1
|
Li J, Gu T, Hu S, Jin B. Anti-proliferative effect of Cannabidiol in Prostate cancer cell PC3 is mediated by apoptotic cell death, NFκB activation, increased oxidative stress, and lower reduced glutathione status. PLoS One 2023; 18:e0286758. [PMID: 37796968 PMCID: PMC10553363 DOI: 10.1371/journal.pone.0286758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/23/2023] [Indexed: 10/07/2023] Open
Abstract
Prostate cancer is the second most frequent cancer diagnosed in men in the world today. Almost all prostate cancers are adenocarcinomas and develop from gland cells. We used the PC3 prostate cancer cell line, which is well studied and derived from a bone metastasis of a grade IV prostatic adenocarcinoma. Cannabidiol (CBD), a major non-psychoactive constituent of cannabis, is a cannabinoid with anti-tumor properties but its effects on prostate cancer cells are not studied in detail. Here, we found cannabidiol decreased prostate cancer cell (PC3) viability up to 37.25% and induced apoptotic cell death in a time and dose-dependent manner. We found that CBD activated the caspases 3/7 pathways and increased DNA fragmentation. Furthermore, we observed an increase of pro-apoptotic genes Bax, an increased level of reactive oxygen species, lower reduced glutathione level, and altered mitochondrial potential in response to CBD treatment leading to lower cellular ATP. Overall, our results suggest that CBD may be effective against prostate cancer cells.
Collapse
Affiliation(s)
- Jie Li
- Department of Urology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
- Department of Urology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Tengfei Gu
- Department of Urology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Shengping Hu
- Department of Urology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Baiye Jin
- Department of Urology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou, China
| |
Collapse
|
2
|
Oronsky B, Takahashi L, Gordon R, Cabrales P, Caroen S, Reid T. RRx-001: a chimeric triple action NLRP3 inhibitor, Nrf2 inducer, and nitric oxide superagonist. Front Oncol 2023; 13:1204143. [PMID: 37313460 PMCID: PMC10258348 DOI: 10.3389/fonc.2023.1204143] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/18/2023] [Indexed: 06/15/2023] Open
Abstract
RRx-001 is a shape shifting small molecule with Fast Track designation for the prevention/amelioration of chemoradiation-induced severe oral mucositis (SOM) in newly diagnosed Head and Neck cancer. It has been intentionally developed or "engineered" as a chimeric single molecular entity that targets multiple redox-based mechanisms. Like an antibody drug conjugate (ADC), RRx-001 contains, at one end a "targeting" moiety, which binds to the NLRP3 inflammasome and inhibits it as well as Kelch-like ECH-associated protein 1 (KEAP1), the negative regulator of Nrf2, and, at the other end, a conformationally constrained, dinitro containing 4 membered ring, which fragments under conditions of hypoxia and reduction to release therapeutically active metabolites i.e., the payload. This "payload", which is delivered specifically to hypoperfused and inflamed areas, includes nitric oxide, nitric oxide related species and carbon-centered radicals. As observed with ADCs, RRx-001 contains a backbone amide "linker" attached to a binding site, which correlates with the Fab region of an antibody, and to the dinitroazetidine payload, which is microenvironmentally activated. However, unlike ADCs, whose large size impacts their pharmacokinetic properties, RRx-001 is a nonpolar small molecule that easily crosses cell membranes and the blood brain barrier (BBB) and distributes systemically. This short review is organized around the de novo design and in vivo pro-oxidant/pro-inflammatory and antioxidant/anti-inflammatory activity of RRx-001, which, in turn, depends on the reduced to oxidized glutathione ratio and the oxygenation status of tissues.
