1
|
Sun S, Choe J, Cho J. Photo-triggered NO release of nitrosyl complexes bearing first-row transition metals and therapeutic applications. Chem Sci 2024; 15:20155-20170. [PMID: 39583571 PMCID: PMC11580031 DOI: 10.1039/d4sc06820c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
In biological systems, nitric oxide (NO) is a crucial signaling molecule that regulates a wide range of physiological and pathological processes. Given the significance of NO, there has been considerable interest in delivering NO exogenously, particularly through light as a non-invasive therapeutic approach. However, due to the high reactivity and instability of NO under physiological conditions, directly delivering NO to targeted sites remains challenging. In recent decades, photo-responsive transition metal-nitrosyl complexes, especially based on first-row transition metals such as Mn, Fe, and Co, have emerged as efficient NO donors, offering higher delivery efficiency and quantum yields than heavy metal-nitrosyl complexes under light exposure. This review provides a comprehensive overview of current knowledge and recent developments in the field of photolabile first-row transition metal-nitrosyl complexes, focusing on the structural and electronic properties, photoreactivity, photodissociation mechanisms, and potential therapeutic applications. By consolidating the key features of photoactive nitrosyl complexes, the review offers deeper insights and highlights the potential of first-row transition metal-nitrosyl complexes as versatile tools for photo-triggered NO delivery.
Collapse
Affiliation(s)
- Seungwon Sun
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Jisu Choe
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Jaeheung Cho
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
- Graduate School of Carbon Neutrality, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| |
Collapse
|
2
|
Liang S, Xiao L, Fang Y, Chen T, Xie Y, Peng Z, Wu M, Liu Y, Xie J, Nie Y, Zhao X, Deng Y, Zhao C, Mai Y. A nanocomposite hydrogel for co-delivery of multiple anti-biofilm therapeutics to enhance the treatment of bacterial biofilm-related infections. Int J Pharm 2024; 649:123638. [PMID: 38008233 DOI: 10.1016/j.ijpharm.2023.123638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023]
Abstract
The characteristics of biofilms have exacerbated the issue of clinical antibiotic resistance, rendering it a pressing challenge in need of resolution. The combination of biofilm-dispersing agents and antibiotics can eliminate biofilms and promote healing synergistically in infected wounds. In this study, we developed a novel nanocomposite hydrogel (NC gel) comprised of the poly(lactic acid)-hyperbranched polyglycerol (PLA-HPG) based bioadhesive nanoparticles (BNPs) and a hydrophilic carboxymethyl chitosan (CS) network. The NC gel was designed to co-deliver two biofilm-dispersing agents (an NO-donor SNO, and an α-amylase Am) and an antibiotic, cefepime (Cef), utilizing a synergistic anti-biofilm mechanism in which Am loosens the matrix structure and NO promotes the release of biofilm bacteria via quorum sensing, and Cef kills bacteria. The drug-loaded NC gel (SNO/BNP/CS@Am-Cef) demonstrated sustained drug release, minimal cytotoxicity, and increased drug-bacterial interactions at the site of infection. When applied to mice infected with methicillin-resistant Staphylococcus aureus (MRSA) biofilms in vivo, SNO/BNP/CS@Am-Cef enhanced biofilm elimination and promoted wound healing compared to traditional antibiotic treatments. Our work demonstrates the feasibility of the co-delivery of biofilm-dispersing agents and antibiotics using the NC gel and presents a promising approach for the polytherapy of bacterial biofilm-related infections.
Collapse
Affiliation(s)
- Shu Liang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Lingyun Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China; Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Yixuan Fang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Tian Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuan Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Julin Xie
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Yichu Nie
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-sen University Foshan Hospital, Foshan 528000, China
| | - Xizhe Zhao
- Department of Chemistry, College of Staten Island, City University of New York, NY 10314, USA
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Chao Zhao
- Department of Chemical and Biological Engineering, Center for Convergent Biosciences and Medicine, Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL 35487, USA.
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
3
|
da Silva Filho PM, Paz IA, Nascimento NRFD, Abreu DS, Lopes LGDF, Sousa EHS, Longhinotti E. Nitroprusside─Expanding the Potential Use of an Old Drug Using Nanoparticles. Mol Pharm 2023; 20:6-22. [PMID: 36350781 DOI: 10.1021/acs.molpharmaceut.2c00661] [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: 11/11/2022]
Abstract
For more than 70 years, sodium nitroprusside (SNP) has been used to treat severe hypertension in hospital emergency settings. During this time, a few other clinical uses have also emerged such as in the treatment of acute heart failure as well as improving mitral incompetence and in the intra- and perioperative management during heart surgery. This drug functions by releasing nitric oxide (NO), which modulates several biological processes with many potential therapeutic applications. However, this small molecule has a short lifetime, and it has been administered through the use of NO donor molecules such as SNP. On the other hand, SNP also has some setbacks such as the release of cyanide ions, high water solubility, and very fast NO release kinetics. Currently, there are many drug delivery strategies that can be applied to overcome many of these limitations, providing novel opportunities for the use of old drugs, including SNP. This Perspective describes some nitroprusside properties and highlights new potential therapeutic uses arising from the use of drug delivery systems, mainly silica-based nanoparticles. There is a series of great opportunities to further explore SNP in many medical issues as reviewed, which deserves a closer look by the scientific community.
Collapse
Affiliation(s)
- Pedro Martins da Silva Filho
- Laboratório de Métodos de Análises e Modificação de Materiais (LABMA), Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, 60440-900, Fortaleza, Ceará, Brazil.,Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza, Ceará, Brazil
| | - Iury Araújo Paz
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, 60714-110, Fortaleza, Ceará, Brazil
| | | | - Dieric S Abreu
- Laboratory of Materials & Devices (Lab MaDe), Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, 60440-900, Fortaleza, Ceará, Brazil
| | - Luiz Gonzaga de França Lopes
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza, Ceará, Brazil
| | - Eduardo Henrique Silva Sousa
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza, Ceará, Brazil
| | - Elisane Longhinotti
- Laboratório de Métodos de Análises e Modificação de Materiais (LABMA), Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, 60440-900, Fortaleza, Ceará, Brazil.,Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza, Ceará, Brazil
| |
Collapse
|
4
|
Harb I, Ashour H, Rashed LA, Mostafa A, Samir M, Aboulhoda BE, El-Hanbuli H, Rashwan E, Mahmoud H. Nicorandil mitigates amiodarone-induced pulmonary toxicity and fibrosis in association with the inhibition of lung TGF-β1/PI3K/Akt1-p/mTOR axis in rats. Clin Exp Pharmacol Physiol 2023; 50:96-106. [PMID: 36208078 DOI: 10.1111/1440-1681.13728] [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: 04/02/2022] [Revised: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 12/13/2022]
Abstract
The long-term side effect of the antiarrhythmic drug, amiodarone (AMIO), such as lung toxicity, remains a critical clinical issue. The previous knowledge denotes diverse antioxidant, anti-inflammatory, and antifibrotic properties of the anti-anginal drug, nicorandil (NI). Therefore, we aimed to investigate the possible protective effect of NI on pulmonary tissue remodelling following AMIO-induced lung toxicity. The included rats were assigned into four equal groups (n = 8): (1) control, (2) control group that received NI 10 mg kg-1 day-1 , (3) model group that received AMIO in a dose of 60 mg kg-1 day-1 , and (4) treated group (AMIO-NI) that were treated with AMIO plus NI as shown above. Drug administration continued for 10 weeks. AMIO resulted in deteriorated (p < 0.001) pulmonary functions accompanied by respiratory acidosis. AMIO showed an obvious histological injury score with intense collagen deposition, disturbed nitric oxide synthase enzymes (NOS/iNOS), and increased alpha smooth muscle actin expression. Furthermore, AMIO upregulated the transforming growth factor (TGF-β1)/phosphoinositide-3 kinase (PI3K)-Akt1-p/mammalian target of rapamycin (mTOR) axis, which determined the possible mechanism of AMIO on pulmonary remodelling. NI treatment significantly (p < 0.001) prevented the AMIO-induced lung toxicity, as well as inhibited the TGF-β1/PI3K/Akt1-p/mTOR axis in the lung tissue of rats. The results were confirmed by an in-vitro study. CONCLUSION: The current results revealed that NI was effective in preserving the lung structure and functions. Amelioration of the oxidative stress and modulation of TGF-β1/PI3K/Akt1-p/mTOR have been achieved. This study suggests NI administration as a preventive therapy from the serious pulmonary fibrosis side effect of AMIO.
