1
|
Shi J, Liu Y, Jiao S, Wu T, Wang A, Wang W, Xie L, Liu Y, Wang H. Synthesis, structure, spectra, cytotoxicity and photo induced NO release of four isomeric nitrosylruthenium complexes. Nitric Oxide 2024; 152:58-68. [PMID: 39313019 DOI: 10.1016/j.niox.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Four isomeric nitrosyl ruthenium complexes [RuCl(2mqn)(Val)(NO)] (1-4) were prepared (2mqn, 2-methyl-8-hydroxyquinoline; Val, l-valine) and characterized by 1H NMR, 13C NMR, absorption spectrum, electrospray ionization mass spectrometry, and X-ray crystal diffraction. Time-resolved FT-IR and fluorescence spectroscopy were used to monitor photo-induced NO release in solution, while NO released in living cells was imaged using a selective fluorescent probe. The isomeric complexes showed different levels of cytotoxicity against HeLa cells, and slightly photo-enhanced anti-proliferative activity was observed. The isomeric complexes 1-4 inhibited the growth of HeLa cells by inducing apoptosis and promoted cell cycle arrest in the S phase. Furthermore, they showed relatively lower cytotoxicity against the human liver cell line HL-7702. The different spatial configurations of the complexes is close related with the selective binding of the isomeric complexes with serum albumin, which provide insight into the potential applications of the nitrosyl ruthenium complexes.
Collapse
Affiliation(s)
- Jia Shi
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, China; Fenyang College of Shanxi Medical University, Fenyang, 032200, China
| | - Yuhua Liu
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, China
| | - Shuxiang Jiao
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, China
| | - Tao Wu
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, China
| | - Ai Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, China
| | - Wenming Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, China
| | - Leilei Xie
- Experimental Management Center, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yanhong Liu
- Techinical Institute of Physics & Chemistry, CAS, Beijing, 100190, China
| | - Hongfei Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
2
|
Annaji M, Mita N, Heard J, Kang X, Poudel I, Boddu SHS, Tiwari AK, Babu RJ. Long-Acting Drug Delivery Technologies for Meloxicam as a Pain Medicine. Crit Rev Ther Drug Carrier Syst 2024; 41:111-150. [PMID: 38608134 DOI: 10.1615/critrevtherdrugcarriersyst.2024048988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Meloxicam, a selective COX-2 inhibitor, has demonstrated clinical effectiveness in managing inflammation and acute pain. Although available in oral and parenteral formulations such as capsule, tablet, suspension, and solution, frequent administration is necessary to maintain therapeutic efficacy, which can increase adverse effects and patient non-compliance. To address these issues, several sustained drug delivery strategies such as oral, transdermal, transmucosal, injectable, and implantable drug delivery systems have been developed for meloxicam. These sustained drug delivery strategies have the potential to improve the therapeutic efficacy and safety profile of meloxicam, thereby reducing the frequency of dosing and associated gastrointestinal side effects. The choice of drug delivery system will depend on the desired release profile, the target site of inflammation, and the mode of administration. Overall, meloxicam sustained delivery systems offer better patient compliance, and reduce the side effects, thereby improving the clinical applications of this drug. Herein, we discuss in detail different strategies for sustained delivery of meloxicam.