Collapse
Affiliation(s)
- Bryan Oronsky
- Drug Development, EpicentRx, Torrey Pines, CA, United States
| | - Lori Takahashi
- Drug Development, EpicentRx, Torrey Pines, CA, United States
| | - Richard Gordon
- Department of Translational Neuroscience, University of Queensland Centre for Clinical Research, Brisbane, QLD, Australia
| | - Pedro Cabrales
- Department of Bioengineering, University of California at San Diego, La Jolla, CA, United States
| | - Scott Caroen
- Drug Development, EpicentRx, Torrey Pines, CA, United States
| | - Tony Reid
- Drug Development, EpicentRx, Torrey Pines, CA, United States
| |
Collapse
|
3
|
Yang C, Mu G, Zhang Y, Gao Y, Zhang W, Liu J, Zhang W, Li P, Yang L, Yang Z, Gao J, Liu J. Supramolecular Nitric Oxide Depot for Hypoxic Tumor Vessel Normalization and Radiosensitization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202625. [PMID: 35906003 DOI: 10.1002/adma.202202625] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/15/2022] [Indexed: 06/15/2023]
Abstract
In cancer radiotherapy, the lack of fixed DNA damage by oxygen in hypoxic microenvironment of solid tumors often leads to severe radioresistance. Nitric oxide (NO) is a potent radiosensitizer that acts in two ways. It can directly react with the radical DNA thus fixing the damage. It also normalizes the abnormal tumor vessels, thereby increasing blood perfusion and oxygen supply. To achieve these functions, the dosage and duration of NO treatment need to be carefully controlled, otherwise it will lead to the exact opposite outcomes. However, a delivery method that fulfills both requirements is still lacking. A NO depot is designed for the control of NO releasing both over quantity and duration for hypoxic tumor vessel normalization and radiosensitization. In B16-tumor-bearing mice, the depot can provide low dosage NO continuously and release large amount of NO immediately before irradiation for a short period of time. These two modes of treatment work in synergy to reverse the radioresistance of B16 tumors more efficiently than releasing at single dosage.
Collapse
Affiliation(s)
- Cuihong Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Ganen Mu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Yang Gao
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Wenxue Zhang
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Wenwen Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Paiyun Li
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Lijun Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| |
Collapse
|
4
|
Xu Y, Li H, Xu S, Liu X, Lin J, Chen H, Yuan Z. Light-Triggered Fluorescence Self-Reporting Nitric Oxide Release from Coumarin Analogues for Accelerating Wound Healing and Synergistic Antimicrobial Applications. J Med Chem 2021; 65:424-435. [PMID: 34918930 DOI: 10.1021/acs.jmedchem.1c01591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nitric oxide (NO) has an important class of endogenous diatomic molecules that play a key regulatory role in many physiological and biochemical processes. However, the type of nitrosamine NO donor stimulated by light has many advantages compared to the conventional NO donors such as diazeniumdiolates and S-nitrosothiols compounds, including easy synthesis, good stability, and controllable release. In addition, NO release can be regulated by light induction with a built-in calibration mechanism fluorescence. Here, we report that the migration and proliferation of human umbilical vein vascular endothelial cells could be accelerated by the light-triggered NO donors, leading to the angiogenesis. Meanwhile, the screened NO donor 3a with Levofloxacin (Lev) showed synergistic effects to eradicate Methicillin-resistant Staphylococcus aureus (MRSA) biofilms in vitro and treat bacteria-infected wound in vivo.
Collapse
Affiliation(s)
- Yue Xu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Hua Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Shufen Xu
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, China.,Department of Oncology, Second Clinical Medical College of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Xian Liu
- The Hong Kong Polytechnic University, 11 Yuk Choi Road, Hung Hom, Kowloon 999077, Hong Kong, China
| | - Jingjing Lin
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 24 Tongjia Lane, Gulou District, Nanjing 210009, China
| |
Collapse
|
5
|
Guo X, Yu H, Shen W, Cai R, Li Y, Li G, Zhao W, Wang S. Synthesis and biological evaluation of NO-donor containing photosensitizers to induce ferroptosis of cancer cells. Bioorg Chem 2021; 116:105355. [PMID: 34592689 DOI: 10.1016/j.bioorg.2021.105355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 11/26/2022]
Abstract
Photodynamic therapy (PDT) is a non-invasive treatment method for tumors by exciting photosensitizers (PS) upon light irradiation to generate cytotoxic reactive oxygen species (ROS). However, the low oxygen concentration near the tumor tissue limits the therapeutic effect of PDT. Herein, we synthesized six chlorin e6 derivatives containing NO-donors to enhance their antitumor activity by synergistic effect of ROS and NO. The results revealed that the new NO-donor containing photosensitizers (PS-NO) exhibited more potent photodynamic activity than chlorin e6, and the introduction of NO donor moieties to chlorin e6 increased the level of NO and ROS in cells. The addition of Ferrostatin-1, a ferroptosis inhibitor, markedly reduced the photodynamic activity of PS-NO as well as the level of NO and ROS in cells. Mechanism studies further showed that PS-NO could reduce intracellular GSH level, inhibit GPX4 activity and promote malondialdehyde (MDA) accumulation upon light irradiation, which suggested the ferroptosis mechanism underlying the PDT effect of PS-NO.