Collapse
Affiliation(s)
- Inas Harb
- Department of Pharmacology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Hend Ashour
- Department of Physiology, Faculty of Medicine, KingKhalid University, Abha, Saudi Arabia.,Department of Physiology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Laila A Rashed
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Abeer Mostafa
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Mai Samir
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Hala El-Hanbuli
- Department of Pathology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Eman Rashwan
- Department of Physiology, Faculty of Medicine, Jouf University, Sakakah, Saudi Arabia.,Department of Physiology, Faculty of Medicine, Al-Azhar University, Assuit, Egypt
| | - Heba Mahmoud
- Department of Pharmacology, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
5
|
Dal Negro RW, Turco P, Povero M. Nebivolol: an effective option against long-lasting dyspnoea following COVID-19 pneumonia - a pivotal double-blind, cross-over controlled study. Multidiscip Respir Med 2022; 17:886. [PMID: 36636645 PMCID: PMC9830396 DOI: 10.4081/mrm.2022.886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/14/2022] [Indexed: 12/25/2022] Open
Abstract
Background Pulmonary microvascular occlusions can aggravate SARS-CoV-2 pneumonia and result in a variable decrease in capillary blood volume (Vc). Dyspnoea may persist for several weeks after hospital discharge in many patients who have "radiologically recovered" from COVID-19 pneumonia. Dyspnoea is frequently "unexplained" in these cases because abnormalities in lung vasculature are understudied. Furthermore, even when they are identified, therapeutic options are still lacking in clinical practice, with nitric oxide (NO) supplementation being used only for severe respiratory failure in the hospital setting. Nebivolol is the only selective β1 adrenoceptor antagonist capable of inducing nitric oxide-mediated vasodilation by stimulating endothelial NO synthase via β3 agonism. The purpose of this study was to compare the effect of nebivolol versus placebo in patients who had low Vc and complained of dyspnoea for several weeks after COVID-19 pneumonia. Methods Patients of both genders, aged ≥18 years, non-smokers, who had a CT scan that revealed no COVID-related parenchymal lesions but still complaining of dyspnoea 12-16 weeks after hospital discharge, were recruited. Spirometrical volumes, blood haemoglobin, SpO2, simultaneous diffusing capacity for carbon monoxide (CO) and NO (DLCO and DLNO, respectively), DLNO/DLCO ratio, Vc and exhaled NO (eNO) were measured together with their dyspnoea score (DS), heart frequency (HF), and blood arterial pressure (BAP). Data were collected before and one week after both placebo (P) and nebivolol (N) (2.5 mg od) double-blind cross-over administered at a two-week interval. Data were statistically compared, and p<0.05 assumed as statistically significant. Results Eight patients (3 males) were investigated. In baseline, their mean DS was 2.5±0.6 SD, despite the normality of lung volumes. DLCO and DLNO mean values were lower than predicted, while mean DLNO/DLCO ratio was higher. Mean Vc proved substantially reduced. Placebo did not modify any variable (all p=ns) while N improved DLco and Vc significantly (+8.5%, p<0.04 and +17.7%, p<0.003, respectively). eNO also was significantly increased (+17.6%, p<0.002). Only N lowered the dyspnoea score (-76%, p<0.001). Systolic and diastolic BAP were slightly lowered (-7.5%, p<0.02 and -5.1%, p<0.04, respectively), together with HF (-16.8%, p<0.03). Conclusions The simultaneous assessment of DLNO, DLCO, DLNO/DLCO ratio, and Vc confirmed that long-lasting dyspnoea is related to hidden abnormalities in the lung capillary vasculature. These abnormalities can persist even after the complete resolution of parenchymal lesions regardless of the normality of lung volumes. Nebivolol, but not placebo, improves DS and Vc significantly. The mechanism suggested is the NO-mediated vasodilation via the β3 adrenoceptor stimulation of endothelial NO synthase. This hypothesis is supported by the substantial increase of eNO only assessed after nebivolol. As the nebivolol tolerability in these post-COVID normotensive patients was very good, the therapeutic use of nebivolol against residual and symptomatic signs of long-COVID can be suggested in out-patients.
Collapse
Affiliation(s)
- Roberto W. Dal Negro
- National Centre for Respiratory Pharmacoeconomics and Pharmacoepidemiology - CESFAR, Verona ,National Centre for Respiratory Pharmacoeconomics and Pharmacoepidemiology, Via G. Rossetti 4, 37124 Verona, Italy.
| | - Paola Turco
- Research & Clinical Governance, Verona, Italy
| | | |
Collapse
|
6
|
Ji X, Zhong Z. External stimuli-responsive gasotransmitter prodrugs: Chemistry and spatiotemporal release. J Control Release 2022; 351:81-101. [PMID: 36116579 DOI: 10.1016/j.jconrel.2022.09.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022]
Abstract
Gasotransmitters like nitric oxide, carbon monoxide, and hydrogen sulfide with unique pleiotropic pharmacological effects in mammals are an emerging therapeutic modality for different human diseases including cancer, infection, ischemia-reperfusion injuries, and inflammation; however, their clinical translation is hampered by the lack of a reliable delivery form, which delivers such gasotransmitters to the action site with precisely controlled dosage. The external stimuli-responsive prodrug strategy has shown tremendous potential in developing gasotransmitter prodrugs, which affords precise temporospatial control and better dose control compared with endogenous stimuli-sensitive prodrugs. The promising external stimuli employed for gasotransmitter activation range from photo, ultrasound, and bioorthogonal click chemistry to exogenous enzymes. Herein, we highlight the recent development of external stimuli-mediated decaging chemistry for the temporospatial delivery of gasotransmitters including nitric oxide, carbon monoxide, hydrogen sulfide and sulfur dioxide, and discuss the pros and cons of different designs.
Collapse
Affiliation(s)
- Xingyue Ji
- College of Pharmaceutical Sciences, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
7
|
Ashour H, Elsayed MH, Elmorsy S, Harb IA. Hypothesis: The potential therapeutic role of nicorandil in COVID-19. Clin Exp Pharmacol Physiol 2020; 47:1791-1797. [PMID: 32881062 PMCID: PMC7436472 DOI: 10.1111/1440-1681.13395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/14/2020] [Accepted: 08/02/2020] [Indexed: 12/16/2022]
Abstract
At present, there is yet no specific antiviral treatment or immunization against the newly identified human severe acute respiratory syndrome virus (SARS-CoV2) that results in a rapidly progressive pandemic coronavirus disease 2019 (COVID-19). We believe in a crucial need for a clinical strategy to counteract this viral pandemic based on the known pathogenesis throughout the disease course. Evidence suggests that exaggerated patient's inflammatory response and oxidative stress are likely to aggravate the disease pathology. The resulting endothelial dysfunction further induces fibrosis and coagulopathy. These disturbances can generate severe acute respiratory distress syndrome (ARDS) that can progress into respiratory and circulatory failure. Nicorandil is an anti-anginal vasodilator drug acts by increasing nitric oxide bioavailability and opening of the KATP channel. Recently, nicorandil has been recognized to possess multiple protective effects against tissue injury. Here, we address a possible modulatory role of nicorandil against COVID-19 pathogenesis. We hypothesise nicorandil would be an effective form of adjuvant therapy against COVID-19.
Collapse
Affiliation(s)
- Hend Ashour
- Faculty of MedicineDepartment of PhysiologyKing Khalid UniversityAbhaSaudi Arabia
- Faculty of MedicineDepartment of PhysiologyKasr AlainyCairo UniversityCairoEgypt
| | - Mohamed H. Elsayed
- Department of Pediatrics ICUAl‐Ahrar Teaching HospitalZagazigEgypt
- Department of Pediatrics ICUKing Fahd Armed Forces HospitalKhamis MushaitSaudi Arabia
| | - Soha Elmorsy
- Faculty of MedicineDepartment of Medical PharmacologyCairo UniversityCairoEgypt
| | - Inas A. Harb
- Faculty of MedicineDepartment of Medical PharmacologyCairo UniversityCairoEgypt
| |
Collapse
|
8
|
Wang H, Wang L, Xie Z, Zhou S, Li Y, Zhou Y, Sun M. Nitric Oxide (NO) and NO Synthases (NOS)-Based Targeted Therapy for Colon Cancer. Cancers (Basel) 2020; 12:E1881. [PMID: 32668616 PMCID: PMC7408898 DOI: 10.3390/cancers12071881] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal malignancies worldwide and CRC therapy remains unsatisfactory. In recent decades, nitric oxide (NO)-a free-radical gas-plus its endogenous producer NO synthases (NOS), have attracted considerable attention. NO exerts dual effects (pro- and anti-tumor) in cancers. Endogenous levels of NO promote colon neoplasms, whereas exogenously sustained doses lead to cytotoxic functions. Importantly, NO has been implicated as an essential mediator in many signaling pathways in CRC, such as the Wnt/β-catenin and extracellular-signal-regulated kinase (ERK) pathways, which are closely associated with cancer initiation, metastasis, inflammation, and chemo-/radio-resistance. Therefore, NO/NOS have been proposed as promising targets in the regulation of CRC carcinogenesis. Clinically relevant NO-donating agents have been developed for CRC therapy to deliver a high level of NO to tumor sites. Notably, inducible NOS (iNOS) is ubiquitously over-expressed in inflammatory-associated colon cancer. The development of iNOS inhibitors contributes to targeted therapies for CRC with clinical benefits. In this review, we summarize the multifaceted mechanisms of NO-mediated networks in several hallmarks of CRC. We review the clinical manifestation and limitations of NO donors and NOS inhibitors in clinical trials. We also discuss the possible directions of NO/NOS therapies in the immediate future.