Collapse
Affiliation(s)
- Manjusha Annaji
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | | | - Jessica Heard
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Xuejia Kang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Ishwor Poudel
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, UAE; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, UAE
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
3
|
Madka V, Patlolla JMR, Venkatachalam K, Zhang Y, Pathuri G, Stratton N, Lightfoot S, Janakiram NB, Mohammed A, Rao CV. Chemoprevention of Colon Cancer by DFMO, Sulindac, and NO-Sulindac Administered Individually or in Combinations in F344 Rats. Cancers (Basel) 2023; 15:4001. [PMID: 37568816 PMCID: PMC10417047 DOI: 10.3390/cancers15154001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are promising colorectal cancer (CRC) chemopreventive drugs; however, to overcome NSAIDs' associated side effects, there is a need to develop safer and efficacious approaches. The present study was designed to evaluate (i) the efficacy of nitric-oxide releasing (NO)-Sulindac as compared to Sulindac; (ii) whether NO-Sulindac is superior to Sulindac in enhancing low-dose difluoromethylornithine (DFMO)-induced chemopreventive efficacy, and (iii) assessing the key biomarkers associated with colon tumor inhibition by these combinations. In F344 rats, colonic tumors were induced by azoxymethane (AOM). At the adenoma stage (13 weeks post AOM), groups of rats were fed the experimental diets containing 0 ppm, 500 ppm DFMO, 150 ppm Sulindac, and 200 ppm NO-Sulindac, individually or in combinations, for 36 weeks. Colon tumors were evaluated histopathologically and assayed for expression levels of proliferative, apoptotic, and inflammatory markers. Results suggest that (except for NO-Sulindac alone), DFMO, Sulindac individually, and DFMO combined with Sulindac or NO-Sulindac significantly suppressed AOM-induced adenocarcinoma incidence and multiplicities. DFMO and Sulindac suppressed adenocarcinoma multiplicity by 63% (p < 0.0001) and 51% (p < 0.0011), respectively, whereas NO-Sulindac had a modest effect (22.8%, p = 0.09). Combinations of DFMO plus Sulindac or NO-Sulindac suppressed adenocarcinoma incidence (60%, p < 0.0001; 50% p < 0.0004), and multiplicity (81%, p < 0.0001; 62%, p < 0.0001). Rats that were fed the combination of DFMO plus Sulindac showed significant inhibition of tumor cell proliferation and induction of apoptosis. In addition, enhancement of p21, Bax, and caspases; downregulation of Ki-67, VEGF, and β-catenin; and modulation of iNOS, COX-2, and ODC activities in colonic tumors were observed. These observations show that a lower-dose of DFMO and Sulindac significantly enhanced CRC chemopreventive efficacy when compared to NO-Sulindac alone, and the combination of DFMO and NO-Sulindac was modestly efficacious as compared to DFMO alone.
Collapse
Affiliation(s)
- Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Department of Internal Medicine, Hem-Onc Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.M.)
| | - Jagan M. R. Patlolla
- Center for Cancer Prevention and Drug Development, Department of Internal Medicine, Hem-Onc Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.M.)
| | - Karthikkumar Venkatachalam
- Center for Cancer Prevention and Drug Development, Department of Internal Medicine, Hem-Onc Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.M.)
| | - Yuting Zhang
- Center for Cancer Prevention and Drug Development, Department of Internal Medicine, Hem-Onc Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.M.)
| | - Gopal Pathuri
- Center for Cancer Prevention and Drug Development, Department of Internal Medicine, Hem-Onc Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.M.)
| | - Nicole Stratton
- Center for Cancer Prevention and Drug Development, Department of Internal Medicine, Hem-Onc Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.M.)
| | - Stanley Lightfoot
- Center for Cancer Prevention and Drug Development, Department of Internal Medicine, Hem-Onc Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.M.)
| | - Naveena B. Janakiram
- Translational Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD 20892, USA
| | - Altaf Mohammed
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20850, USA
| | - Chinthalapally V. Rao
- Center for Cancer Prevention and Drug Development, Department of Internal Medicine, Hem-Onc Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (V.M.)
- VA Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
4
|
Baylow HE, Esfandiarei M, Ratiu I. Swallowing and quality of life in individuals with Marfan syndrome: a cross-sectional study. Qual Life Res 2022; 31:3365-3375. [PMID: 35867322 DOI: 10.1007/s11136-022-03192-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Marfan syndrome (MFS) is a connective tissue disorder that affects skeletal, ocular, pulmonary, cardiovascular, and central nervous systems. Symptoms may lead to diminished quality of life (QoL) in individuals with MFS compared with healthy individuals. Currently, there is little evidence regarding the impact of MFS on swallowing and QoL. This study examined perceptions of swallowing difficulties and QoL among persons with MFS. METHOD A total of 356 participants (1% response rate) with a self-reported diagnosis of MFS provided medication regime and completed a Quality-of-Life Index and a modified version of the SWAL-QOL used to assess 8 QOL concepts related to swallowing: burden, duration, desire, food choice, fear, mental health, social concerns, and fatigue. RESULTS Dysphagia symptoms were reported by 62% (N = 356) of the participants. Analyses assessing correlations between responses to SWAL-QOL questions, QoL scores, and reported medications were conducted. Further, responses on the SWAL-QOL predicted QoL satisfaction, even after controlling for medications. Findings revealed that greater swallowing difficulty affects QoL satisfaction and overall QoL, but not QoL importance. Further, specific medications were associated with differences in swallowing difficulty as well as QoL satisfaction. CONCLUSIONS The findings of the current study suggest that individuals with MFS may experience specific swallowing difficulties which impact QoL. Specific classes of drugs may also be associated with reported swallowing QoL and QoL satisfaction in MFS. These findings have implications for clinicians who work with individuals with MFS.
