. Several studies have confirmed the ability of polyphenols to induce apoptosis and regression of tumours in animal models. However, the mechanism through which polyphenols modulate their malignant cell selective anticancer effects has not been clearly established. While it is believed that the antioxidant properties of these molecules may contribute to lowering the risk of cancer induction by causing oxidative damage to DNA, it could not be held responsible for chemotherapeutic properties and apoptosis induction. It is a well known fact that cellular copper increases within the malignant cell and in serum of patients harboring malignancies. This phenomenon is independent of the cellular origin of malignancies. Based on our own observations and those of others; over the last 30 years our laboratory has shown that cellular copper reacts with plant derived polyphenolic compounds, by a Fenton like reaction, which generates reactive oxygen species and leads to genomic DNA damage. This damage then causes an apoptosis like cell death of malignant cells, while sparing normal cells. This communication reviews our work in this area and lays the basis for understanding how plant derived polyphenols can behave as prooxidants (and not antioxidants) within the microenvironment of a malignancy (elevated copper levels) and gives rationale for their preferential cytotoxicity towards malignant cells.
Collapse
Affiliation(s)
- Mohd Farhan
- Department of Basic Sciences, Preparatory Year Deanship, King Faisal University, Al-Ahsa, Saudi Arabia
- *Correspondence: Mohd Farhan,
| | - Asim Rizvi
- Department of Kulliyat, Faculty of Unani Medicine, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
16
|
Abbotts R, Dellomo AJ, Rassool FV. Pharmacologic Induction of BRCAness in BRCA-Proficient Cancers: Expanding PARP Inhibitor Use. Cancers (Basel) 2022; 14:2640. [PMID: 35681619 PMCID: PMC9179544 DOI: 10.3390/cancers14112640] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 12/17/2022] Open
Abstract
The poly(ADP-ribose) polymerase (PARP) family of proteins has been implicated in numerous cellular processes, including DNA repair, translation, transcription, telomere maintenance, and chromatin remodeling. Best characterized is PARP1, which plays a central role in the repair of single strand DNA damage, thus prompting the development of small molecule PARP inhibitors (PARPi) with the intent of potentiating the genotoxic effects of DNA damaging agents such as chemo- and radiotherapy. However, preclinical studies rapidly uncovered tumor-specific cytotoxicity of PARPi in a subset of cancers carrying mutations in the BReast CAncer 1 and 2 genes (BRCA1/2), which are defective in the homologous recombination (HR) DNA repair pathway, and several PARPi are now FDA-approved for single agent treatment in BRCA-mutated tumors. This phenomenon, termed synthetic lethality, has now been demonstrated in tumors harboring a number of repair gene mutations that produce a BRCA-like impairment of HR (also known as a 'BRCAness' phenotype). However, BRCA mutations or BRCAness is present in only a small subset of cancers, limiting PARPi therapeutic utility. Fortunately, it is now increasingly recognized that many small molecule agents, targeting a variety of molecular pathways, can induce therapeutic BRCAness as a downstream effect of activity. This review will discuss the potential for targeting a broad range of molecular pathways to therapeutically induce BRCAness and PARPi synthetic lethality.
Collapse
Affiliation(s)
- Rachel Abbotts
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.J.D.); (F.V.R.)
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| | - Anna J. Dellomo
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.J.D.); (F.V.R.)
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| | - Feyruz V. Rassool
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.J.D.); (F.V.R.)
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| |
Collapse
|
17
|
Cao Y, Liang Q, Lan Y, Liu Y. The therapeutic efficacy and safety improvements of crizotinib prodrug micelles on breast cancer treatment. Pharm Dev Technol 2022; 27:469-478. [PMID: 35579888 DOI: 10.1080/10837450.2022.2078984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nowadays, breast cancer has become a major killer threatening women health. MET is a receptor tyrosine kinase that upon binding of its ligand, hepatocyte growth factor, activates downstream pathways with diverse cellular functions which are important in occurrence and development of breast cancer. Crizotinib (Cro) is multi-target tyrosine kinase inhibitor targeting ALK gene recombination, MET gene amplification and ROS gene. Although, Cro has ideal treatment of breast cancer, Cro has stronger hepatotoxicity and lacks targeting capacity to tumor cell, which limited Cro to effectively therapy breast cancer. In this study, we develop a novel prodrug micelle through polymerization reaction polymerizing Cro onto the chain to form POEG-b-PCro prodrug micelles, which the drug loading capacity of Cro was significantly increased to improve cumulant of tumor. Pharmacokinetic and biodistribution studies illustrated POEG-b-PCro prodrug micelles had significant effect through improving Cro content in tumor. Meanwhile, antitumor mechanism of POEG-b-PCro prodrug micelles proved POEG-b-PCro prodrug micelles had stronger effect through reducing negative regulatory proteins. POEG-b-PCro prodrug micelles had splendid safety through safety study in vivo to account for POEG-b-PCro prodrug micelles. Therefore, POEG-b-PCro prodrug micelles are a promising drug delivery strategy for reducing toxicity and enhancing efficacy of Cro.
Collapse
Affiliation(s)
- Yongjing Cao
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan, 750004, China
| | - Qiangwei Liang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan, 750004, China
| | - Yang Lan
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan, 750004, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan, 750004, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ningxia Medical University, Yinchuan, 750004, China
| |
Collapse
|
18
|
Sabarwal A, Wedel J, Liu K, Zurakowski D, Chakraborty S, Flynn E, Briscoe DM, Balan M, Pal S. A Combination therapy using an mTOR inhibitor and Honokiol effectively induces autophagy through the modulation of AXL and Rubicon in renal cancer cells and restricts renal tumor growth following organ transplantation. Carcinogenesis 2021; 43:360-370. [PMID: 34965300 PMCID: PMC9118982 DOI: 10.1093/carcin/bgab126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 12/31/2022] Open
Abstract
Development of cancer, including renal cancer, is a major problem in immunosuppressed patients. The mTOR inhibitor Rapamycin (RAPA) is used as an immunosuppressive agent in patients with organ transplants and other immunological disorders; and it also has antitumorigenic potential. However, long-term use of RAPA causes reactivation of Akt, and ultimately leads to enhanced tumor growth. Honokiol (HNK) is a natural compound, which possesses both anti-inflammatory and antitumorigenic properties. In this study, we investigated the effect of a novel combination therapy using RAPA + HNK on allograft survival and post-transplantation renal tumor growth. We observed that it effectively modulated the expression of some key regulatory molecules (like Carabin, an endogenous Ras inhibitor; and Rubicon, a negative regulator of autophagy) that play important roles in tumor cell growth and survival. This combination induced toxic autophagy and apoptosis to promote cancer cell death; and was associated with a reduced expression of the tumor-promoting receptor tyrosine kinase AXL. Finally, we utilized a novel murine model to examine the effect of RAPA + HNK on post-transplantation renal tumor growth. The combination treatment prolonged the allograft survival and significantly inhibited post-transplantation tumor growth. It was associated with reduced tumor expression of Rubicon and the cytoprotective/antioxidant heme oxygenase-1 to overcome therapeutic resistance. It also downregulated the coinhibitory programmed death-1 ligand, which plays major role(s) in the immune escape of tumor cells. Together, this combination treatment has a great potential to restrict renal tumor growth in transplant recipients as well as other immunosuppressed patients.