Collapse
Affiliation(s)
- Hao Wang
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, China;
| | - Liye Wang
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX, 77204, USA; (L.W.); (Z.X.); (S.Z.); (Y.L.)
| | - Zuoxu Xie
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX, 77204, USA; (L.W.); (Z.X.); (S.Z.); (Y.L.)
| | - Shuang Zhou
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX, 77204, USA; (L.W.); (Z.X.); (S.Z.); (Y.L.)
| | - Yan Li
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX, 77204, USA; (L.W.); (Z.X.); (S.Z.); (Y.L.)
| | - Yue Zhou
- Department of Statistics, North Dakota University, Fargo, ND 58105, USA;
| | - Meiyan Sun
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, China;
| |
Collapse
|
9
|
Ückert S, Kedia GT, Tsikas D, Simon A, Bannowsky A, Kuczyk MA. Emerging drugs to target lower urinary tract symptomatology (LUTS)/benign prostatic hyperplasia (BPH): focus on the prostate. World J Urol 2019; 38:1423-1435. [PMID: 31506747 DOI: 10.1007/s00345-019-02933-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 08/28/2019] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES The benign prostatic syndrome, comprising lower urinary tract symptomatology secondary to benign prostatic hyperplasia/enlargement, represents a major health care issue in westernized countries. The pharmacological management involves alpha-adrenoceptor antagonists, intervention into the hormonal control of prostate growth using inhibitors of the enzyme 5-alpha-reductase, and stimulation of the nitric oxide/cyclic GMP pathway by tadalafil, an inhibitor of the phosphodiesterase type 5. METHODS This review summarizes the achievements which have been made in the development of drug candidates assumed to offer opportunities as beneficial treatment options in the management of the benign prostatic syndrome. RESULTS A review of the literature has revealed that the line of development is focusing on drugs interfering with peripheral neuromuscular/neuronal mechanisms (nitric oxide donor drugs, agonists/antagonists of endogenous peptides, botulinum toxin, NX-1207), the steroidal axis (cetrorelix) or the metabolic turn-over (lonidamine), as well as the combination of drugs already established in the treatment of lower urinary tract symptomatology/benign prostatic hyperplasia (phosphodiesterase 5 inhibitor plus alpha-adrenoceptor antagonist). CONCLUSION Many research efforts have provided the basis for the development of new therapeutic modalities for the management of lower urinary tract dysfunctions, some of which might be offered to the patients in the near future.
Collapse
Affiliation(s)
- Stefan Ückert
- Division of Surgery, Department of Urology and Urological Oncology, Hannover Medical School, 30623, Hannover, Germany.
| | - George T Kedia
- Division of Surgery, Department of Urology and Urological Oncology, Hannover Medical School, 30623, Hannover, Germany
| | - Dimitrios Tsikas
- Core Unit Proteomics, Center of Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Annika Simon
- Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | | | - Markus A Kuczyk
- Division of Surgery, Department of Urology and Urological Oncology, Hannover Medical School, 30623, Hannover, Germany
| |
Collapse
|
10
|
Sun F, Wang Y, Luo X, Ma Z, Xu Y, Zhang X, Lv T, Zhang Y, Wang M, Huang Z, Zhang J. Anti-CD24 Antibody-Nitric Oxide Conjugate Selectively and Potently Suppresses Hepatic Carcinoma. Cancer Res 2019; 79:3395-3405. [PMID: 30918001 DOI: 10.1158/0008-5472.can-18-2839] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/04/2019] [Accepted: 03/22/2019] [Indexed: 11/16/2022]
Abstract
Nitric oxide (NO) has a wide range of potential applications in tumor therapy. However, a targeted delivery system for NO donors has remained elusive, creating a bottleneck that limits its druggability. The antibody-drug conjugate (ADC) is a targeted drug delivery system composed of an antibody linked to an active cytotoxic drug. This design may compensate for the weak targeting ability and various biological functions of the NO donor. In this study, we designed the NO donor HL-2, which had a targeted, cleaved disulfide bond and an attachable maleimide terminal. We conjugated HL-2 with an antibody that targeted CD24 through a thioether bond to generate an ADC-like immunoconjugate, antibody-nitric oxide conjugate (ANC), which we named HN-01. HN-01 showed efficient internalization and significantly increased the release of NO in hepatic carcinoma cells in vitro. HN-01 induced apoptosis of tumor cells and suppressed tumor growth in hepatic carcinoma-bearing nude mice through antibody-dependent co-toxicity; HN-01 also increased NO levels in tumor cells. Collectively, this study expands the concept of ADC and provides an innovative NO donor and ANC to address current challenges in targeted delivery of NO. This new inspiration for an ANC design can also be used in future studies for other molecules with intracellular targets. SIGNIFICANCE: This study is the first to expand the concept of ADC with an antibody-nitric oxide conjugate that suppresses hepatic carcinoma in vitro and in vivo.
Collapse
Affiliation(s)
- Fumou Sun
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Yang Wang
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaojun Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, China
| | - Zhaoxiong Ma
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Yao Xu
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Xinrong Zhang
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Tian Lv
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, China
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, China
| | - Min Wang
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, China.
| | - Juan Zhang
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
11
|
Wang H, Liu Y, Wang Z, Yang M, Gu Y. 808 nm-light-excited upconversion nanoprobe based on LRET for the ratiometric detection of nitric oxide in living cancer cells. NANOSCALE 2018; 10:10641-10649. [PMID: 29845132 DOI: 10.1039/c8nr03078b] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
NO (nitric oxide) has dual functions in cancer, promoting carcinogenesis in low concentrations and inducing tumor cell apoptosis at high concentrations. The dual-edged-sword functions of NO make it particularly appealing to develop a sensitive and specific chemical probe for its detection. However, most NO sensors suffer from poor Stokes shifts and are limited by ultraviolet (UV) or visible light excitation, which render it difficult to avoid the intrinsic background signal. In this study, an 808 nm laser-excited Nd3+-sensitized upconversion nanoprobe based on LRET (luminescence resonance energy-transfer) for NO detection was constructed for the first time. This probe was composed of Nd3+-sensitized core-shell upconversion nanoparticles (540 nm and 660 nm emission) as the energy donor and RhBs as the acceptor. In the presence of NO, RhBs was converted into Rhodamine B and its strong absorption subsequently quenched the 540 nm fluorescence of UCNPs, while the emission at 660 nm remained constant. The ratiometric detection of the fluorescence at 540 nm, as compared to 660 nm, can precisely respond to the difference in NO levels with a detection limit of 0.21 μM. Importantly, compared with conventional UCNPs excited at 980 nm, the 808 nm light excitation led to lower water absorption and deeper tissue penetration, thus avoiding overheating and allowing for long-term biological imaging. This strategy has been perfectly applied to detecting the NO levels in living cells to differentiate the tumor cells from normal cells based on varied intracellular NO concentration. Further, the nanosystem realized the real-time monitoring of NO during treatment with NO donor drugs, which could inspire the future application of this probe to guide NO therapy.
Collapse
Affiliation(s)
- Han Wang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Yi Liu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhaohui Wang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Man Yang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
12
|
Eilertsen M, Allin SM, Pearson RJ. New 4-aryl-1,3,2-oxathiazolylium-5-olates: Chemical synthesis and photochemical stability of a novel series of S-nitrosothiols. Bioorg Med Chem Lett 2018; 28:1106-1110. [PMID: 29482942 DOI: 10.1016/j.bmcl.2018.01.059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/25/2018] [Accepted: 01/27/2018] [Indexed: 01/12/2023]
Abstract
S-nitrosothiols (RSNOs) remain one of the most popular classes of NO-donating compounds due to their ability to release nitric oxide (NO) under non-enzymatic means whilst producing an inert disulphide by-product. However, alligning these compounds to the different biological fields of NO research has proved to be problematic due to the inherent instability of such compounds under a variety of conditions including heat, light and the presence of copper ions. 1,3,2-Oxathiazolylium-5-olates (OZOs) represent an interesting subclass of S-nitrosothiols that lock the -SNO moiety into a five membered heterocyclic ring in an attempt to improve the compound's overall stability. The synthesis of a novel series of halogen-containing OZOs was comprehensively studied resulting in a seven-step route and overall yields ranging between 21 and 37%. The photochemical stability of these compounds was assessed to determine if S-nitrosothiols locked within these mesoionic ring systems can offer greater stability and thereby release NO in a more controllable fashion than their non-cyclic counterparts.