Collapse
Affiliation(s)
- Hope E Baylow
- Department of Health and Human Performance, The University of Scranton, Edward Leahy Hall Rm. 824, 800 Linden St, Scranton, PA, 18510, USA.
| | - Mitra Esfandiarei
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Ileana Ratiu
- Department of Speech and Hearing Science, Arizona State University, Tempe, AZ, USA
- Speech-Language Pathology Program, Midwestern University, Glendale, AZ, USA
| |
Collapse
|
5
|
Saletti M, Maramai S, Reale A, Paolino M, Brogi S, Di Capua A, Cappelli A, Giorgi G, D'Avino D, Rossi A, Ghelardini C, Di Cesare Mannelli L, Sardella R, Carotti A, Woelkart G, Klösch B, Bigogno C, Dondio G, Anzini M. Novel analgesic/anti-inflammatory agents: 1,5-Diarylpyrrole nitrooxyethyl sulfides and related compounds as Cyclooxygenase-2 inhibitors containing a nitric oxide donor moiety endowed with vasorelaxant properties. Eur J Med Chem 2022; 241:114615. [DOI: 10.1016/j.ejmech.2022.114615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022]
|
6
|
Ribeiro H, Rodrigues I, Napoleão L, Lira L, Marques D, Veríssimo M, Andrade JP, Dourado M. Non-steroidal anti-inflammatory drugs (NSAIDs), pain and aging: Adjusting prescription to patient features. Biomed Pharmacother 2022; 150:112958. [PMID: 35453005 DOI: 10.1016/j.biopha.2022.112958] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/02/2022] Open
Abstract
A narrative review of papers published from January 2011 to December 2021, after a literature search in selected databases using the terms "pharmacokinetics", "ibuprofen", "diclofenac", "acemetacin", "naproxen", "etodolac" and "etoricoxib" was performed. From 828 articles identified, only eight met the inclusion criteria. Selective COX-2 inhibitors are associated with higher cardiovascular risk, while non-selective COX inhibitors are associated with higher gastrointestinal risk. NSAIDs with lower renal excretion with phase 2 metabolism are less likely to induce adverse effects and drug-drug interactions. Patients with frequent NSAID use needs, such as elderly patients and patients with cardiovascular disease or impaired renal function, will benefit from lower renal excretion (e.g. acemethacin, diclofenac, and etodolac) (level of evidence 3). Polymedicated patients, elderly patients, and patients with chronic alcohol abuse will be at a lower risk for adverse effects with NSAIDs that undergo phase 2 liver biotransformation, namely, acemethacin and diclofenac (level of evidence 3). Young patients, patients dealing with acute pain, or with active and/or chronic symptomatic gastritis, selective COX-2 inhibitors (celecoxib or etoricoxib) may be a better option (level of evidence 2). Knowing the individual characteristics of the patients, combined with knowledge on basic pharmacology, offers greater safety and better adherence to therapy. PERSPECTIVE: Although there are several NSAIDs options to treat pain, physicians usually take special care to its prescription regarding cardiovascular and gastrointestinal side effects, despite the age of the patient. In this paper, based on the best evidence, the authors present a review of the safest NSAIDs to use in the elderly.
Collapse
Affiliation(s)
- Hugo Ribeiro
- Palliative Care Unit of Health Centers Cluster Gaia, PhD Palliative Care Student in Faculty of Medicine of University of Porto, Invited Assistant of Faculty of Medicine of University of Coimbra, Portugal.