Collapse
Affiliation(s)
- Akash Sabarwal
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Johannes Wedel
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Kaifeng Liu
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - David Zurakowski
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Samik Chakraborty
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Evelyn Flynn
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - David M Briscoe
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA,Transplant Research Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Murugabaskar Balan
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Soumitro Pal
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA,To whom correspondence should be addressed. Division of Nephrology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA. Tel: +1 617 919 2989; Fax: +1 617 730 0365;
| |
Collapse
|
19
|
Shi J, Xiong Z, Wang K, Yuan C, Huang Y, Xiao W, Meng X, Chen Z, Lv Q, Miao D, Liang H, Xu T, Xie K, Yang H, Zhang X. HIF2α promotes tumour growth in clear cell renal cell carcinoma by increasing the expression of NUDT1 to reduce oxidative stress. Clin Transl Med 2021; 11:e592. [PMID: 34841698 PMCID: PMC8567048 DOI: 10.1002/ctm2.592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The key role of hypoxia-inducible factor 2alpha (HIF2α) in the process of renal cancer has been confirmed. In the field of tumour research, oxidative stress is also considered to be an important influencing factor. However, the relationship and biological benefits of oxidative stress and HIF2α in ccRCC remain unclear. This research attempts to explore the effect of oxidative stress on the cancer-promoting effect of HIF2α in ccRCC and reveal its mechanism of action. METHODS The bioinformatics analysis for ccRCC is based on whole transcriptome sequencing and TCGA database. The detection of the expression level of related molecules is realised by western blot and PCR. The expression of Nucleoside diphosphate-linked moiety X-type motif 1 (NUDT1) was knocked down by lentiviral infection technology. The functional role of NUDT1 were further investigated by CCK8 assays, transwell assays and cell oxidative stress indicator detection. The exploration of related molecular mechanisms is realised by Luciferase assays and Chromatin immunoprecipitation (ChIP) assays. RESULTS Molecular screening based on knockdown HIF2α sequencing data and oxidative stress related data sets showed that NUDT1 is considered to be an important molecule for the interaction of HIF2α with oxidative stress. Subsequent experimental results showed that NUDT1 can cooperate with HIF2α to promote the progression of ccRCC. And this biological effect was found to be caused by the oxidative stress regulated by NUDT1. Mechanistically, HIF2α transcription activates the expression of NUDT1, thereby inhibiting oxidative stress and promoting the progression of ccRCC. CONCLUSIONS This research clarified a novel mechanism by which HIF2α stabilises sirtuin 3 (SIRT3) through direct transcriptional activation of NUDT1, thereby inhibiting oxidative stress to promote the development of ccRCC. It provided the possibility for the selection of new therapeutic targets for ccRCC and the study of combination medication regimens.
Collapse
Affiliation(s)
- Jian Shi
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Zhiyong Xiong
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Keshan Wang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Changfei Yuan
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Yu Huang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Wen Xiao
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Xiangui Meng
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Zhixian Chen
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Qingyang Lv
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Daojia Miao
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Huageng Liang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Tianbo Xu
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Kairu Xie
- Department of Pathogenic BiologySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Hongmei Yang
- Department of Pathogenic BiologySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Xiaoping Zhang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| |
Collapse
|
20
|
Lawal B, Wang YC, Wu ATH, Huang HS. Pro-Oncogenic c-Met/EGFR, Biomarker Signatures of the Tumor Microenvironment are Clinical and Therapy Response Prognosticators in Colorectal Cancer, and Therapeutic Targets of 3-Phenyl-2H-benzo[e][1,3]-Oxazine-2,4(3H)-Dione Derivatives. Front Pharmacol 2021; 12:691234. [PMID: 34512327 PMCID: PMC8429938 DOI: 10.3389/fphar.2021.691234] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022] Open
Abstract
Genetic and environmental factors play important roles in cancer progression, metastasis, and drug resistance. Herein, we used a multiomics data analysis to evaluate the predictive and prognostic roles of genetic and epigenetic modulation of c-MET (hepatocyte growth factor receptor)/epidermal growth factor receptor (EGFR) in colorectal cancer (CRC). First, we found that overexpressions of c-MET/EGFR were associated with the infiltration of tumor immune cells and cancer-associated fibroblasts, and were of prognostic relevance in CRC cohorts. We also observed that genetic alterations of c-MET/EGFR in CRC co-occurred with other gene alterations and were associated with overexpression of messenger (m)RNA of some cancer hallmark proteins. More specifically, DNA-methylation and somatic copy number alterations of c-MET/EGFR were associated with immune infiltration, dysfunctional T-cell phenotypes, and poor prognoses of the cohorts. Moreover, we describe two novel gefitinib-inspired small molecules derivatives of 3-phenyl-2H-benzo[e] [1,3]-oxazine-2,4(3H)-dione, NSC777205 and NSC777207, which exhibited wide-spectrum antiproliferative activities and selective cytotoxic preference for drug-sensitive and multidrug-resistant melanoma, renal, central nervous system, colon, and non-small cell lung cancer cell lines. We further provided in silico mechanistic evidence implicating c-MET/EGFR/phosphatidylinositol 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) inhibition in anticancer activities of those compounds. Our overall structure-activity relationship study revealed that the addition of an –OCH3 group to salicylic core of NSC777207 was not favorable, as the added moiety led to overall less-favorable drug properties as well as weaker anticancer activities compared to the properties and activities demonstrated by NSC777205 that has no –OCH3 substituent group. Further in vitro and in vivo analyses in tumor-bearing mice are ongoing in our lab to support this claim and to unravel the full therapeutic efficacies of NSC777205 and NSC777207 in CRC.
Collapse
Affiliation(s)
- Bashir Lawal
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan.,Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chi Wang
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Alexander T H Wu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,The PhD Program of Translational Medicine, College of Science and Technology, Taipei Medical University, Taipei, Taiwan.,Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan.,Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,School of Pharmacy, National Defense Medical Center, Taipei, Taiwan.,PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
21
|
The Role of HO-1 and Its Crosstalk with Oxidative Stress in Cancer Cell Survival. Cells 2021; 10:cells10092401. [PMID: 34572050 PMCID: PMC8471703 DOI: 10.3390/cells10092401] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Heme oxygenases (HOs) act on heme degradation to produce carbon monoxide (CO), free iron, ferritin, and biliverdin. Upregulation of cellular HO-1 levels is signature of oxidative stress for its downstream effects particularly under pro-oxidative status. Subcellular traffics of HO-1 to different organelles constitute a network of interactions compromising a variety of effectors such as pro-oxidants, ROS, mitochondrial enzymes, and nucleic transcription factors. Some of the compartmentalized HO-1 have been demonstrated as functioning in the progression of cancer. Emerging data show the multiple roles of HO-1 in tumorigenesis from pathogenesis to the progression to malignancy, metastasis, and even resistance to therapy. However, the role of HO-1 in tumorigenesis has not been systematically addressed. This review describes the crosstalk between HO-1 and oxidative stress, and following redox regulation in the tumorigenesis. HO-1-regulated signaling pathways are also summarized. This review aims to integrate basic information and current progress of HO-1 in cancer research in order to enhance the understandings and facilitate following studies.