Collapse
Affiliation(s)
- Monica Eilertsen
- School of Pharmacy, Keele University, Hornbeam Building, Keele, Staffordshire ST5 5BG, UK
| | - Steve M Allin
- Department of Chemistry and Forensics, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Russell J Pearson
- School of Pharmacy, Keele University, Hornbeam Building, Keele, Staffordshire ST5 5BG, UK.
| |
Collapse
|
13
|
Shahbazi S, Kaur J, Singh S, Achary KG, Wani S, Jema S, Akhtar J, Sobti RC. Impact of novel N-aryl piperamide NO donors on NF-κB translocation in neuroinflammation: rational drug-designing synthesis and biological evaluation. Innate Immun 2017; 24:24-39. [PMID: 29145791 PMCID: PMC6830765 DOI: 10.1177/1753425917740727] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
NO donor drugs showed a significant therapeutic effect in the treatment of many
diseases, such as arteriopathies, various acute and chronic inflammatory
conditions, and several degenerative diseases. NO-releasing anti-inflammatory
drugs are the prototypes of a novel class of compounds, combining the
pharmacological activities of anti-inflammatory and anti-nociceptive of drugs
with those of NO, thus possessing potential therapeutic applications in a great
variety of diseases. In this study, we designed and predicted biological
activity by targeting cyclooxygenase type 2 (COX-2) and NF-κB subunits and
pharmacological profiling along with toxicity predictions of various
N-aryl piperamides linked via an ester bond to a spacer
that is bound to a NO-releasing moiety (-ONO2). The result of absorption,
distribution, metabolism and excretion and Docking studies indicated that among
51 designed molecules PA-3′K showed the best binding potential in both the
substrate and inhibitory binding pocket of the COX-2 enzyme with affinity values
of –9.33 and –5.12 for PDB ID 1CVU and 3LN1, respectively, thereby having the
potential to be developed as a therapeutic agent. The results of cell
viabilities indicated that PA-3′k possesses the best cell viability property
with respect to its dose (17.33 ng/ml), with 67.76% and 67.93% viable cells for
CHME3 and SVG cell lines, respectively.
Collapse
Affiliation(s)
- Sajad Shahbazi
- Department of Biotechnology, Panjab
University, Chandigarh, India
- Sajad Shahbazi, Department of Biotechnology,
Panjab University, Chandigarh, 160014, India.
| | - Jagdeep Kaur
- Department of Biotechnology, Panjab
University, Chandigarh, India
| | - Shikha Singh
- Center of Biotechnology, Siksha O
Anusandhan University, Khandagiri, Bhubaneswar, Odisha, India
| | | | - Sameena Wani
- Department of Experimental Medicine and
Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh,
India
| | | | - Jabed Akhtar
- Imgenex India, E5, Infocity,
Bhubaneswar, Odisha, India
| | - Ranbir Chander Sobti
- Department of Biotechnology, Panjab
University, Chandigarh, India
- Babasaheb Bhimrao Ambedkar University,
Lucknow, India
| |
Collapse
|
14
|
Wajih N, Basu S, Jailwala A, Kim HW, Ostrowski D, Perlegas A, Bolden CA, Buechler NL, Gladwin MT, Caudell DL, Rahbar E, Alexander-Miller MA, Vachharajani V, Kim-Shapiro DB. Potential therapeutic action of nitrite in sickle cell disease. Redox Biol 2017; 12:1026-1039. [PMID: 28511346 PMCID: PMC5430577 DOI: 10.1016/j.redox.2017.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/07/2017] [Accepted: 05/08/2017] [Indexed: 12/26/2022] Open
Abstract
Sickle cell disease is caused by a mutant form of hemoglobin that polymerizes under hypoxic conditions, increasing rigidity, fragility, calcium influx-mediated dehydration, and adhesivity of red blood cells. Increased red cell fragility results in hemolysis, which reduces nitric oxide (NO) bioavailability, and induces platelet activation and inflammation leading to adhesion of circulating blood cells. Nitric Oxide inhibits adhesion and platelet activation. Nitrite has emerged as an attractive therapeutic agent that targets delivery of NO activity to areas of hypoxia through bioactivation by deoxygenated red blood cell hemoglobin. In this study, we demonstrate anti-platelet activity of nitrite at doses achievable through dietary interventions with comparison to similar doses with other NO donating agents. Unlike other NO donating agents, nitrite activity is shown to be potentiated in the presence of red blood cells in hypoxic conditions. We also show that nitrite reduces calcium associated loss of phospholipid asymmetry that is associated with increased red cell adhesion, and that red cell deformability is also improved. We show that nitrite inhibits red cell adhesion in a microfluidic flow-channel assay after endothelial cell activation. In further investigations, we show that leukocyte and platelet adhesion is blunted in nitrite-fed wild type mice compared to control after either lipopolysaccharide- or hemolysis-induced inflammation. Moreover, we demonstrate that nitrite treatment results in a reduction in adhesion of circulating blood cells and reduced red blood cell hemolysis in humanized transgenic sickle cell mice subjected to local hypoxia. These data suggest that nitrite is an effective anti-platelet and anti-adhesion agent that is activated by red blood cells, with enhanced potency under physiological hypoxia and in venous blood that may be useful therapeutically.
Collapse
Affiliation(s)
- Nadeem Wajih
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, United States; Translational Science Center, Wake Forest University, Winston-Salem, NC 27109, United States
| | - Swati Basu
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, United States; Translational Science Center, Wake Forest University, Winston-Salem, NC 27109, United States
| | - Anuj Jailwala
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, United States
| | - Hee Won Kim
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, United States
| | - David Ostrowski
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, United States
| | - Andreas Perlegas
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, United States
| | - Crystal A Bolden
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, United States
| | - Nancy L Buechler
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| | - Mark T Gladwin
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - David L Caudell
- Department of Pathology-Comparative Medicine, Section on Rheumatology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| | - Elaheh Rahbar
- Department of Biomedical Engineering, Section on Rheumatology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| | - Martha A Alexander-Miller
- Department of Microbiology and Immunology, Section on Rheumatology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| | - Vidula Vachharajani
- Translational Science Center, Wake Forest University, Winston-Salem, NC 27109, United States; Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States.
| | - Daniel B Kim-Shapiro
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, United States; Translational Science Center, Wake Forest University, Winston-Salem, NC 27109, United States.
| |
Collapse
|
15
|
Serafim RAM, Pernichelle FG, Ferreira EI. The latest advances in the discovery of nitric oxide hybrid drug compounds. Expert Opin Drug Discov 2017; 12:941-953. [PMID: 28664751 DOI: 10.1080/17460441.2017.1344400] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION There is a great interest in Nitric oxide (NO) within medicinal chemistry since it's involved in human signaling pathways. Prodrugs or hybrid compounds containing NO-donor scaffolds linked to an active compound are valuable, due to their potential for modulating many pathological conditions due to NO's biological properties when released in addition to the native drug. Compounds that selectively inhibit nitric oxide synthase isoforms (NOS) can also increase therapeutic capacity, particularly in the treatment of chronic diseases. However, search for bioactive compounds to efficiently and selectively modulate NO is still a challenge in drug discovery. Areas covered: In this review, the authors highlight the recent advances in the strategies used to discover NO-hybrid derivatives, especially those related to anti-inflammatory, cardiovascular, anticancer and anti-microorganism activities. They also focus on: nitric oxide synthase inhibitors, NO delivery materials and other related activities. Expert opinion: The process of molecular hybridization can be used to obtain NO-releasing compounds that also interact with different targets. The main problem with this approach is to control NO multiple actions in the right biological system. However, the use of NO-releasing groups with many different scaffolds leads to new molecular structures for bioactive compounds, suggesting synergies.
Collapse
Affiliation(s)
- Ricardo A M Serafim
- a LAPEN: Laboratory of Design and Synthesis of Chemotherapeutic Potentially Active against Neglected Diseases, Department of Pharmacy, Faculty of Pharmaceutical Sciences , University of São Paulo - FCF/USP , São Paulo , Brazil
| | - Filipe G Pernichelle
- a LAPEN: Laboratory of Design and Synthesis of Chemotherapeutic Potentially Active against Neglected Diseases, Department of Pharmacy, Faculty of Pharmaceutical Sciences , University of São Paulo - FCF/USP , São Paulo , Brazil
| | - Elizabeth I Ferreira
- a LAPEN: Laboratory of Design and Synthesis of Chemotherapeutic Potentially Active against Neglected Diseases, Department of Pharmacy, Faculty of Pharmaceutical Sciences , University of São Paulo - FCF/USP , São Paulo , Brazil
| |
Collapse
|
16
|
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
|
17
|
Wo Y, Brisbois EJ, Bartlett RH, Meyerhoff ME. Recent advances in thromboresistant and antimicrobial polymers for biomedical applications: just say yes to nitric oxide (NO). Biomater Sci 2016; 4:1161-83. [PMID: 27226170 PMCID: PMC4955746 DOI: 10.1039/c6bm00271d] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biomedical devices are essential for patient diagnosis and treatment; however, when blood comes in contact with foreign surfaces or homeostasis is disrupted, complications including thrombus formation and bacterial infections can interrupt device functionality, causing false readings and/or shorten device lifetime. Here, we review some of the current approaches for developing antithrombotic and antibacterial materials for biomedical applications. Special emphasis is given to materials that release or generate low levels of nitric oxide (NO). Nitric oxide is an endogenous gas molecule that can inhibit platelet activation as well as bacterial proliferation and adhesion. Various NO delivery vehicles have been developed to improve NO's therapeutic potential. In this review, we provide a summary of the NO releasing and NO generating polymeric materials developed to date, with a focus on the chemistry of different NO donors, the polymer preparation processes, and in vitro and in vivo applications of the two most promising types of NO donors studied thus far, N-diazeniumdiolates (NONOates) and S-nitrosothiols (RSNOs).