| | - Inês Rodrigues
- Family Health Unit Barão do Corvo of Health Centers Cluster Gaia, Portugal
| | - Leonardo Napoleão
- Family Health Unit Canelas of Health Centers Cluster Espinho/Gaia, Portugal
| | - Luís Lira
- Family Health Unit Barão do Corvo of Health Centers Cluster Gaia, Portugal
| | | | - Manuel Veríssimo
- Coimbra University Hospital and Faculty of Medicine of University of Coimbra, Portugal
| | - José Paulo Andrade
- Department of Biomedicine - Unit of Anatomy, Faculty of Medicine of University of Porto, Portugal
| | | |
Collapse
|
7
|
Xie L, Bai H, Song L, Liu C, Gong W, Wang W, Zhao X, Takemoto C, Wang H. Structural and Photodynamic Studies on Nitrosylruthenium-Complexed Serum Albumin as a Delivery System for Controlled Nitric Oxide Release. Inorg Chem 2021; 60:8826-8837. [PMID: 34060309 DOI: 10.1021/acs.inorgchem.1c00762] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
How to deliver nitric oxide (NO) to a physiological target and control its release quantitatively is a key issue for biomedical applications. Here, a water-soluble nitrosylruthenium complex, [(CH3)4N][RuCl3(5cqn)(NO)] (H5cqn = 5-chloro-8-quinoline), was synthesized, and its structure was confirmed with 1H NMR and X-ray crystal diffraction. Photoinduced NO release was investigated with time-resolved Fourier transform infrared and electron paramagnetic resonance (EPR) spectroscopies. The binding constant of the [RuCl3(5cqn)(NO)]- complex with human serum albumin (HSA) was determined by fluorescence spectroscopy, and the binding mode was identified by X-ray crystallography of the HSA and Ru-NO complex adduct. The crystal structure reveals that two molecules of the Ru-NO complex are located in the subdomain IB, which is one of the major drug binding regions of HSA. The chemical structures of the Ru complexes were [RuCl3(5cqn)(NO)]- and [RuCl3(Glycerin)NO]-, in which the electron densities for all ligands to Ru are unambiguously identified. EPR spin-trapping data showed that photoirradiation triggered NO radical generation from the HSA complex adduct. Moreover, the near-infrared image of exogenous NO from the nitrosylruthenium complex in living cells was observed using a NO-selective fluorescent probe. This study provides a strategy to design an appropriate delivery system to transport NO and metallodrugs in vivo for potential applications.
Collapse
Affiliation(s)
- Leilei Xie
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Hehe Bai
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Luna Song
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Chenyang Liu
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Wenjun Gong
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Wenming Wang
- Key Laboratory of Pharmaceutical Biotechnology of Shanxi Provence, Shanxi, Taiyuan 030006, China
| | - Xuan Zhao
- Department of Chemistry, University of Memphis, Memphis, Tennessee 38152, United States
| | - Chie Takemoto
- RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa 230-0045, Japan
| | - Hongfei Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
8
|
Guo CG, Leung WK. Potential Strategies in the Prevention of Nonsteroidal Anti-inflammatory Drugs-Associated Adverse Effects in the Lower Gastrointestinal Tract. Gut Liver 2021; 14:179-189. [PMID: 31547642 PMCID: PMC7096237 DOI: 10.5009/gnl19201] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 07/09/2019] [Indexed: 12/21/2022] Open
Abstract
With the increasing use of nonsteroidal anti-inflammatory drugs (NSAIDs), the incidence of lower gastrointestinal (GI) complications is expected to increase. However, unlike upper GI complications, the burden, pathogenesis, prevention and treatment of NSAID-associated lower GI complications remain unclear. To date, no cost-effective and safe protective agent has been developed that can completely prevent or treat NSAID-related lower GI injuries. Selective COX-2 inhibitors, misoprostol, intestinal microbiota modulation, and some mucoprotective agents have been reported to show protective effects on NSAID-induced lower GI injuries. This review aims to provide an overview of the current evidence on the prevention of NSAID-related lower GI injuries.
Collapse
Affiliation(s)
- Chuan-Guo Guo
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Wai K Leung
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
9
|
Dillon KM, Carrazzone RJ, Matson JB, Kashfi K. The evolving landscape for cellular nitric oxide and hydrogen sulfide delivery systems: A new era of customized medications. Biochem Pharmacol 2020; 176:113931. [PMID: 32224139 PMCID: PMC7263970 DOI: 10.1016/j.bcp.2020.113931] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/20/2020] [Indexed: 02/09/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are industrial toxins or pollutants; however, both are produced endogenously and have important biological roles in most mammalian tissues. The recognition that these gasotransmitters have a role in physiological and pathophysiological processes has presented opportunities to harness their intracellular effects either through inhibition of their production; or more commonly, through inducing their levels and or delivering them by various modalities. In this review article, we have focused on an array of NO and H2S donors, their hybrids with other established classes of drugs, and the various engineered delivery platforms such a fibers, polymers, nanoparticles, hydrogels, and others. In each case, we have reviewed the rationale for their development.