Collapse
|
22
|
Yang H, Luo F, Wei Y, Jiao Y, Qian J, Chen S, Gong Y, Tang L. TGR5 protects against cholestatic liver disease via suppressing the NF-κB pathway and activating the Nrf2/HO-1 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1158. [PMID: 34430599 PMCID: PMC8350648 DOI: 10.21037/atm-21-2631] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/28/2021] [Indexed: 01/13/2023]
Abstract
Background Characterized by the presence of inflammation, fibrosis, and bile duct proliferation, cholestatic liver disease (CLD) affects people of all age groups. Takeda G-protein-coupled receptor (TGR5) has been implicated in the suppression of inflammation via toll-like receptor 4 (TLR4) and nuclear factor kappa B (NF-κB). Kupffer cells and their M1 polarization play important roles in inflammation and cholestatic liver injury via production of pro-inflammatory cytokines. Nevertheless, the function of TGR5 signaling in CLD is largely unknown. Methods We conducted liver tissue experiments, animal experiments, serum marker testing, liver histology analysis, Kupffer cell experiments, RNA extraction and Real-time PCR, western blotting, evaluation of ROS production by flow cytometry and statistical differences were analyzed by student t-test using GraphPad Prism. Results We found that serum bile acid (BA) and TGR5 levels were elevated in patients with cholestasis cirrhosis. Knockout of TGR5 in animals significantly increased bile duct ligation (BDL)-caused liver injury through increasing oxidative stress, promoting M1-predominant polarization of Kupffer cells, and elevating the serum levels of inflammatory cytokines. In contrast, TGR5 activation inhibited ROS production, secretion of pro-inflammatory cytokines, and M1-predominant polarization of Kupffer cells. Moreover, results showed that TGR5 exerted its effects via suppressing NF-κB signaling and activating nuclear factor 2 (Nrf2)/HO-1 signaling. Finally, the effect of TGR5 on cholestatic liver damage was also confirmed in vivo. Conclusions TGR5 activation protected against BDL-induced CLD by both suppressing inflammation via inhibiting the NF-κB pathway and reducing ROS production via activation of Nrf2/HO-1 signaling. These findings show the importance of TGR5 in CLD and provide new insight into therapeutic strategies for CLD.
Collapse
Affiliation(s)
- Haojun Yang
- Gastrointestinal Surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Fengyong Luo
- School of Graduate, Dalian Medical University, Dalian, China
| | - Yi Wei
- Gastrointestinal Surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yuwen Jiao
- Gastrointestinal Surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Jun Qian
- Gastrointestinal Surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Shuai Chen
- Gastrointestinal Surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yu Gong
- Gastrointestinal Surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Liming Tang
- Gastrointestinal Surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
23
|
Chakraborty S, Balan M, Sabarwal A, Choueiri TK, Pal S. Metabolic reprogramming in renal cancer: Events of a metabolic disease. Biochim Biophys Acta Rev Cancer 2021; 1876:188559. [PMID: 33965513 PMCID: PMC8349779 DOI: 10.1016/j.bbcan.2021.188559] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022]
Abstract
Recent studies have established that tumors can reprogram the pathways involved in nutrient uptake and metabolism to withstand the altered biosynthetic, bioenergetics and redox requirements of cancer cells. This phenomenon is called metabolic reprogramming, which is promoted by the loss of tumor suppressor genes and activation of oncogenes. Because of alterations and perturbations in multiple metabolic pathways, renal cell carcinoma (RCC) is sometimes termed as a "metabolic disease". The majority of metabolic reprogramming in renal cancer is caused by the inactivation of von Hippel-Lindau (VHL) gene and activation of the Ras-PI3K-AKT-mTOR pathway. Hypoxia-inducible factor (HIF) and Myc are other important players in the metabolic reprogramming of RCC. All types of RCCs are associated with reprogramming of glucose and fatty acid metabolism and the tricarboxylic acid (TCA) cycle. Metabolism of glutamine, tryptophan and arginine is also reprogrammed in renal cancer to favor tumor growth and oncogenesis. Together, understanding these modifications or reprogramming of the metabolic pathways in detail offer ample opportunities for the development of new therapeutic targets and strategies, discovery of biomarkers and identification of effective tumor detection methods.
Collapse
Affiliation(s)
- Samik Chakraborty
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Murugabaskar Balan
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Akash Sabarwal
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Toni K Choueiri
- Dana Farber Cancer Institute, Boston, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Soumitro Pal
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America.
| |
Collapse
|
24
|
Dahiya M, Dureja H. Sorafenib for hepatocellular carcinoma: potential molecular targets and resistance mechanisms. J Chemother 2021; 34:286-301. [PMID: 34291704 DOI: 10.1080/1120009x.2021.1955202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most widespread typical therapy-resistant, unresectable type of malignant solid tumour with a high death rate constituting huge medical concern. Sorafenib is a small molecule oral multi-target kinase potent inhibitor that acts by suppressing/blocking the multiplication of the tumour cells, angiogenesis, and encouraging apoptosis of the tumour cells. Though, the precise mechanism of tumour cell death induction by sorafenib is yet under exploration. Furthermore, genetic heterogeneity plays a critical role in developing sorafenib resistance, which leads the way to identify the need for predictive biomarkers responsible for drug resistance. Therefore, it is essential to find out the fundamental resistance mechanisms to expand therapeutic plans. The authors summarize the molecular concepts of resistance, progression, potential molecular targets, HCC management therapies, and discussion on the advancements expected in the coming future, inclusive of biomarker-driven treatment strategies, which may provide the prospects to design innovative therapeutically targeted strategies for the HCC treatment and the clinical implementation of emerging targeted agents.
Collapse
Affiliation(s)
- Mandeep Dahiya
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
25
|
Dempke WCM, Reck M. KEAP1/NRF2 (NFE2L2) mutations in NSCLC - Fuel for a superresistant phenotype? Lung Cancer 2021; 159:10-17. [PMID: 34303275 DOI: 10.1016/j.lungcan.2021.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/04/2021] [Accepted: 07/10/2021] [Indexed: 12/18/2022]
Abstract
The transcription factor NRF2 (nuclear factor E2-related factor 2) (also known as nuclear factor, erythroid 2 like 2 [NFE2L2]) is the master regulator of cellular antioxidant responses. NRF2 is repressed by interaction with a redox-sensitive protein KEAP1 (Kelch-like ECH-associated protein 1). Dysregulation of KEAP1/NRF2 transcriptional activity has been associated with the pathogenesis of multiple diseases, and the KEAP1/NRF2 axis has emerged to be the most important modulator of cellular homeostasis. Oxidative stress plays an important role in the initiation and progression of many chronic diseases, including diabetes, cancer, and neurodegenerative diseases. Although its role in immunotherapy is still somewhat controversial, it is well documented from clinical studies that KEAP1/NRF2 mutations in NSCLCs are associated with resistance to various cancer treatments including chemotherapy, X-irradiation, TKI treatment, and a shorter OS and currently available results from clinical trials suggest that KEAP1/NRF2 mutations can be used as a prognostic biomarker (poorer prognosis) for determining prognosis following immunotherapy and a predictive marker for chemo-, radio-, immunotherapy- and TKI-resistance. Despite overwhelming enthusiasm about the various KEAP1/NRF2 inhibitors that have been described during the last decades, none of these inhibitors are currently explored in clinical trials or in clinical applications which clearly add weight to the proposal that the development of these inhibitors remains challenging, but will be beneficial for novel treatment approaches in NSCLC in the near future. In this review we highlight the molecular features, the key components, and possible inhibitors of the KEAP1/NRF2 pathway, its role as prognostic and predictive biomarker, and the resulting clinical implications in NSCLC patients.