Collapse
Affiliation(s)
- Yaqi Wo
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | |
Collapse
|
18
|
Duan W, Li J, Inks ES, Chou CJ, Jia Y, Chu X, Li X, Xu W, Zhang Y. Design, synthesis, and antitumor evaluation of novel histone deacetylase inhibitors equipped with a phenylsulfonylfuroxan module as a nitric oxide donor. J Med Chem 2015; 58:4325-38. [PMID: 25906087 DOI: 10.1021/acs.jmedchem.5b00317] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
On the basis of the strategy of creating multifunctional drugs, a novel series of phenylsulfonylfuroxan-based hydroxamates with histone deacetylase (HDAC) inhibitory and nitric oxide (NO) donating activities were designed, synthesized, and evaluated. The most potent NO donor-HDAC inhibitor (HDACI) hybrid, 5c, exhibited a much greater in vitro antiproliferative activity against the human erythroleukemia (HEL) cell line than that of the approved drug SAHA (Vorinostat), and its antiproliferative activity was diminished by the NO scavenger hemoglobin in a dose-dependent manner. Further mechanism studies revealed that 5c strongly induced cellular apoptosis and G1 phase arrest in HEL cells. Animal experiment identified 5c as an orally active agent with potent antitumor activity in a HEL cell xenograft model. Interestingly, although compound 5c was remarkably HDAC6-selective at the molecular level, it exhibited pan-HDAC inhibition in a western blot assay, which is likely due to class I HDACs inhibition caused by NO release at the cellular level.
Collapse
Affiliation(s)
- Wenwen Duan
- †Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| | - Jin Li
- †Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| | - Elizabeth S Inks
- ‡Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - C James Chou
- ‡Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Yuping Jia
- §Shandong Academy of Pharmaceutical Sciences, Ji'nan, Shandong 250101, People's Republic of China
| | - Xiaojing Chu
- ∥Weifang Bochuang International Biological Medicinal Institute, Weifang, Shandong 261061, People's Republic of China
| | - Xiaoyang Li
- †Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| | - Wenfang Xu
- †Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| | - Yingjie Zhang
- †Department of Medicinal Chemistry, School of Pharmacy, Shandong University, Ji'nan, Shandong 250012, People's Republic of China
| |
Collapse
|
19
|
New organic nitrate-containing benzyloxy isonipecotanilide derivatives with vasodilatory and anti-platelet activity. Eur J Pharm Sci 2015; 72:69-80. [DOI: 10.1016/j.ejps.2015.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 02/19/2015] [Accepted: 03/03/2015] [Indexed: 01/01/2023]
|
20
|
Flister M, Timerghazin QK. Structure, Stability, and Substituent Effects in Aromatic S-Nitrosothiols: The Crucial Effect of a Cascading Negative Hyperconjugation/Conjugation Interaction. J Phys Chem A 2014; 118:9914-24. [DOI: 10.1021/jp5079136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Matthew Flister
- Department of Chemistry, Marquette University, P.O. Box
1881, Milwaukee, Wisconsin 53201-1881, United States
| | - Qadir K. Timerghazin
- Department of Chemistry, Marquette University, P.O. Box
1881, Milwaukee, Wisconsin 53201-1881, United States
| |
Collapse
|
21
|
Santana PDPB, Silva TVG, da Costa NN, da Silva BB, Carter TF, Cordeiro MDS, da Silva BJM, Santos SDSD, Herculano AM, Adona PR, Ohashi OM, Miranda MDS. Supplementation of bovine embryo culture medium with L-arginine improves embryo quality via nitric oxide production. Mol Reprod Dev 2014; 81:918-27. [PMID: 25236163 DOI: 10.1002/mrd.22387] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 08/01/2014] [Indexed: 12/30/2022]
Abstract
Nitric oxide (NO) is a cell-signaling molecule that regulates a variety of molecular pathways. We investigated the role of NO during preimplantation embryonic development by blocking its production with an inhibitor or supplementing in vitro bovine embryo cultures with its natural precursor, L-arginine, over different periods. Endpoints evaluated included blastocyst rates, development kinetics, and embryo quality. Supplementation with the NO synthase inhibitor N-Nitro-L-arginine-methyl ester (L-NAME) from Days 1 to 8 of culture decreased blastocyst (P < 0.05) and hatching (P < 0.05) rates. When added from Days 1 to 8, 50 mM L-arginine decreased blastocyst rates (P < 0.001); in contrast, when added from Days 5 to 8, 1 mM L-arginine improved embryo hatching rates (P < 0.05) and quality (P < 0.05) as well as increased POU5F1 gene expression (P < 0.05) as compared to the untreated control. Moreover, NO levels in the medium during this culture period positively correlated with the increased embryo hatching rates and quality (P < 0.05). These data suggest exerts its positive effects during the transition from morula to blastocyst stage, and that supplementing the embryo culture medium with L-arginine favors preimplantation development of bovine embryos.
Collapse
|
22
|
Zhang QY, Wang ZY, Wen F, Ren L, Li J, Teoh SH, Thian ES. Gelatin-siloxane nanoparticles to deliver nitric oxide for vascular cell regulation: synthesis, cytocompatibility, and cellular responses. J Biomed Mater Res A 2014; 103:929-38. [PMID: 24853642 DOI: 10.1002/jbm.a.35239] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/10/2014] [Accepted: 05/19/2014] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) is an important mediator in cardiovascular system to regulate vascular tone and maintain tissue homeostasis. Its role in vascular cell regulation makes it promising to address the post-surgery restenosis problem. However, the application of NO is constrained by its high reactivity. Here, we developed a novel NO-releasing gelatin-siloxane nanoparticle (GS-NO NP) to deliver NO effectively for vascular cell regulation. Results showed that gelatin-siloxane nanoparticles (GS NPs) could be synthesized via sol-gel chemistry with a diameter of ∼200 nm. It could be modified into GS-NO NPs via S-nitrosothiol (RSNO) modification. The synthesized GS-NO NPs could release a total of ∼0.12 µmol/mg NO sustainably for 7 days following a first-order exponential profile. They showed not only excellent cytocompatibility, but also rapid intracellularization within 2 h. GS-NO NPs showed inhibition of human aortic smooth muscle cell (AoSMC) proliferation and promotion of human umbilical vein endothelial cell (HUVEC) proliferation in a dose-dependent manner, which is an important approach to prevent restenosis. With GS-NO NP dose at 100 µg/mL, the proliferation of AoSMCs could be slowed down whereas the growth of HUVECs was significantly promoted. We concluded that GS-NO NPs could have potential to be used as a promising nano-system to deliver NO for vascular cell regulation.
Collapse
Affiliation(s)
- Qin-Yuan Zhang
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
23
|
Sharma K, Sengupta K, Chakrapani H. Nitroreductase-activated nitric oxide (NO) prodrugs. Bioorg Med Chem Lett 2013; 23:5964-7. [DOI: 10.1016/j.bmcl.2013.08.066] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/25/2013] [Accepted: 08/14/2013] [Indexed: 01/27/2023]
|
24
|
Costa ISF, de Souza GFP, de Oliveira MG, Abrahamsohn IDA. S-nitrosoglutathione (GSNO) is cytotoxic to intracellular amastigotes and promotes healing of topically treated Leishmania major or Leishmania braziliensis skin lesions. J Antimicrob Chemother 2013; 68:2561-8. [DOI: 10.1093/jac/dkt210] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
25
|
Carpenter AW, Schoenfisch MH. Nitric oxide release: part II. Therapeutic applications. Chem Soc Rev 2012; 41:3742-52. [PMID: 22362384 DOI: 10.1039/c2cs15273h] [Citation(s) in RCA: 678] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A wide range of nitric oxide (NO)-releasing materials has emerged as potential therapeutics that exploit NO's vast biological roles. Macromolecular NO-releasing scaffolds are particularly promising due to their ability to store and deliver larger NO payloads in a more controlled and effective manner compared to low molecular weight NO donors. While a variety of scaffolds (e.g., particles, dendrimers, and polymers/films) have been cleverly designed, the ultimate clinical utility of most NO-releasing macromolecules remains unrealized. Although not wholly predictive of clinical success, in vitro and in vivo investigations have enabled a preliminary evaluation of the therapeutic potential of such materials. In this tutorial review, we review the application of macromolecular NO therapies for cardiovascular disease, cancer, bacterial infections, and wound healing.