Collapse
Affiliation(s)
- Xiuhan Guo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, Liaoning, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, Zhejiang, China.
| | - Haoze Yu
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Wanjie Shen
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Rui Cai
- Center of Analysis and Research, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Yueqing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, Liaoning, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, Zhejiang, China
| | - Guangzhe Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Shisheng Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, Liaoning, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, Zhejiang, China.
| |
Collapse
|
6
|
Small molecule inhibitors and stimulators of inducible nitric oxide synthase in cancer cells from natural origin (phytochemicals, marine compounds, antibiotics). Biochem Pharmacol 2020; 176:113792. [PMID: 31926145 DOI: 10.1016/j.bcp.2020.113792] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023]
Abstract
Nitric oxide synthases (NOS) are a family of isoforms, which generate nitric oxide (NO). NO is one of the smallest molecules in nature and acts mainly as a potent vasodilator. It participates in various biological processes ranging from physiological to pathological conditions. Inducible NOS (iNOS, NOS2) is a calcium-independent and inducible isoform. Despite high iNOS expression in many tumors, the role of iNOS is still unclear and complex with both enhancing and prohibiting actions in tumorigenesis. Nature presents a broad variety of natural stimulators and inhibitors, which may either promote or inhibit iNOS response. In the present review, we give an overview of iNOS-modulating agents with a special focus on both natural and synthetic molecules and their effects in related biological processes. The role of iNOS in physiological and pathological conditions is also discussed.
Collapse
|
7
|
Kamm A, Przychodzen P, Kuban-Jankowska A, Jacewicz D, Dabrowska AM, Nussberger S, Wozniak M, Gorska-Ponikowska M. Nitric oxide and its derivatives in the cancer battlefield. Nitric Oxide 2019; 93:102-114. [PMID: 31541733 DOI: 10.1016/j.niox.2019.09.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/06/2019] [Accepted: 09/16/2019] [Indexed: 12/14/2022]
Abstract
Elevated levels of reactive nitrogen species, alteration in redox balance and deregulated redox signaling are common hallmarks of cancer progression and chemoresistance. However, depending on the cellular context, distinct reactive nitrogen species are also hypothesized to mediate cytotoxic activity and are thus used in anticancer therapies. We present here the dual face of nitric oxide and its derivatives in cancer biology. Main derivatives of nitric oxide, such as nitrogen dioxide and peroxynitrite cause cell death by inducing protein and lipid peroxidation and/or DNA damage. Moreover, they control the activity of important protein players within the pro- and anti-apoptotic signaling pathways. Thus, the control of intracellular reactive nitrogen species may become a sophisticated tool in anticancer strategies.
Collapse
Affiliation(s)
- Anna Kamm
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Paulina Przychodzen
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Alicja Kuban-Jankowska
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | | | | | - Stephan Nussberger
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| | - Michal Wozniak
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Magdalena Gorska-Ponikowska
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland; Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany; Euro-Mediterranean Institute of Science and Technology, Palermo, Italy.