Collapse
Affiliation(s)
- Wolfram C M Dempke
- University Clinic LMU Munich, Medical Clinic III, Marchioninistr. 15, D-81377 Munich, Germany.
| | - Martin Reck
- Department of Thoracic Oncology, Airway Research Center North, German Center for Lung Research, LungenClinic, Wöhrendamm 80, D-22927 Grosshansdorf, Germany
| |
Collapse
|
26
|
Li N, Zhao Y, Shen Y, Cheng Y, Qiao M, Song L, Huang X. Protective effects of folic acid on oxidative damage of rat spleen induced by lead acetate. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 211:111917. [PMID: 33497860 DOI: 10.1016/j.ecoenv.2021.111917] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/20/2020] [Accepted: 01/06/2021] [Indexed: 06/12/2023]
Abstract
Lead (Pb) is a heavy metal environmental pollutant that can cause functional damage and anemia of immune organs. More and more evidence indicate that the toxicity of lead was related to apoptosis driven by oxidative stress and endoplasmic reticulum stress. This article mainly discusses the protective effect and mechanism of folic acid intervention on lead-induced spleen injury and apoptosis. In this study, Sprague-Dawley rats were randomly divided into control group, lead exposure group (0.2% lead acetate), folic acid + lead group (0.4 mg/kg folic acid and 0.2% lead acetate), and folic acid group (0.4 mg/kg folic acid). By recording and calculating the rat's initial body weight, final body weight, net weight gain, daily weight gain, and spleen index, observe the rat's weight change and spleen weight. And adopt the immunofluorescence staining method to determine the expression level of NrF2, HO-1, GRP78, CHOP protein in the spleen. The results showed that The 0.4 mg/kg folic acid diet did not significantly improve in the body weight and spleen index of lead-exposed rats (P > 0.05). While compared with the control group, the expression levels of HO-1 and CHOP protein were significantly increased in the lead exposure group (P < 0.05), and the expression levels of HO-1 and CHOP protein were significantly reduced in the folic acid intervention group (P < 0.05). In conclusion, lead exposure increased the expression levels of HO-1 and CHOP in the spleen of rats, and caused damage to the spleen. Folic acid down-regulated the expression levels of HO-1 and CHOP proteins through the two pathways of NrF2/HO-1 and GRP78/CHOP, thereby exerting a certain protective effect and alleviating the spleen caused by lead-induced oxidative stress and endoplasmic reticulum stress damage.
Collapse
Affiliation(s)
- Ning Li
- College of Food Science and Technology, Henan Agriculture University, 450002, China.
| | - Yali Zhao
- College of Food Science and Technology, Henan Agriculture University, 450002, China
| | - Yue Shen
- College of Food Science and Technology, Henan Agriculture University, 450002, China
| | - Yongxia Cheng
- College of Food Science and Technology, Henan Agriculture University, 450002, China
| | - Mingwu Qiao
- College of Food Science and Technology, Henan Agriculture University, 450002, China
| | - Lianjun Song
- College of Food Science and Technology, Henan Agriculture University, 450002, China
| | - Xianqing Huang
- College of Food Science and Technology, Henan Agriculture University, 450002, China.
| |
Collapse
|
27
|
Sun J, Zhou C, Zhao Y, Zhang X, Chen W, Zhou Q, Hu B, Gao D, Raatz L, Wang Z, Nelson PJ, Jiang Y, Ren N, Bruns CJ, Zhou H. Quiescin sulfhydryl oxidase 1 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by driving EGFR endosomal trafficking and inhibiting NRF2 activation. Redox Biol 2021; 41:101942. [PMID: 33770521 PMCID: PMC8024711 DOI: 10.1016/j.redox.2021.101942] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/20/2021] [Accepted: 03/08/2021] [Indexed: 01/01/2023] Open
Abstract
Sorafenib is a first-line molecular-target drug for advanced hepatocellular carcinoma (HCC), but its clinical effects are still limited. In this study we identify Quiescin sulfhydryl oxidase 1 (QSOX1) acting as a cellular pro-oxidant, specifically in the context of sorafenib treatment of HCC. QSOX1 disrupts redox homoeostasis and sensitizes HCC cells to oxidative stress by inhibiting activation of the master antioxidant transcription factor NRF2. A negative correlation between QSOX1 and NRF2 expression was validated in tumor tissues from 151 HCC patients. Mechanistically, QSOX1 restrains EGF-induced EGFR activation by promoting ubiquitination-mediated degradation of EGFR and accelerating its intracellular endosomal trafficking, leading to suppression of NRF2 activity. Additionally, QSOX1 potentiates sorafenib-induced ferroptosis by suppressing NRF2 in vitro and in vivo. In conclusion, the data presented identify QSOX1 as a novel candidate target for sorafenib-based combination therapeutic strategies in HCC or other EGFR-dependent tumor types.
Collapse
Affiliation(s)
- Jialei Sun
- Liver Cancer Institute & Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University & Ministry of Education, Shanghai, China.
| | - Chenhao Zhou
- Liver Cancer Institute & Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University & Ministry of Education, Shanghai, China; Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yue Zhao
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany.
| | - Xiaofei Zhang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China.
| | - Wanyong Chen
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Fudan Minhang Academic Health System, And Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 200032, China.
| | - Qiang Zhou
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Fudan Minhang Academic Health System, And Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 200032, China.
| | - Bo Hu
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Dongmei Gao
- Liver Cancer Institute & Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University & Ministry of Education, Shanghai, China.
| | - Lisa Raatz
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany.
| | - Zhefang Wang
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany.
| | - Peter J Nelson
- Medical Clinic and Policlinic IV, University Clinic, Ludwig-Maximilians-University Munich, Germany.
| | - Yuchao Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| | - Ning Ren
- Liver Cancer Institute & Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University & Ministry of Education, Shanghai, China; Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Fudan Minhang Academic Health System, And Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 200032, China.
| | - Christiane J Bruns
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology (CIO) Achen, Bonn, Cologne and Düsseldorf, Cologne, Germany.
| | - Haijun Zhou
- Liver Cancer Institute & Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis & Cancer Invasion, Fudan University & Ministry of Education, Shanghai, China.
| |
Collapse
|
28
|
Cheng HS, Marvalim C, Zhu P, Law CLD, Low ZYJ, Chong YK, Ang BT, Tang C, Tan NS. Kinomic profile in patient-derived glioma cells during hypoxia reveals c-MET-PI3K dependency for adaptation. Theranostics 2021; 11:5127-5142. [PMID: 33859738 PMCID: PMC8039937 DOI: 10.7150/thno.54741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/17/2021] [Indexed: 01/05/2023] Open
Abstract
Hypoxic microenvironment is a hallmark of solid tumors, especially glioblastoma. The strong reliance of glioma-propagating cells (GPCs) on hypoxia-induced survival advantages is potentially exploitable for drug development. Methods: To identify key signaling pathways for hypoxia adaptation by patient-derived GPCs, we performed a kinase inhibitor profiling by screening 188 small molecule inhibitors against 130 different kinases in normoxia and hypoxia. Potential kinase candidates were prioritized for in vitro and in vivo investigations using a ranking algorithm that integrated information from the kinome connectivity network and estimated patients' survival based on expression status. Results: Hypoxic drug screen highlighted extensive modifications of kinomic landscape and a crucial functionality of c-MET-PI3K. c-MET inhibitors diminished phosphorylation of c-MET and PI3K in GPCs subjected to hypoxia, suggesting its role in the hypoxic adaptation of GPCs. Mechanistically, the inhibition of c-MET and PI3K impaired antioxidant defense, leading to oxidative catastrophe and apoptosis. Repurposed c-MET inhibitors PF04217903 and tivantinib exhibited hypoxic-dependent drug synergism with temozolomide, resulting in reduced tumor load and growth of GPC xenografts. Detailed analysis of bulk and single-cell glioblastoma transcriptomes associates the cellular subpopulation over-expressing c-MET with inflamed, hypoxic, metastatic, and stem-like phenotypes. Conclusions: Thus, our "bench to bedside (the use of patient-derived GPCs and xenografts for basic research) and back (validation with independent glioblastoma transcriptome databases)" analysis unravels the novel therapeutic indications of c-MET and PI3K/Akt inhibitors for the treatment of glioblastoma, and potentially other cancers, in the hypoxic tumor microenvironment.