Collapse
Affiliation(s)
- Alexis W Carpenter
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
26
|
|
27
|
Decaluwé K, Pauwels B, Verpoest S, Van de Voorde J. New Therapeutic Targets for the Treatment of Erectile Dysfunction. J Sex Med 2011; 8:3271-90. [DOI: 10.1111/j.1743-6109.2011.02459.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
28
|
Montenegro MF, Amaral JH, Pinheiro LC, Sakamoto EK, Ferreira GC, Reis RI, Marçal DMO, Pereira RP, Tanus-Santos JE. Sodium nitrite downregulates vascular NADPH oxidase and exerts antihypertensive effects in hypertension. Free Radic Biol Med 2011; 51:144-52. [PMID: 21530643 DOI: 10.1016/j.freeradbiomed.2011.04.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 03/24/2011] [Accepted: 04/05/2011] [Indexed: 02/07/2023]
Abstract
Dietary nitrite and nitrate are important sources of nitric oxide (NO). However, the use of nitrite as an antihypertensive drug may be limited by increased oxidative stress associated with hypertension. We evaluated the antihypertensive effects of sodium nitrite given in drinking water for 4 weeks in two-kidney one-clip (2K1C) hypertensive rats and the effects induced by nitrite on NO bioavailability and oxidative stress. We found that, even under the increased oxidative stress conditions present in 2K1C hypertension, nitrite reduced systolic blood pressure in a dose-dependent manner. Whereas treatment with nitrite did not significantly change plasma nitrite concentrations in 2K1C rats, it increased plasma nitrate levels significantly. Surprisingly, nitrite treatment exerted antioxidant effects in both hypertensive and sham-normotensive control rats. A series of in vitro experiments was carried out to show that the antioxidant effects induced by nitrite do not involve direct antioxidant effects or xanthine oxidase activity inhibition. Conversely, nitrite decreased vascular NADPH oxidase activity. Taken together, our results show for the first time that nitrite has antihypertensive effects in 2K1C hypertensive rats, which may be due to its antioxidant properties resulting from vascular NADPH oxidase activity inhibition.
Collapse
Affiliation(s)
- Marcelo F Montenegro
- Department of Pharmacology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, 14049-900 Ribeirao Preto, SP, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhang Y, Janssens SP, Wingler K, Schmidt HHHW, Moens AL. Modulating endothelial nitric oxide synthase: a new cardiovascular therapeutic strategy. Am J Physiol Heart Circ Physiol 2011; 301:H634-46. [PMID: 21622818 DOI: 10.1152/ajpheart.01315.2010] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The pathogenesis of many cardiovascular diseases is associated with reduced nitric oxide (NO) bioavailability and/or increased endothelial NO synthase (eNOS)-dependent superoxide formation. These findings support that restoring and conserving adequate NO signaling in the heart and blood vessels is a promising therapeutic intervention. In particular, modulating eNOS, e.g., through increasing the bioavailability of its substrate and cofactors, enhancing its transcription, and interfering with other modulators of eNOS pathway, such as netrin-1, has a high potential for effective treatments of cardiovascular diseases. This review provides an overview of the possibilities for modulating eNOS and how this may be translated to the clinic in addition to describing the genetic models used to study eNOS modulation.
Collapse
Affiliation(s)
- Yixuan Zhang
- Department of Cardiology, Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
30
|
Hong SY, Borchert GL, Maciag AE, Nandurdikar RS, Saavedra JE, Keefer LK, Phang JM, Chakrapani H. The Nitric Oxide Prodrug V-PROLI/NO Inhibits Cellular Uptake of Proline. ACS Med Chem Lett 2010; 1:386-389. [PMID: 21212855 DOI: 10.1021/ml1000905] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
V-PYRRO/NO is a well studied nitric oxide (NO) prodrug which has been shown to protect human liver cells from arsenic, acetaminophen, and other toxic assaults in vivo. Its proline-based analogue, V-PROLI/NO, was designed to be a more biocompatible form that decomposes to the naturally occurring metabolites of proline, NO, and glycolaldehyde. Like V-PYRRO/NO, this cytochrome P450-activated prodrug was previously assumed to passively diffuse through the cellular membrane. Using (14)C-labeled proline in a competition assay, we show that V-PROLI/NO is transported through proline transporters into multiple cell lines. A fluorescent NO-sensitive dye (DAF-FM diacetate) and nitrite excretion indicated elevated intracellular NO release after metabolism over V-PYRRO/NO. These results also allowed us to predict and design a more permeable analogue, V-SARCO/NO. We report a proline transporter-based strategy for the selective transport of NO prodrugs that may have enhanced efficacy and aid in development of further NO prodrugs with increased permeability.
Collapse
Affiliation(s)
- Sam Y. Hong
- Chemistry Section, Laboratory of Comparative Carcinogenesis
| | | | | | | | | | | | | | - Harinath Chakrapani
- Department of Chemistry, Indian Institute of Science Education and Research, Pune 411 008, Maharashtra, India
| |
Collapse
|
31
|
|
32
|
Zhang MM, Yang S, Gao FL, Ma HJ, Pi KJ, Cui DL, Zhang ZK. Apoptosis of hepatic stellate cells (HSC-T6) induced by sodium nitroprusside and mechanisms involved. Shijie Huaren Xiaohua Zazhi 2010; 18:761-766. [DOI: 10.11569/wcjd.v18.i8.761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether nitric oxide (NO) donor sodium nitroprusside (SNP) can induce the apoptosis of hepatic stellate cells (HSC-T6) and to explore potential mechanisms involved.
METHODS: The apoptosis of HSC-T6 cells was determined by flow cytometry and Hoechst staining. The nuclear translocation of nuclear factor-κB (NF-κB) p65 was detected by laser scanning confocal microscopy. The expression of tissue inhibitor of matrix metalloproteinase-1 (TIMP-1), type I procollagen (procollagen I), and growth arrest and DNA damage-inducible protein (GADD45β) mRNAs was detected by real-time reverse transcription-polymerase chain reaction (RT-PCR).
RESULTS: The apoptosis rate was significantly higher in HSC-T6 cells treated with SNP than in control cells (20.78% ± 5.91%vs 3.25% ± 1.26%, P = 0.031). Apoptotic HSC-T6 cells showed dense nuclear staining or granular fluorescence after Hoechst staining. Tumor necrosis factor-α (TNF-α)-mediated nuclear translocation of NF-κB p65 was inhibited by SNP treatment. With the increase in SNP dose, the expression levels of TIMP-1, procollagen I and GADD45β mRNAs were reduced (all P < 0.05).
CONCLUSION: SNP can induce the apoptosis of HSC-T6 cells and reduce the expression of TIMP-1 and procollagen I mRNAs perhaps by inhibiting NF-κB activity and reducing GADD45β mRNA expression.
Collapse
|
33
|
Reduced ischemia/reperfusion injury via glutathione-initiated nitric oxide-releasing dendrimers. Nitric Oxide 2009; 22:30-6. [PMID: 19914388 DOI: 10.1016/j.niox.2009.11.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 10/30/2009] [Accepted: 11/07/2009] [Indexed: 11/29/2022]
Abstract
We report the therapeutic potential of S-nitroso-N-acetylpenicillamine-derivatized generation-4 polyamidoamine dendrimers (G4-SNAP) for reducing ischemia/reperfusion (I/R) injury in an isolated, perfused rat heart. The use of this dendrimer scaffold to deliver the nitrosothiol therapeutic did not inhibit NO donor activity as the required dose of G4-SNAP to minimize I/R injury (31nM corresponding to 2microM SNAP) was consistent with the optimum concentration of small molecule SNAP alone. An exploration of G4-SNAP NO release kinetics in the presence of physiologically relevant concentrations of glutathione (GSH) indicated enhanced NO release (t[NO]=1.28microM NO/mg) at 500microM GSH. Reperfusion experiments conducted with 500microM GSH further lowered the optimal therapeutic G4-SNAP dose to 230pM (i.e., 15nM SNAP). The unique combination of G4-SNAP dendrimer and glutathione trigger represents a novel strategy with possible clinical relevance toward salvaging ischemic tissue.
Collapse
|
34
|
Ruiz-Torres MP, Griera M, Chamorro A, Díez-Marqués ML, Rodríguez-Puyol D, Rodríguez-Puyol M. Tirofiban increases soluble guanylate cyclase in rat vascular walls: pharmacological and pathophysiological consequences. Cardiovasc Res 2009; 82:125-32. [DOI: 10.1093/cvr/cvn359] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
35
|
Andrei D, Maciag AE, Chakrapani H, Citro ML, Keefer LK, Saavedra JE. Aryl bis(diazeniumdiolates): potent inducers of S-glutathionylation of cellular proteins and their in vitro antiproliferative activities. J Med Chem 2008; 51:7944-52. [PMID: 19053760 PMCID: PMC2629944 DOI: 10.1021/jm800831y] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A number of bis(diazeniumdiolates) that we designed to release up to 4 mol of nitric oxide (NO) and that are structural analogues of the NO prodrug and anticancer lead compound O(2)-{2,4-dinitro-5-[4-(N-methylamino)benzoyloxy]phenyl} 1-(N,N-dimethylamino)diazen-1-ium-1,2- diolate (PABA/NO) were synthesized and studied. A majority of these compounds yielded higher levels of NO, were better inhibitors of proliferation of a number of cancer cell lines, and more rapidly induced substantially increased levels of S-glutathionylation of cellular proteins in comparison with PABA/NO. In most cases, the antiproliferative activity and extents of S-glutathionylation correlated well with levels of intracellular NO release. We report bis(diazeniumdiolates) to be a class of S-glutathionylating agents with potent antiproliferative and S-glutathionylating activity.