| |
Collapse
|
8
|
Chakraborty S, Datta S, Ghosh S. Induction of autophagy under nitrosative stress: A complex regulatory interplay between SIRT1 and AMPK in MCF7 cells. Cell Signal 2019; 64:109411. [PMID: 31491460 DOI: 10.1016/j.cellsig.2019.109411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 08/30/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Induction of nitrosative stress has been observed in various cancer types and in tumor environment. However, it is still unclear how cancer cells combat the effect of nitrosative stress. The main targets of nitrosative stress in cells are cellular lipids, proteins and DNA. Autophagy or self-cleaning generates energy for cell survival under stress conditions. In the present study we investigated the role of autophagy under nitrosative stress in MCF7, a breast cancer cell line. Interestingly, we observed induction of autophagy associated with cell death when MCF7 cells were treated with NO donor compound DETA-NONOate for eight hours. While investigating the mode of cell death under nitrosative stress in MCF7 cells, it was found that it was neither apoptotic nor necrotic. Moreover, nitrosative stress did not alter mitochondrial membrane potential and cellular redox status in MCF7 cells. But we observed an increase in NAD+/NADH and a drop in NADH level in MCF7 cells following NO donor treatment. Sirtuins having NAD+ dependent deacetylase activity, play an important role in cell survival mechanisms. So we further checked the status of SIRT1 under nitrosative stress in MCF7 cells. Surprisingly, we observed an induction of SIRT1, phospho-AMPK and p53 in MCF7 cells under nitrosative stress. Interestingly, autophagy markers were down regulated in MCF7 cells upon treatment with nicotinamide, an inhibitor of SIRT1 activity and dorsomorphin, a phospho-AMPK inhibitor when treated separately under nitrosative stress. To further confirm the role of SIRT1 in the induction of autophagy associated cell death, it was knocked down using si-RNA and nitrosative stress was applied. SIRT1 knock down led to increase in MCF7 cell viability along with down regulation of autophagic markers and phospho-AMPK as well as accumulation of acetylated p53. The increase in p53 controlled DRAM1 mRNA expression in MCF7 cells under nitrosative stress further confirmed a complex interplay between p53, SIRT1 and AMPK under nitrosative stress in MCF7 cells. Altogether our work for the first time suggests a complex inter-twined partnership between AMPK, SIRT1 and p53 in regulating autophagy in response to nitrosative stress in MCF7 cells.
Collapse
Affiliation(s)
- Subhamoy Chakraborty
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sampurna Datta
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sanjay Ghosh
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
9
|
Metabolic pathways of L-arginine and therapeutic consequences in tumors. Adv Med Sci 2019; 64:104-110. [PMID: 30605863 DOI: 10.1016/j.advms.2018.08.018] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 06/03/2018] [Accepted: 08/31/2018] [Indexed: 02/06/2023]
Abstract
Difference in the metabolism of normal and cancer cells inspires to search for new, more specific and less toxic therapies than those currently used. The development of tumors is conditioned by genetic changes in cancer-transformed cells, immunological tolerance and immunosuppression. At the initial stages of carcinogenesis, the immune system shows anti-tumor activity, however later, cancer disrupts the function of Th1/Th17/Th2 lymphocytes by regulatory T (Treg) cells, tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) and finally causes immunosuppression. Recently, much attention has been devoted to the influence of l-arginine metabolism disorders on both carcinogenesis and the immune system. l-Arginine is essential for the maturation of the T cell receptor zeta (TCRζ), and its absence deprives T-cells of the ability to interact with tumor antigens. MDSCs deplete l-arginine due to a high expression of arginase 1 (ARG1) and their number increases 4-10 times depending on the type of the cancer. L-Arginine has been shown to be essential for the survival and progression of arginine auxotrophic tumors. However, the progression of arginine non-auxotrophic tumors is independent of exogenous l-arginine, because these tumors have arginine-succinate synthetase (ASS1) activity and are available to produce l-arginine from citrulline. Clinical studies have confirmed the high efficacy of arginine auxotrophic tumors therapy based on the elimination of l-arginine. However, l-arginine supplementation may improve the results of treatment of patients with arginine non-auxotrophic cancer. This review is an attempt to explain the seemingly contradictory results of oncological therapies based on the deprivation or supplementation of l-arginine.