Collapse
|
29
|
Capmatinib for patients with non-small cell lung cancer with MET exon 14 skipping mutations: A review of preclinical and clinical studies. Cancer Treat Rev 2021; 95:102173. [PMID: 33740553 DOI: 10.1016/j.ctrv.2021.102173] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 12/24/2022]
Abstract
The mesenchymal-epithelial transition (MET) receptor tyrosine kinase binds the hepatocyte growth factor to activate downstream cell signaling pathways involved in cell proliferation, survival, and migration. Several genetic mechanisms can result in an aberrant activation of this receptor in cancer cells. One such activating mechanism involves the acquisition of gene mutations that cause MET exon 14 skipping (METex14) during mRNA splicing. Mutations leading to METex14 are found in approximately 3-4% of patients with non-small cell lung cancer (NSCLC). Accumulating evidence suggests that METex14 is a true, independent oncogenic driver in NSCLC, as well as being an independent prognostic factor for poorer survival in patients with NSCLC. The successes of target therapies have relied on improved understanding of the genetic alterations that lead to the dysregulation of the molecular pathways and more advanced molecular diagnostics. Multiple efforts have been made to target the MET pathway in cancer; however, real clinical progress has only occurred since the emergence of METex14 as a valid biomarker for MET inhibition. Capmatinib is a highly potent and selective type Ib inhibitor of MET. Following preclinical demonstration of activity against MET-dependent cancer cell line growth and MET-driven tumor growth in xenograft models, data from a phase 1 clinical trial showed an acceptable safety profile of capmatinib and preliminary evidence of efficacy in patients with MET-dysregulated NSCLC. The multicohort GEOMETRY mono-1 phase 2 trial reported objective response rates of 68% and 41% in treatment-naïve and in pre-treated patients with METex14 advanced NSCLC, respectively. These results have supported the approval of capmatinib by the US Food and Drug Administration for patients with metastatic NSCLC harboring METex14.
Collapse
|
30
|
Moosavi F, Giovannetti E, Peters GJ, Firuzi O. Combination of HGF/MET-targeting agents and other therapeutic strategies in cancer. Crit Rev Oncol Hematol 2021; 160:103234. [PMID: 33497758 DOI: 10.1016/j.critrevonc.2021.103234] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
MET receptor has emerged as a druggable target across several human cancers. Agents targeting MET and its ligand hepatocyte growth factor (HGF) including small molecules such as crizotinib, tivantinib and cabozantinib or antibodies including rilotumumab and onartuzumab have proven their values in different tumors. Recently, capmatinib was approved for treatment of metastatic lung cancer with MET exon 14 skipping. In this review, we critically examine the current evidence on how HGF/MET combination therapies may take advantage of synergistic effects, overcome primary or acquired drug resistance, target tumor microenvironment, modulate drug metabolism or tackle pharmacokinetic issues. Preclinical and clinical studies on the combination of HGF/MET-targeted agents with conventional chemotherapeutics or molecularly targeted treatments (including EGFR, VEGFR, HER2, RAF/MEK, and PI3K/Akt targeting agents) and also the value of biomarkers are examined. Our deeper understanding of molecular mechanisms underlying successful pharmacological combinations is crucial to find the best personalized treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
31
|
Combined crizotinib and endocrine drugs inhibit proliferation, migration, and colony formation of breast cancer cells via downregulation of MET and estrogen receptor. Med Oncol 2021; 38:8. [DOI: 10.1007/s12032-021-01458-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023]
|
32
|
Nalbach L, Roma LP, Schmitt BM, Becker V, Körbel C, Wrublewsky S, Pack M, Später T, Metzger W, Menger MM, Frueh FS, Götz C, Lin H, EM Fox J, MacDonald PE, Menger MD, Laschke MW, Ampofo E. Improvement of islet transplantation by the fusion of islet cells with functional blood vessels. EMBO Mol Med 2021; 13:e12616. [PMID: 33135383 PMCID: PMC7799357 DOI: 10.15252/emmm.202012616] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic islet transplantation still represents a promising therapeutic strategy for curative treatment of type 1 diabetes mellitus. However, a limited number of organ donors and insufficient vascularization with islet engraftment failure restrict the successful transfer of this approach into clinical practice. To overcome these problems, we herein introduce a novel strategy for the generation of prevascularized islet organoids by the fusion of pancreatic islet cells with functional native microvessels. These insulin-secreting organoids exhibit a significantly higher angiogenic activity compared to freshly isolated islets, cultured islets, and non-prevascularized islet organoids. This is caused by paracrine signaling between the β-cells and the microvessels, mediated by insulin binding to its corresponding receptor on endothelial cells. In vivo, the prevascularized islet organoids are rapidly blood-perfused after transplantation by the interconnection of their autochthonous microvasculature with surrounding blood vessels. As a consequence, a lower number of islet grafts are required to restore normoglycemia in diabetic mice. Thus, prevascularized islet organoids may be used to improve the success rates of clinical islet transplantation.