Collapse
Affiliation(s)
- Daniela Andrei
- To whom correspondence should be addressed. Phone: 708-524-6540. Fax: 708-524-6055. E-mail: . Phone: 301-846-1467. Fax: 301-846-5946. E-mail:
| | | | | | | | - Larry K. Keefer
- To whom correspondence should be addressed. Phone: 708-524-6540. Fax: 708-524-6055. E-mail: . Phone: 301-846-1467. Fax: 301-846-5946. E-mail:
| | | |
Collapse
|
36
|
Chakrapani H, Kalathur RC, Maciag AE, Citro ML, Ji X, Keefer LK, Saavedra JE. Synthesis, mechanistic studies, and anti-proliferative activity of glutathione/glutathione S-transferase-activated nitric oxide prodrugs. Bioorg Med Chem 2008; 16:9764-71. [PMID: 18930407 PMCID: PMC2631660 DOI: 10.1016/j.bmc.2008.09.063] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 09/24/2008] [Accepted: 09/26/2008] [Indexed: 10/21/2022]
Abstract
Nitric oxide (NO) prodrugs such as O(2)-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate (JS-K) are a growing class of promising NO-based therapeutics. Nitric oxide release from the anti-cancer lead compound, JS-K, is proposed to occur through a nucleophilic aromatic substitution by glutathione (GSH) catalyzed by glutathione S-transferase (GST) to form a diazeniumdiolate anion that spontaneously releases NO. In this study, a number of structural analogues of JS-K were synthesized and their chemical and biological properties were compared with those of JS-K. The homopiperazine analogue of JS-K showed anti-cancer activity that is comparable with that of JS-K but with a diminished reactivity towards both GSH and GSH/GST; both the aforementioned compounds displayed no cytotoxic activity towards normal renal epithelial cell line at concentrations where they significantly diminished the proliferation of a panel of renal cancer cell lines. These properties may prove advantageous in the further development of this class of nitric oxide prodrugs as cancer therapeutic agents.
Collapse
Affiliation(s)
- Harinath Chakrapani
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Ravi C. Kalathur
- Biomolecular Structure Section, Macromolecular Crystallography Section, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Anna E. Maciag
- Basic Research Program, SAIC-Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Michael L. Citro
- Basic Research Program, SAIC-Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Xinhua Ji
- Biomolecular Structure Section, Macromolecular Crystallography Section, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Larry K. Keefer
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Joseph E. Saavedra
- Basic Research Program, SAIC-Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| |
Collapse
|
37
|
Chakrapani H, Maciag AE, Citro ML, Keefer LK, Saavedra JE. Cell-permeable esters of diazeniumdiolate-based nitric oxide prodrugs. Org Lett 2008; 10:5155-8. [PMID: 18956868 DOI: 10.1021/ol8020989] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although O(2)-(2,4-dinitrophenyl) derivatives of diazeniumdiolate-based nitric oxide (NO) prodrugs bearing a free carboxylic acid group were activated by glutathione to release NO, these compounds were poor sources of intracellular NO and showed diminished antiproliferative activity against human leukemia HL-60 cells. The carboxylic acid esters of these prodrugs, however, were found to be superior sources of intracellular NO and potent inhibitors of HL-60 cell proliferation.
Collapse
Affiliation(s)
- Harinath Chakrapani
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA.
| | | | | | | | | |
Collapse
|
38
|
Synthesis of S-[13N]nitrosoglutathione (13N-GSNO) as a new potential PET imaging agent. Appl Radiat Isot 2008; 67:95-9. [PMID: 19019692 DOI: 10.1016/j.apradiso.2008.09.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 08/27/2008] [Accepted: 09/19/2008] [Indexed: 01/26/2023]
Abstract
In the present paper, a fast and reproducible method for the synthesis of S-[(13)N]nitrosoglutathione is reported for the first time. The labeling strategy is based on the production of [(13)N]NO(3)(-) via the (16)O(p,alpha)(13)N nuclear reaction in water, as opposed to the standardized production of [(13)N]NH(4)(+) in 2mM aqueous ethanol. Following the reduction of [(13)N]NO(3)(-) to [(13)N]NO(2)(-), the reaction with glutathione in the presence of hydrochloric acid led to the desired radiotracer with a good radiochemical yield (24.2+/-2.0% end of synthesis, n=5) in a short production time (3min from the end of bombardment).
Collapse
|
39
|
Rigas B, Williams JL. NO-donating NSAIDs and cancer: an overview with a note on whether NO is required for their action. Nitric Oxide 2008; 19:199-204. [PMID: 18486630 DOI: 10.1016/j.niox.2008.04.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 04/18/2008] [Indexed: 12/11/2022]
Abstract
Nitric oxide-donating nonsteroidal anti-inflammatory drugs (NO-NSAIDs) consist of a conventional NSAID to which an NO-releasing moiety is attached covalently, often via a spacer molecule. NO-NSAIDs represent an emerging class of compounds with chemopreventive properties against a variety of cancers, demonstrated in preclinical models including cell culture systems and animal tumor models; their potential efficacy in humans has not been assessed. Their mechanism of action appears complex and involves the generation of reactive oxygen species, suppression of microsatellite instability in mismatch repair-deficient cells, and modulation of several signaling cascades that culminate in inhibited cell renewal and enhanced apoptosis. NO, long appreciated to be able to protect from and also promote cancer, is released form NO-NSAIDs and constitutes their defining property. Existing data are consistent with the notion that NO may mediate their anticancer effect. In addition there is evidence that long-term administration of NO-donating compounds is not associated with increased incidence of colon cancer. Whether NO release is required for the anticancer effect of NO-NSAIDs has being questioned by recent data indicating that, at least in the case of NO-aspirin, the NO-releasing moiety may serve as a leaving group while the spacer actually being the moiety responsible for its pharmacological action. Regardless of mechanistic issues, these compounds promise to contribute to the control of cancer.
Collapse
Affiliation(s)
- Basil Rigas
- Division of Cancer Prevention, Stony Brook University, Life Sciences Building, Stony Brook, NY 11794-5200, USA.
| | | |
Collapse
|
40
|
Chakrapani H, Wilde TC, Citro ML, Goodblatt MM, Keefer LK, Saavedra JE. Synthesis, nitric oxide release, and anti-leukemic activity of glutathione-activated nitric oxide prodrugs: Structural analogues of PABA/NO, an anti-cancer lead compound. Bioorg Med Chem 2008; 16:2657-64. [PMID: 18060792 PMCID: PMC2631658 DOI: 10.1016/j.bmc.2007.11.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Revised: 11/09/2007] [Accepted: 11/13/2007] [Indexed: 10/22/2022]
Abstract
Diazeniumdiolate anions and their prodrug forms are reliable sources of nitric oxide (NO) that have generated interest as promising therapeutic agents. A number of structural analogues of O(2)-(2,4-dinitro-5-(4-(N-methylamino)benzoyloxy)phenyl) 1-(N,N-dimethylamino)diazen-1-ium-1,2-diolate (PABA/NO), an anti-cancer lead compound that is designed to release NO upon activation by glutathione, were prepared. The nitric oxide release patterns of these O(2)-(2,4-dinitrophenyl) diazeniumdiolates in the presence of glutathione were tested and it was found that in the absence of competing pathways, these compounds release nearly quantitative amounts of NO. The ability of PABA/NO and its structural analogues to inhibit human leukemia cell proliferation was determined and it was found that compounds releasing elevated amounts of NO displayed superior cytotoxic effects.
Collapse
Affiliation(s)
- Harinath Chakrapani
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Thomas C. Wilde
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Michael L. Citro
- Basic Research Program, SAIC-Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Michael M. Goodblatt
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Larry K. Keefer
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | - Joseph E. Saavedra
- Basic Research Program, SAIC-Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| |
Collapse
|
41
|
Abstract
Impaired cutaneous nitric oxide (NO) production is associated with diminished antioxidative capacity in skin cells, hindered wound healing, unbalanced inflammatory reactions, and disturbed immunological responses. Use of topically applied NO donors might represent an auspicious new therapeutic approach in the field of dermatology. But what is the appropriate NO-generating compound or system? In this issue, Mowbray et al. describe a new chemical inert NO donor that per se produces little inflammation in the skin.