Collapse
|
10
|
Kashfi K. The dichotomous role of H 2S in cancer cell biology? Déjà vu all over again. Biochem Pharmacol 2018; 149:205-223. [PMID: 29397935 PMCID: PMC5866221 DOI: 10.1016/j.bcp.2018.01.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/17/2018] [Indexed: 02/09/2023]
Abstract
Nitric oxide (NO) a gaseous free radical is one of the ten smallest molecules found in nature, while hydrogen sulfide (H2S) is a gas that bears the pungent smell of rotten eggs. Both are toxic yet they are gasotransmitters of physiological relevance. There appears to be an uncanny resemblance between the general actions of these two gasotransmitters in health and disease. The role of NO and H2S in cancer has been quite perplexing, as both tumor promotion and inflammatory activities as well as anti-tumor and antiinflammatory properties have been described. These paradoxes have been explained for both gasotransmitters in terms of each having a dual or biphasic effect that is dependent on the local flux of each gas. In this review/commentary, I have discussed the major roles of NO and H2S in carcinogenesis, evaluating their dual nature, focusing on the enzymes that contribute to this paradox and evaluate the pros and cons of inhibiting or inducing each of these enzymes.
Collapse
Affiliation(s)
- Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA.
| |
Collapse
|
11
|
Shafran Y, Zurgil N, Ravid-Hermesh O, Sobolev M, Afrimzon E, Hakuk Y, Shainberg A, Deutsch M. Nitric oxide is cytoprotective to breast cancer spheroids vulnerable to estrogen-induced apoptosis. Oncotarget 2017; 8:108890-108911. [PMID: 29312577 PMCID: PMC5752490 DOI: 10.18632/oncotarget.21610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 08/17/2017] [Indexed: 12/24/2022] Open
Abstract
Estrogen-induced apoptosis has become a successful treatment for postmenopausal metastatic, estrogen receptor-positive breast cancer. Nitric oxide involvement in the response to this endocrine treatment and its influence upon estrogen receptor-positive breast cancer progression is still unclear. Nitric oxide impact on the MCF7 breast cancer line, before and after estrogen-induced apoptosis, was investigated in 3D culture systems using unique live-cell imaging methodologies. Spheroids were established from MCF7 cells vulnerable to estrogen-induced apoptosis, before and after exposure to estrogen. Spheroids derived from estrogen-treated cells exhibited extensive apoptosis levels with downregulation of estrogen receptor expression, low proliferation rate and reduced metabolic activity, unlike spheroids derived from non-treated cells. In addition to basic phenotypic differences, these two cell cluster types are diverse in their reactions to exogenous nitric oxide. A dual effect of nitric oxide was observed in the breast cancer phenotype sensitive to estrogen-induced apoptosis. Nitric oxide, at the nanomolar level, induced cell proliferation, high metabolic activity, downregulation of estrogen receptor and enhanced collective invasion, contributing to a more aggressive phenotype. Following hormone supplementation, breast cancer 3D clusters were rescued from estrogen-induced apoptosis by these low nitric oxide-donor concentrations, since nitric oxide attenuates cell death levels, upregulates survivin expression and increases metabolic activity. Higher nitric oxide concentrations (100nM) inhibited cell growth, metabolism and promoted apoptosis. These results suggest that nitric oxide, in nanomolar concentrations, may inhibit estrogen-induced apoptosis, playing a major role in hormonal therapy. Inhibiting nitric oxide activity may benefit breast cancer patients and ultimately reduce tumor recurrence.
Collapse
Affiliation(s)
- Yana Shafran
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Naomi Zurgil
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Orit Ravid-Hermesh
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Maria Sobolev
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Elena Afrimzon
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Yaron Hakuk
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Asher Shainberg
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 52900, Israel
| | - Mordechai Deutsch
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| |
Collapse
|
12
|
The Role of Nitric Oxide from Neurological Disease to Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:71-88. [PMID: 28840553 DOI: 10.1007/978-3-319-60733-7_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
13
|
Abstract
The increasing understanding of the role of nitric oxide (NO) in cancer biology has generated significant progress in the use of NO donor-based therapy to fight cancer. These advances strongly suggest the potential adoption of NO donor-based therapy in clinical practice, and this has been supported by several clinical studies in the past decade. In this review, we first highlight several types of important NO donors, including recently developed NO donors bearing a dinitroazetidine skeleton, represented by RRx-001, with potential utility in cancer therapy. Special emphasis is then given to the combination of NO donor(s) with other therapies to achieve synergy and to the hybridization of NO donor(s) with an anticancer drug/agent/fragment to enhance the activity or specificity or to reduce toxicity. In addition, we briefly describe inducible NO synthase gene therapy and nanotechnology, which have recently entered the field of NO donor therapy.