Collapse
Affiliation(s)
- Lisa Nalbach
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Leticia P Roma
- Biophysics DepartmentCenter for Human and Molecular BiologySaarland UniversityHomburg/SaarGermany
| | - Beate M Schmitt
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Vivien Becker
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Christina Körbel
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Selina Wrublewsky
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Mandy Pack
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Thomas Später
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Wolfgang Metzger
- Department of Trauma, Hand and Reconstructive SurgerySaarland UniversityHomburgGermany
| | - Maximilian M Menger
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
- Departement of Trauma and Reconstructive SurgeryEberhar Karls University TuebingenTuebingenGermany
| | - Florian S Frueh
- Division of Plastic Surgery and Hand SurgeryUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Claudia Götz
- Medical Biochemistry and Molecular BiologySaarland UniversityHomburgGermany
| | - Haopeng Lin
- Department of PharmacologyAlberta Diabetes InstituteUniversity of AlbertaEdmontonABCanada
| | - Joseline EM Fox
- Department of PharmacologyAlberta Diabetes InstituteUniversity of AlbertaEdmontonABCanada
| | - Patrick E MacDonald
- Department of PharmacologyAlberta Diabetes InstituteUniversity of AlbertaEdmontonABCanada
| | - Michael D Menger
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Matthias W Laschke
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental SurgerySaarland UniversityHomburg/SaarGermany
| |
Collapse
|
33
|
Wang J, Bai Y, Yin S, Cui J, Zhang Y, Wang X, Zhang F, Li H, Tang Y, Wang J. Circadian clock gene BMAL1 reduces urinary calcium oxalate stones formation by regulating NRF2/HO-1 pathway. Life Sci 2020; 265:118853. [PMID: 33278384 DOI: 10.1016/j.lfs.2020.118853] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 02/08/2023]
Abstract
Calcium oxalate stones are closely related to oxalate metabolism and oxidative stress injury. Normal metabolism homeostasis and tissue repair are often affected by the biological rhythm, which plays an indispensable role in maintaining the homeostasis of the organism. Nuclear factor erythroid 2-related factor/heme oxygenase-1 (NRF2/HO-1) is one pathway related to oxidative stress injury in human body. Normal operation of this pathway is conducive to the resistance against oxidative stress-related injury. This study was mainly aimed to explore whether the rhythm gene "brain and muscle ARNT-like 1" (BMAL1) was involved in regulating oxidative stress-related NRF2/HO-1 pathway to reduce the formation of urinary calcium oxalate stones. In vitro experiment found that the activation of NRF2/HO-1 can significantly reduce the oxalate-induced oxidative damage and urinary calcium oxalate stone formation, and the relative expression of BMAL1 was increased. Then overexpression of circadian gene BMAL1 can activate the NRF2/HO-1 pathway and reduce the oxalate-induced oxidative damage. In the hyperoxaluria animal model, the BMAL1 expression level decreased obviously, and the production of calcium oxalate stones was significantly reduced after activating NRF2/HO-1. Finally, we further verified the BMAL1 expression in blood samples from the patients, and analysis of several single nucleotide polymorphisms showed BMAL1 was related to calcium oxalate stones. Therefore, maintaining normal biorhythms and appropriately intervening related rhythm genes and their downstream antioxidant pathways may play an important role in the prevention and postoperative recurrence of urinary calcium oxalate calculi, which may open up new directions for the treatment of urinary calculi.
Collapse
Affiliation(s)
- Jiahao Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yunjin Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Yin
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianwei Cui
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoming Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Facai Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yin Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| | - Jia Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
34
|
Thakur S, Sarkar B, Dhiman M, Mantha AK. Organophosphate-pesticides induced survival mechanisms and APE1-mediated Nrf2 regulation in non-small-cell lung cancer cells. J Biochem Mol Toxicol 2020; 35:e22640. [PMID: 33078895 DOI: 10.1002/jbt.22640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/11/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022]
Abstract
Epidemiological and molecular studies have indicated that environmental exposure to organophosphate pesticides (OPPs) is associated with increased cancer risk; however, the underlying molecular mechanisms still need to be explained. Increasing cancer incidence is linked to OPPs-induced oxidative stress (OS). Our study evaluates monocrotophos (MCP) and chlorpyrifos (CP)-induced OS responses and apurinic/apyrimidinic endonuclease 1 (APE1) role in human non-small-cell lung cancer (NSCLC) cells. Our prior study has implicated OPPs-induced base excision repair (BER)-pathway dysregulation and APE1-mediated regulation of transcription factor (TF) c-jun in A549 cells. We further investigated the effects of MCP and CP on apoptosis, proliferation, and APE1's redox-regulation of nuclear factor-like 2 (Nrf2). Data demonstrates that MCP and CP at subtoxic concentrations induced reactive oxygen species generation and oxidative DNA base damage 8-oxo-dG lesions in NCI-H1299 cells. CP moderately upregulated the apoptosis-inducing factor (AIF) in A549 cells, however, it did not trigger other pro-apoptotic factors viz. caspase-9 and caspase-3, suggesting early caspase-independent apoptosis. However, dose-dependent AIF-downregulation was observed for MCP treatment. Furthermore, CP and MCP treatments upregulated proliferating cell nuclear antigen levels. Immunofluorescent confocal imaging showed the colocalization of APE1 with Nrf2 in 10 µM CP- and MCP-treated NCI-H1299 cells. Immunoprecipitation confirmed that APE1 and Nrf2 physically interacted, indicating the role of APE1-mediated Nrf2 activation following OPPs treatment. This study suggests that low concentration MCP and CP exposure generates OS along with DNA damage, and modulates apoptosis, and APE1-mediated Nrf2 activation, which might be considered as the possible mechanism promoting lung cancer cell survival, suggesting that APE1 may have the potential to become a therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Shweta Thakur
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | - Bibekananda Sarkar
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
- Department of Zoology, B.S.S. College (affiliated to the B. N. Mandal University, Madhepura, Bihar), Supaul, Bihar, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Anil K Mantha
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
35
|
Jaganjac M, Milkovic L, Gegotek A, Cindric M, Zarkovic K, Skrzydlewska E, Zarkovic N. The relevance of pathophysiological alterations in redox signaling of 4-hydroxynonenal for pharmacological therapies of major stress-associated diseases. Free Radic Biol Med 2020; 157:128-153. [PMID: 31756524 DOI: 10.1016/j.freeradbiomed.2019.11.023] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
Abstract
Modern analytical methods combined with the modern concepts of redox signaling revealed 4-hydroxy-2-nonenal (4-HNE) as particular growth regulating factor involved in redox signaling under physiological and pathophysiological circumstances. In this review current knowledge of the relevance of 4-HNE as "the second messenger of reactive oxygen species" (ROS) in redox signaling of representative major stress-associated diseases is briefly summarized. The findings presented allow for 4-HNE to be considered not only as second messenger of ROS, but also as one of fundamental factors of the stress- and age-associated diseases. While standard, even modern concepts of molecular medicine and respective therapies in majority of these diseases target mostly the disease-specific symptoms. 4-HNE, especially its protein adducts, might appear to be the bioactive markers that would allow better monitoring of specific pathophysiological processes reflecting their complexity. Eventually that could help development of advanced integrative medicine approach for patients and the diseases they suffer from on the personalized basis implementing biomedical remedies that would optimize beneficial effects of ROS and 4-HNE to prevent the onset and progression of the illness, perhaps even providing the real cure.
Collapse
Affiliation(s)
- Morana Jaganjac
- Qatar Analytics & BioResearch Lab, Anti Doping Lab Qatar, Sport City Street, Doha, Qatar
| | - Lidija Milkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Div. of Molecular Medicine, Bijenicka 54, Zagreb, Croatia
| | - Agnieszka Gegotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Marina Cindric
- University of Zagreb, School of Medicine, Div. of Pathology, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| | - Kamelija Zarkovic
- University of Zagreb, School of Medicine, Div. of Pathology, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| | - Elzbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Neven Zarkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Div. of Molecular Medicine, Bijenicka 54, Zagreb, Croatia.