Collapse
Affiliation(s)
- Klaus-D Kröncke
- Institute of Biochemistry and Molecular Biology I, Medical Department of the Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | | |
Collapse
|
42
|
Chakrapani H, Goodblatt MM, Udupi V, Malaviya S, Shami PJ, Keefer LK, Saavedra JE. Synthesis and in vitro anti-leukemic activity of structural analogues of JS-K, an anti-cancer lead compound. Bioorg Med Chem Lett 2008; 18:950-3. [PMID: 18178089 PMCID: PMC2278236 DOI: 10.1016/j.bmcl.2007.12.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Accepted: 12/17/2007] [Indexed: 11/20/2022]
Abstract
Structural analogues of JS-K, an anti-cancer lead compound, were prepared and their in vitro anti-leukemic activity was determined. The rate of nitric oxide release from the corresponding diazeniumdiolate anions did not appear to affect the anti-leukemic activity of the prodrug forms. Two compounds with potent inhibitory activity and a potentially favorable toxicological profile were identified.
Collapse
Affiliation(s)
- Harinath Chakrapani
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, PO Box B, Frederick, MD 21702, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Scatena R. New nitric oxide-releasing polymers for implantable devices. Expert Opin Ther Pat 2007. [DOI: 10.1517/13543776.17.11.1397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Ishida H, Ray R, Ray P. Sulfur mustard downregulates iNOS expression to inhibit wound healing in a human keratinocyte model. J Dermatol Sci 2007; 49:207-16. [PMID: 17964119 DOI: 10.1016/j.jdermsci.2007.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2007] [Revised: 08/18/2007] [Accepted: 09/03/2007] [Indexed: 01/29/2023]
Abstract
BACKGROUND Increased nitric oxide (NO) synthesized by inducible NO synthase (iNOS) is involved in inflammatory and pathological conditions. iNOS also regulates several biomarkers that accelerate normal wound healing. Effects of exposure to sulfur mustard (SM) on the skin include formation of blisters and slow-healing injuries. Promoting re-epithelialization is a challenging issue in the treatment of the delayed healing of SM-induced skin injuries. OBJECTIVES To clarify the role(s) of iNOS in wound healing and the effect of SM on iNOS expression in an in vitro wound assay to eventually develop therapies for SM skin injuries. METHODS A wound was created by scratching normal human epidermal keratinocytes grown in vitro. iNOS expression was monitored by Western blotting, fluorescence microscopy, and real-time RT-PCR. Wound healing was analyzed using digitalized image analysis software. RESULTS The level of iNOS peaked 24-48h after wounding. SM exposure strongly reduced iNOS protein and mRNA levels. Fluorescence microscopy revealed that induction of iNOS expression by wounding and inhibition of iNOS expression by SM occurred not only in the cells at the wound edge but also in cells in the surrounding area, suggesting that wounding may induce and SM may inhibit release of cytokines that stimulate iNOS expression. iNOS-specific small interfering RNAs caused a marked decrease of iNOS expression irrespective of wounding. Gene silencing also completely inhibited wound healing. CONCLUSION These results suggest that preventing SM-induced inhibition of iNOS may be a prospective strategy to promote wound healing in SM-exposed skin.
Collapse
Affiliation(s)
- Hiroshi Ishida
- Molecular Biology Section, Department of Biology, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | | |
Collapse
|
45
|
Chakrapani H, Showalter BM, Citro ML, Keefer LK, Saavedra JE. Nitric Oxide Prodrugs: Diazeniumdiolate Anions of Hindered Secondary Amines. Org Lett 2007; 9:4551-4. [DOI: 10.1021/ol7019636] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Harinath Chakrapani
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, and Basic Research Program, SAIC Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702
| | - Brett M. Showalter
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, and Basic Research Program, SAIC Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702
| | - Michael L. Citro
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, and Basic Research Program, SAIC Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702
| | - Larry K. Keefer
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, and Basic Research Program, SAIC Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702
| | - Joseph E. Saavedra
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, and Basic Research Program, SAIC Frederick, National Cancer Institute at Frederick, Frederick, Maryland 21702
| |
Collapse
|
46
|
Griffith D, Bergamo A, Pin S, Vadori M, Müller-Bunz H, Sava G, Marmion CJ. Novel platinum pyridinehydroxamic acid complexes: Synthesis, characterisation, X-ray crystallographic study and nitric oxide related properties. Polyhedron 2007. [DOI: 10.1016/j.poly.2007.03.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
47
|
Whissell P, Persinger M. Developmental effects of perinatal exposure to extremely weak 7 Hz magnetic fields and nitric oxide modulation in the Wistar albino rat. Int J Dev Neurosci 2007; 25:433-9. [DOI: 10.1016/j.ijdevneu.2007.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Revised: 09/02/2007] [Accepted: 09/04/2007] [Indexed: 12/22/2022] Open
Affiliation(s)
- P.D. Whissell
- Neuroscience Research Group, Department of BiologyLaurentian UniversityCanada
| | - M.A. Persinger
- Neuroscience Research Group, Department of BiologyLaurentian UniversityCanada
- Behavioural Neuroscience and Biomolecular Sciences ProgramsLaurentian UniversitySudburyOntarioCanada
| |
Collapse
|
48
|
Abstract
Peripheral arterial disease (PAD) encompasses the vascular diseases caused primarily by atherosclerosis and thromboembolic pathophysiological processes that alter the normal structure and function of the aorta, its visceral arterial branches and the arteries of the upper and lower extremities. PAD is associated with an increased risk for cardiovascular morbidity and mortality. The goals for pharmacological therapy in PAD should focus on reducing cardiovascular risk, improving walking distance and preventing critical limb ischaemia. Exercise training plays a key role in the therapeutic assessment, as well stopping smoking. Antiplatelet therapy (aspirin) should be given to every PAD patient if there are no contraindications. Neither their combination nor anticoagulant therapy has shown additional benefit in PAD patients. Several pharmacological agents have been developed to improve the functional state of the claudicant and to relieve the symptoms. Many studied drugs have shown either no, a small or a potential benefit. With future development of new drugs for PAD, there is an absolute need for very strict well-designed protocols in order to evaluate the claudication distance, the progression of the disease and the reduction in cardiovascular morbidity and mortality. New developments should focus on improvement of endothelial function, vascular repair and enhancement of collateral circulation.
Collapse
Affiliation(s)
- Daniel A Duprez
- University of Minnesota, Cardiovascular Division, Medical School, VCRC-Room 270, Minneapolis, MN 55455, USA.
| |
Collapse
|
49
|
Chakrapani H, Showalter BM, Kong L, Keefer LK, Saavedra JE. V-PROLI/NO, a prodrug of the nitric oxide donor, PROLI/NO. Org Lett 2007; 9:3409-12. [PMID: 17658755 DOI: 10.1021/ol701419a] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The sensitivity to decomposition of the nitric oxide (NO) donor ion, 1-[2-(carboxylato)pyrrolidin-1-yl]diazen-1-ium-1,2-diolate (PROLI/NO), complicates direct electrophilic substitution to form useful prodrug derivatives. A modified general synthetic approach involving 1-[2-(hydroxymethyl)pyrrolidin-1-yl]diazen-1-ium-1,2-diolate ion (structure A, above) was used to prepare several PROLI/NO prodrugs including the previously inaccessible O2-vinyl derivative, V-PROLI/NO. Metabolism of V-PROLI/NO by liver microsomes enriched in human cytochrome P450 isoforms was demonstrated.
Collapse
Affiliation(s)
- Harinath Chakrapani
- Chemistry Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | | | | | | | | |
Collapse
|
50
|
Scatena R, Bottoni P, Botta G, Martorana GE, Giardina B. The role of mitochondria in pharmacotoxicology: a reevaluation of an old, newly emerging topic. Am J Physiol Cell Physiol 2007; 293:C12-21. [PMID: 17475665 DOI: 10.1152/ajpcell.00314.2006] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In addition to their well-known critical role in energy metabolism, mitochondria are now recognized as the location where various catabolic and anabolic processes, calcium fluxes, various oxygen-nitrogen reactive species, and other signal transduction pathways interact to maintain cell homeostasis and to mediate cellular responses to different stimuli. It is important to consider how pharmacological agents affect mitochondrial biochemistry, not only because of toxicological concerns but also because of potential therapeutic applications. Several potential targets could be envisaged at the mitochondrial level that may underlie the toxic effects of some drugs. Recently, antiviral nucleoside analogs have displayed mitochondrial toxicity through the inhibition of DNA polymerase-γ (pol-γ). Other drugs that target different components of mitochondrial channels can disrupt ion homeostasis or interfere with the mitochondrial permeability transition pore. Many known inhibitors of the mitochondrial electron transfer chain act by interfering with one or more of the respiratory chain complexes. Nonsteroidal anti-inflammatory drugs (NSAIDs), for example, may behave as oxidative phosphorylation uncouplers. The mitochondrial toxicity of other drugs seems to depend on free radical production, although the mechanisms have not yet been clarified. Meanwhile, drugs targeting mitochondria have been used to treat mitochondrial dysfunctions. Importantly, drugs that target the mitochondria of cancer cells have been developed recently; such drugs can trigger apoptosis or necrosis of the cancer cells. Thus the aim of this review is to highlight the role of mitochondria in pharmacotoxicology, and to describe whenever possible the main molecular mechanisms underlying unwanted and/or therapeutic effects.
Collapse
Affiliation(s)
- Roberto Scatena
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy.
| | | | | | | | | |
Collapse
|