Collapse
Affiliation(s)
- Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , Nanjing 210009, P. R. China
| | - Junjie Fu
- Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University , Nanjing 211166, P.R. China
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University , Nanjing 210009, P. R. China
| |
Collapse
|
14
|
Cabrales P, Caroen S, Oronsky A, Carter C, Trepel J, Summers T, Reid T, Oronsky N, Lybeck M, Oronsky B. The macrophage stimulating anti-cancer agent, RRx-001, protects against ischemia-reperfusion injury. Expert Rev Hematol 2017; 10:575-582. [PMID: 28448172 DOI: 10.1080/17474086.2017.1324779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND RRx-001, a clinical macrophage-stimulating anti-cancer agent that also produces nitric oxide (NO) was studied in a model of ischemia-reperfusion injury. METHODS The production of NO is dependent on the oxygen tension because nitric oxide synthases convert l-arginine to NO and l-citrulline in the presence of O2. Since the P450 enzymes, which metabolize nitrate esters such as nitroglycerin are dependent on oxygen, the generation of 'exogenous' NO is also sensitive to alterations in tissue PO2. I/R injury was studied in a hamster chamber window, with compression of the periphery of the window for 1 h to induce ischemia. Animals received RRx-001 (5 mg/kg) 24 h before ischemia and sodium nitrite (10 nmols/kg) was supplemented 10 min after the start of reperfusion. Vessel diameter, blood flow, adherent leukocytes, and functional capillary density were assessed by intravital microscopy at 0.5, 2, and 24 h following the release of the ischemia. RESULTS The results demonstrated that, compared to control, RRx-001 preconditioning increased blood flow and functional capillary density, and preserved tissue viability in the absence of side effects over a sustained time period. CONCLUSION Thus, RRx-001 may serve as a long-lived protective agent during postsurgical restoration of flow and other ischemia-reperfusion associated conditions, increasing blood flow and functional capillary density as well as preserving tissue viability in the absence of side effects.
Collapse
Affiliation(s)
- Pedro Cabrales
- a Department of Bioengineering , University of California San Diego (UCSD) , La Jolla , CA , USA
| | | | | | - Corey Carter
- d Walter Reed Military Medical Center , Murtha Cancer Center , Bethesda , MD , USA
| | - Jane Trepel
- e Moores Cancer Center , University of California San Diego (UCSD) , La Jolla , CA , USA
| | - Thomas Summers
- d Walter Reed Military Medical Center , Murtha Cancer Center , Bethesda , MD , USA
| | | | | | | | | |
Collapse
|
15
|
Zou Y, Yan C, Knaus EE, Zhang H, Zhang Y, Huang Z. Discovery of phosphorodiamidate mustard-based O2-phosphorylated diazeniumdiolates with potent anticancer activity. RSC Adv 2017. [DOI: 10.1039/c7ra00401j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Diazeniumdiolates are an important class of NO donors. Herein, we describe the design, synthesis and biological evaluation of a group of phosphorodiamidate mustard-based O2-phosphorylated diazeniumdiolates.