| |
Collapse
|
36
|
Carnosic acid increases sorafenib-induced inhibition of ERK1/2 and STAT3 signaling which contributes to reduced cell proliferation and survival of hepatocellular carcinoma cells. Oncotarget 2020; 11:3129-3143. [PMID: 32913557 PMCID: PMC7443370 DOI: 10.18632/oncotarget.27687] [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: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 11/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has increasing worldwide incidence but when unresectable lacks curative options. Treatment with a kinase inhibitor Sorafenib (Sf), while initially effective, results in only short increases in patient survival, thus there is a need for improved treatment regimens. Numerous treatment regimens have been explored wherein Sf is combined with other agents, such as non-toxic botanicals like Curcumin or Silibinin. Recently, we have shown that carnosic acid (CA), a component of the food preservative Rosemary Extract, can markedly enhance the cytotoxic actions of Sf in several cell lines derived from HCC, but not in the cell line Hu1545 derived from normal hepatocytes. CA has been shown to enhance Sf-induced cell death in the neoplastic cell lines, principally due to the composite of increased apoptosis and cytotoxic autophagy. In the present study we focused on the mechanisms that underlie the reduced proliferation and survival of HCC cells when CA is added to Sf and how this relates to the increase in Sf-induced DNA damage as well as to the elevation of cytoplasmic levels of reactive oxygen species (ROS). Importantly, the elevation of ROS levels induced by Sf was increased by adding CA. We found that CA enhanced Sf-induced prolongation of cell cycle, and the overall decrease in cell growth was associated with reduced activation of both STAT3 transcription factor (TF) and extracellular signal-regulated protein kinase (Erk)1/2. Our data suggest that a regimen incorporating CA, an inexpensive and non-toxic food additive, in the treatment of advanced HCC merits clinical evaluation.
Collapse
|
37
|
Sabarwal A, Chakraborty S, Mahanta S, Banerjee S, Balan M, Pal S. A Novel Combination Treatment with Honokiol and Rapamycin Effectively Restricts c-Met-Induced Growth of Renal Cancer Cells, and also Inhibits the Expression of Tumor Cell PD-L1 Involved in Immune Escape. Cancers (Basel) 2020; 12:cancers12071782. [PMID: 32635337 PMCID: PMC7408055 DOI: 10.3390/cancers12071782] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
The mTOR inhibitor Rapamycin has tumor inhibitory properties; and it is also used as an immunosuppressive agent after organ transplantation. However, prolonged Rapamycin treatment re-activates Akt and can promote cancer growth. Honokiol is a natural compound with both anti-tumorigenic and anti-inflammatory properties. Here, we assessed the anti-tumor effects of Rapamycin and Honokiol combination in renal cell carcinoma (RCC). Receptor tyrosine kinase c-Met-mediated signaling plays a major role in RCC growth. We observed that compared with Rapamycin alone, Rapamycin + Honokiol combination can effectively down-regulate c-Met-induced Akt phosphorylation in renal cancer cells; and it markedly inhibited Ras activation and cell proliferation and promoted G1 phase cell cycle arrest. The combination treatment significantly induced ROS generation and cancer cell apoptosis even when c-Met is activated. Importantly, Honokiol, but not Rapamycin, decreased c-Met-induced expression of the co-inhibitory molecule PD-L1, implied in the immune escape of renal cancer cells. In mouse renal cancer cells and Balb/c splenocytes co-culture assay, Rapamycin + Honokiol markedly potentiated immune-cell-mediated killing of cancer cells, possibly through the down-regulation of PD-L1. Together, Honokiol can effectively overcome the limitation of Rapamycin treatment alone; and the combination treatment can markedly restrict the growth of RCC, with particular importance to post-transplantation renal cancer.
Collapse
Affiliation(s)
- Akash Sabarwal
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.S.); (S.C.); (S.M.); (S.B.); (M.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Samik Chakraborty
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.S.); (S.C.); (S.M.); (S.B.); (M.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Simran Mahanta
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.S.); (S.C.); (S.M.); (S.B.); (M.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Selina Banerjee
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.S.); (S.C.); (S.M.); (S.B.); (M.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Murugabaskar Balan
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.S.); (S.C.); (S.M.); (S.B.); (M.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Soumitro Pal
- Division of Nephrology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.S.); (S.C.); (S.M.); (S.B.); (M.B.)
- Harvard Medical School, Boston, MA 02115, USA
- Correspondence: ; Tel.: +1-617-919-2989
| |
Collapse
|
38
|
Coskun ZM, Ersoz M, Gecili M, Ozden A, Acar A. Cytotoxic and apoptotic effects of ethanolic propolis extract on C6 glioma cells. ENVIRONMENTAL TOXICOLOGY 2020; 35:768-773. [PMID: 32061154 DOI: 10.1002/tox.22911] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 06/10/2023]
Abstract
Propolis is a natural resinous substance obtained from beehives, and emerging evidence supports that it has antitumor, antiinflammatory, antioxidant, and antimicrobial activities. The aim of the study is to examine the cytotoxic, antioxidant, and apoptotic features of ethanolic propolis extract (PE) on C6 glioma cells. The cells were treated with ethanolic PE at various concentrations for 24 hours, after which the total antioxidant status (TAS) and total oxidant status; malondialdehyde, protein carbonyl, 8-hydroxy-2'-deoxyguanosine, and glutathione (GSH) levels; Cu/Zn-superoxide dismutase (Cu/Zn-SOD) activity; and apoptotic markers were measured. Ethanolic PE at 100, 250, and 500 μg/mL concentrations showed optimal activity on C6 glioma cells. TAS and GSH levels were significantly increased in C6 glioma cells treated with 100 and 500 μg/mL PE compared to control cells (P < .05). Similarly, the activity of Cu/Zn-SOD was higher in C6 glioma cells treated with 250 or 500 μg/mL ethanolic PE compared to control cells (P < .05), as was the caspase-3 mRNA expression level. The highest levels of caspase-8 and -9 expression were in C6 glioma cells treated with 500 μg/mL PE. Collectively, our results indicate that ethanolic PE has cytotoxic and apoptotic effects on C6 glioma cells. Furthermore, it may provide a protective role in the antioxidant defense system. PE shows potential for development as a natural antioxidant and apoptotic agent for the treatment of brain tumors.
Collapse
Affiliation(s)
- Zeynep M Coskun
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Demiroglu Bilim University, Istanbul, Turkey
| | - Melike Ersoz
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Demiroglu Bilim University, Istanbul, Turkey
| | - Melike Gecili
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Demiroglu Bilim University, Istanbul, Turkey
| | - Aytek Ozden
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Demiroglu Bilim University, Istanbul, Turkey
| | - Aynur Acar
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Demiroglu Bilim University, Istanbul, Turkey
| |
Collapse
|
39
|
Artemisinin attenuates early renal damage on diabetic nephropathy rats through suppressing TGF-β1 regulator and activating the Nrf2 signaling pathway. Life Sci 2020; 256:117966. [PMID: 32535079 DOI: 10.1016/j.lfs.2020.117966] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/17/2022]
Abstract
AIM The present study aims to investigate the protective effects of artemisinin (ATZ) on early renal damage in experimental diabetic rats and its probable mechanism. METHODS Models of diabetic nephropathy (DN) rats was established utilizing streptozotocin (STZ)-injection intraperitoneally (55 mg/kg) method. All rats were subsequently divided into normal control group, model group and ATZ (25, 50, 75 mg/kg) group randomly. Biochemical parameters including body weight, kidney index, blood glucose, 24 h UAER, Scr, BUN, T-SOD, GSH-Px and MDA were comprehensively determined after 8-week consecutive administrations. HE and PAS stainings were performed to observe the histopathological alterations of kidney. Western blot was conducted to detect the expressions of TGF-β1, Nrf2, HQ-1 and NQO1. KEY FINDINGS ATZ at three concentrations in ATZ group significantly increased the body weight. Biochemical parameters altered significantly between model group and ATZ group. Moreover, ATZ inhibited TGF-β1 protein expression and activated the Nrf2 signaling pathway. Pathological histology results revealed the alterations including mesangial cells proliferation, thickness of glomerular capillary basement membrane, extracellular matrix (ECM) and the 24 h UAER. Western blot analysis demonstrated the increase of antioxidant proteins HO-1 and NQO1 and Nrf2-related proteins. SIGNIFICANCE ATZ could reduce early renal oxidative stress damage in DN rats by inhibiting TGF-β1 protein expression in kidney tissues as well as activating the Nrf2 signaling pathway and enhancing the expression of antioxidant proteins, thereby exerting the protective effects on DN kidney. The current study is the first report of ATZ on attenuating effects on kidney of DN rats, which could lay solid theoretical foundations on clinical application of ATZ to treat DN.
Collapse
|
40
|
Erkan H, Telci D, Dilek O. Design of Fluorescent Probes for Bioorthogonal Labeling of Carbonylation in Live Cells. Sci Rep 2020; 10:7668. [PMID: 32376913 PMCID: PMC7203098 DOI: 10.1038/s41598-020-64790-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
With the rapid development of chemical biology, many diagnostic fluorophore-based tools were introduced to specific biomolecules by covalent binding. Bioorthogonal reactions have been widely utilized to manage challenges faced in clinical practice for early diagnosis and treatment of several tumor samples. Herein, we designed a small molecule fluorescent-based biosensor, 2Hydrazine-5nitrophenol (2Hzin5NP), which reacts with the carbonyl moiety of biomolecules through bioorthogonal reaction, therefore can be utilized for the detection of biomolecule carbonylation in various cancer cell lines. Our almost non-fluorescent chemical probe has a fast covalent binding with carbonyl moieties at neutral pH to form a stable fluorescent hydrazone product leading to a spectroscopic alteration in live cells. Microscopic and fluorometric analyses were used to distinguish the exogenous and endogenous ROS induced carbonylation profile in human dermal fibroblasts along with A498 primary site and ACHN metastatic site renal cell carcinoma (RRC) cell lines. Our results showed that carbonylation level that differs in response to exogenous and endogenous stress in healthy and cancer cells can be detected by the newly synthesized bioorthogonal fluorescent probe. Our results provide new insights into the development of novel bioorthogonal probes that can be utilized in site-specific carbonylation labeling to enhance new diagnostic approaches in cancer.
Collapse
Affiliation(s)
- Hazel Erkan
- Department of Biotechnology, Yeditepe University, Istanbul, 34755, Turkey
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Department of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse, 6/4 8010, Graz, Austria
| | - Dilek Telci
- Department of Biotechnology, Yeditepe University, Istanbul, 34755, Turkey.
| | - Ozlem Dilek
- Department of Chemistry, University of Saint Joseph, West Hartford, 06117, Connecticut, USA.
| |
Collapse
|
41
|
Qi M, Sun LA, Jiang XC, Han YL, Wang L, Niu WH, Fei MX, Zhaba WD, Zheng LR, Zhou ML. FOXO4 expression associates with glioblastoma development and FOXO4 expression inhibits cell malignant phenotypes in vitro and in vivo. Life Sci 2020; 247:117436. [PMID: 32070707 DOI: 10.1016/j.lfs.2020.117436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 02/13/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIM Forkhead box protein O4 (FOXO4) is a transcription factor, and aberrant FOXO4 expression is associated with development of various human cancers. This study explored the role of FOXO4 in glioma in vitro and in vivo. METHODS FOXO4 expression was first assessed in normal brain tissues, low-grade glioma, glioblastoma multiforme (GBM), normal human astrocytes (HA), and GBM cell lines, while manipulation of FOXO4 expression in glioma cell lines was assessed using qRT-PCR, Western blot, and cell viability CCK-8, Transwell, and a nude mouse subcutaneous xenograft assays. KEY FINDINGS The data showed downregulated FOXO4 expression in GBM tissues and cell lines. FOXO4 overexpression induced by transfection with FOXO4 cDNA significantly inhibited GBM cell proliferation, migration, and invasion, but increased tumor cells to undergo apoptosis in vitro, while suppressed growth of GBM cell subcutaneous xenografts in nude mice. In conclusion, FOXO4 possesses an anti-cancer glioma activity, which could be a novel target for future control of GBM.
Collapse
Affiliation(s)
- Min Qi
- Anatomy Experimental Center, Wannan Medical College, Wuhu 241002, Anhui, China; Graduate School of Wannan Medical College, Wuhu 241002, Anhui, China; Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Le-An Sun
- Graduate School of Wannan Medical College, Wuhu 241002, Anhui, China; Department of Neurosurgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, Anhui, China
| | - Xiao-Chun Jiang
- Department of Neurosurgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, Anhui, China
| | - Yan-Ling Han
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Lin Wang
- Department of Pathophysiology, Basic Medical College, Wannan Medical College, Wuhu 241002, Anhui, China
| | - Wen-Hao Niu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Mao-Xing Fei
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Wang-Dui Zhaba
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, China
| | - Lan-Rong Zheng
- Department of Pathology, Basic Medical College, Wannan Medical College, Wuhu 241002, Anhui, China.
| | - Meng-Liang Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, China.
| |
Collapse
|
42
|
Aggarwal V, Tuli HS, Varol A, Thakral F, Yerer MB, Sak K, Varol M, Jain A, Khan MA, Sethi G. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules 2019; 9:735. [PMID: 31766246 PMCID: PMC6920770 DOI: 10.3390/biom9110735] [Citation(s) in RCA: 676] [Impact Index Per Article: 112.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) play a pivotal role in biological processes and continuous ROS production in normal cells is controlled by the appropriate regulation between the silver lining of low and high ROS concentration mediated effects. Interestingly, ROS also dynamically influences the tumor microenvironment and is known to initiate cancer angiogenesis, metastasis, and survival at different concentrations. At moderate concentration, ROS activates the cancer cell survival signaling cascade involving mitogen-activated protein kinase/extracellular signal-regulated protein kinases 1/2 (MAPK/ERK1/2), p38, c-Jun N-terminal kinase (JNK), and phosphoinositide-3-kinase/ protein kinase B (PI3K/Akt), which in turn activate the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), matrix metalloproteinases (MMPs), and vascular endothelial growth factor (VEGF). At high concentrations, ROS can cause cancer cell apoptosis. Hence, it critically depends upon the ROS levels, to either augment tumorigenesis or lead to apoptosis. The major issue is targeting the dual actions of ROS effectively with respect to the concentration bias, which needs to be monitored carefully to impede tumor angiogenesis and metastasis for ROS to serve as potential therapeutic targets exogenously/endogenously. Overall, additional research is required to comprehend the potential of ROS as an effective anti-tumor modality and therapeutic target for treating malignancies.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Histopathology, Post Graduate Institute of Medical Education and Research (PGIMER), Punjab, Chandigarh 160012, India;
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India;
| | - Ayşegül Varol
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskişehir TR26470, Turkey;
| | - Falak Thakral
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India;
| | - Mukerrem Betul Yerer
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Turkey;
| | | | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Kotekli Campus, Mugla Sitki Kocman University, Mugla TR48000, Turkey;
| | - Aklank Jain
- Department of Animal Sciences, Central University of Punjab, City Campus, Mansa Road, Bathinda 151001, India;
| | - Md. Asaduzzaman Khan
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China;
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| |
Collapse
|