Collapse
Affiliation(s)
- Yu Zou
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Jiangsu Key Laboratory of Drug Screening
- China Pharmaceutical University
- Nanjing 210009
| | - Chang Yan
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Jiangsu Key Laboratory of Drug Screening
- China Pharmaceutical University
- Nanjing 210009
| | - Edward E. Knaus
- Faculty of Pharmacy and Pharmaceutical Sciences
- University of Alberta
- Edmonton
- Canada
| | - Huibin Zhang
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Jiangsu Key Laboratory of Drug Screening
- China Pharmaceutical University
- Nanjing 210009
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Jiangsu Key Laboratory of Drug Screening
- China Pharmaceutical University
- Nanjing 210009
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Jiangsu Key Laboratory of Drug Screening
- China Pharmaceutical University
- Nanjing 210009
| |
Collapse
|
16
|
Fens MH, Cabrales P, Scicinski J, Larkin SK, Suh JH, Kuypers FA, Oronsky N, Lybeck M, Oronsky A, Oronsky B. Targeting tumor hypoxia with the epigenetic anticancer agent, RRx-001: a superagonist of nitric oxide generation. Med Oncol 2016; 33:85. [PMID: 27377482 DOI: 10.1007/s12032-016-0798-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/15/2016] [Indexed: 12/17/2022]
Abstract
This study reveals a novel interaction between deoxyhemoglobin, nitrite and the non-toxic compound, RRx-001, to generate supraphysiologic levels of nitric oxide (NO) in blood. We characterize the nitrite reductase activity of deoxyhemoglobin, which in the presence of bound RRx-001 reduces nitrite at a much faster rate, leading to markedly increased NO generation. These data expand on the paradigm that hemoglobin generates NO via nitrite reduction during hypoxia and ischemia when nitric oxide synthase (NOS) function is limited. Here, we demonstrate that RRx-001 greatly enhances NO generation from nitrite reduction. RRx-001 is thus the first example of a functional superagonist for nitrite reductase. We hypothesize that physiologically this reaction releases the potentially cytotoxic effector NO selectively in hypoxic tumor regions. It may be that a binary NO-H2O2 trigger is indirectly responsible for the observed tumoricidal activity of RRx-001 since NO is known to inhibit mitochondrial respiration.
Collapse
Affiliation(s)
- Marcel H Fens
- Children's Hospital Oakland Research Institute (CHORI), 5700 M.L.K. Jr Way, Oakland, CA, 94609, USA
| | - Pedro Cabrales
- Department of Bioengineering, University of California San Diego (UCSD), 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Jan Scicinski
- EpicentRx, Inc., 800 W El Camino Real, Suite 180, Mountain View, CA, 94040, USA
| | - Sandra K Larkin
- Children's Hospital Oakland Research Institute (CHORI), 5700 M.L.K. Jr Way, Oakland, CA, 94609, USA
| | - Jung H Suh
- Children's Hospital Oakland Research Institute (CHORI), 5700 M.L.K. Jr Way, Oakland, CA, 94609, USA
| | - Frans A Kuypers
- Children's Hospital Oakland Research Institute (CHORI), 5700 M.L.K. Jr Way, Oakland, CA, 94609, USA
| | - Neil Oronsky
- CFLS Data, 560 South Winchester Boulevard, San Jose, CA, 95128, USA
| | - Michelle Lybeck
- EpicentRx, Inc., 800 W El Camino Real, Suite 180, Mountain View, CA, 94040, USA
| | - Arnold Oronsky
- InterWest Partners, 2710 Sand Hill Road #200, Menlo Park, CA, 94025, USA
| | - Bryan Oronsky
- EpicentRx, Inc., 800 W El Camino Real, Suite 180, Mountain View, CA, 94040, USA.
| |
Collapse
|
17
|
Vannini F, Kashfi K, Nath N. The dual role of iNOS in cancer. Redox Biol 2015; 6:334-343. [PMID: 26335399 PMCID: PMC4565017 DOI: 10.1016/j.redox.2015.08.009] [Citation(s) in RCA: 380] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/07/2015] [Accepted: 08/10/2015] [Indexed: 01/02/2023] Open
Abstract
Nitric oxide (NO) is one of the 10 smallest molecules found in nature. It is a simple gaseous free radical whose predominant functions is that of a messenger through cGMP. In mammals, NO is synthesized by the enzyme nitric oxide synthase (NOS) of which there are three isoforms. Neuronal (nNOS, NOS1) and endothelial (eNOS, NOS3) are constitutive calcium-dependent forms of the enzyme that regulate neural and vascular function respectively. The third isoform (iNOS, NOS2), is calcium-independent and is inducible. In many tumors, iNOS expression is high, however, the role of iNOS during tumor development is very complex and quite perplexing, with both promoting and inhibiting actions having been described. This review will aim to summarize the dual actions of iNOS-derived NO showing that the microenvironment of the tumor is a contributing factor to these observations and ultimately to cellular outcomes. NO is pro- and anti-tumorigenic. High concentrations of NO maybe anti-tumorigenic. iNOS produces high concentrations of NO and relates to tumor growth or its inhibition. iNOS is associated with cytotoxicity, apoptosis and bystander anti-tumor effects. Tumor- and stromal-iNOS, and the ‘cell situation’ contribute to anti or pro-tumor effects. Dual role of iNOS is influenced by the cell situation and is environment dependent.
Collapse
Affiliation(s)
- Federica Vannini
- Department of Physiology, Pharmacology and Neuroscience, Sophie Davis School of Biomedical Education, City University of New York Medical School, New York, NY 10031, United States
| | - Khosrow Kashfi
- Department of Physiology, Pharmacology and Neuroscience, Sophie Davis School of Biomedical Education, City University of New York Medical School, New York, NY 10031, United States
| | - Niharika Nath
- Department of Life Sciences, New York Institute of Technology, NY 10023, United States.
| |
Collapse
|
18
|
NO to cancer: The complex and multifaceted role of nitric oxide and the epigenetic nitric oxide donor, RRx-001. Redox Biol 2015; 6:1-8. [PMID: 26164533 PMCID: PMC4529402 DOI: 10.1016/j.redox.2015.07.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 01/18/2023] Open
Abstract
The endogenous mediator of vasodilation, nitric oxide (NO), has been shown to be a potent radiosensitizer. However, the underlying mode of action for its role as a radiosensitizer – while not entirely understood – is believed to arise from increased tumor blood flow, effects on cellular respiration, on cell signaling, and on the production of reactive oxygen and nitrogen species (RONS), that can act as radiosensitizers in their own right. NO activity is surprisingly long-lived and more potent in comparison to oxygen. Reports of the effects of NO with radiation have often been contradictory leading to confusion about the true radiosensitizing nature of NO. Whether increasing or decreasing tumor blood flow, acting as radiosensitizer or radioprotector, the effects of NO have been controversial. Key to understanding the role of NO as a radiosensitizer is to recognize the importance of biological context. With a very short half-life and potent activity, the local effects of NO need to be carefully considered and understood when using NO as a radiosensitizer. The systemic effects of NO donors can cause extensive side effects, and also affect the local tumor microenvironment, both directly and indirectly. To minimize systemic effects and maximize effects on tumors, agents that deliver NO on demand selectively to tumors using hypoxia as a trigger may be of greater interest as radiosensitizers. Herein we discuss the multiple effects of NO and focus on the clinical molecule RRx-001, a hypoxia-activated NO donor currently being investigated as a radiosensitizer in the clinic. . NO radiosensitizes by reaction with DNA radicals, by its metabolites and by impact on the vasculature. Understanding the local and context-specific activity of NO is key for radiosensitizer development RRx-001 induces NO production under hypoxia with promising radiosensitizing activity.
Collapse
|
19
|
A549 cells adapted to high nitric oxide show reduced surface CEACAM expression and altered adhesion and migration properties. Tumour Biol 2014; 36:1871-9. [PMID: 25500969 DOI: 10.1007/s13277-014-2789-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/29/2014] [Indexed: 12/12/2022] Open
Abstract
The migration and adhesion properties of tumors affect their metastatic rate. In the present study, we investigated carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 1, 5, and 6 expression in high nitric oxide (HNO)-adapted lung cancer cells compared to parent cells. We observed high transcript levels of CEACAM 1 (4S, 4L), CEACAM 5, and CEACAM 6 in HNO cells compared to parent cells. However, the surface expression was low in HNO cells. Interestingly, the intracellular protein levels were high for these three CEACAMs. We confirmed these results with immunohistochemical experiments. Further, the adhesion and migration assays showed reduced clumping in HNO-adapted A549 (A549-HNO) cells and faster migration rates, respectively. These results document the altered adhesion and migration properties of cells adapted to HNO. Further, our studies also indicate a dynamic regulation of CEACAM protein expression and surface transport in HNO cells.
Collapse
|