Collapse
Affiliation(s)
- Kearsley M Dillon
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, USA; Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, USA
| | - Ryan J Carrazzone
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, USA; Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, USA
| | - John B Matson
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, USA; Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, 160 Convent Avenue, New York, NY 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, NY, USA.
| |
Collapse
|
10
|
Feng CW, Chen NF, Sung CS, Kuo HM, Yang SN, Chen CL, Hung HC, Chen BH, Wen ZH, Chen WF. Therapeutic Effect of Modulating TREM-1 via Anti-inflammation and Autophagy in Parkinson's Disease. Front Neurosci 2019; 13:769. [PMID: 31440123 PMCID: PMC6691936 DOI: 10.3389/fnins.2019.00769] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/09/2019] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common age-related neurodegenerative diseases, and neuroinflammation has been identified as one of its key pathological characteristics. Triggering receptors expressed on myeloid cells-1 (TREM-1) amplify the inflammatory response and play a role in sepsis and cancer. Recent studies have demonstrated that the attenuation of TREM-1 activity produces cytoprotective and anti-inflammatory effects in macrophages. However, no study has examined the role of TREM-1 in neurodegeneration. We showed that LP17, a synthetic peptide blocker of TREM-1, significantly inhibited the lipopolysaccharide (LPS)-induced upregulation of proinflammatory cascades of inducible nitric oxide synthase (iNOS), cyclooxygenase-2, and nuclear factor-kappa B. Moreover, LP17 enhanced the LPS-induced upregulation of autophagy-related proteins such as light chain-3 and histone deacetylase-6. We also knocked down TREM-1 expression in a BV2 cell model to further confirm the role of TREM-1. LP17 inhibited 6-hydroxydopamine-induced locomotor deficit and iNOS messenger RNA expression in zebrafish. We also observed therapeutic effects of LP17 administration in 6-hydroxydopamine-induced PD syndrome using a rat model. These data suggest that the attenuation of TREM-1 could ameliorate neuroinflammatory responses in PD and that this neuroprotective effect might occur via the activation of autophagy and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Chien-Wei Feng
- National Museum of Marine Biology & Aquarium, Pingtung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung City, Taiwan.,Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung City, Taiwan.,Center for Neuroscience, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - San-Nan Yang
- Department of Pediatrics, E-Da Hospital, Kaohsiung City, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung City, Taiwan
| | - Chien-Liang Chen
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan.,Department of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Han-Chun Hung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung City, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung City, Taiwan.,Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung City, Taiwan.,Department of Neurosurgery, Xiamen Chang Gung Hospital, Xiamen, China
| |
Collapse
|
11
|
Affiliation(s)
- Nathan S. Bryan
- Department of Molecular and Human GeneticsBaylor College of Medicine One Baylor Plaza Alkek Building for Biomedical Research R-850 Houston TX 77030
| |
Collapse
|
12
|
Kłobucki M, Urbaniak A, Grudniewska A, Kocbach B, Maciejewska G, Kiełbowicz G, Ugorski M, Wawrzeńczyk C. Syntheses and cytotoxicity of phosphatidylcholines containing ibuprofen or naproxen moieties. Sci Rep 2019; 9:220. [PMID: 30659229 PMCID: PMC6338774 DOI: 10.1038/s41598-018-36571-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 11/22/2018] [Indexed: 12/12/2022] Open
Abstract
In this study, novel phosphatidylcholines containing ibuprofen or naproxen moieties were synthesized in good yields and high purities. Under the given synthesis conditions, the attached drug moieties racemized, which resulted in the formation of phospholipid diastereomers. The comperative studies of the cytotoxicity of ibuprofen, naproxen and their phosphatidylcholine derivatives against human promyelocytic leukemia HL-60, human colon carcinoma Caco-2, and porcine epithelial intestinal IPEC-J2 cells were carried out. The results of these studies indicated that phospholipids with NSAIDs at both sn-1 and sn-2 positions (15 and 16) were more toxic than ibuprofen or naproxen themselves, whereas 2-lysophosphatidylcholines (7 and 8) were less toxic against all tested cell lines. Phospholipids with NSAIDs at sn-1 and palmitic acid at sn-2 (9 and 10) were also less toxic against Caco-2 and normal cells (IPEC-J2).
Collapse
Affiliation(s)
- Marek Kłobucki
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland
| | - Anna Urbaniak
- Laboratory of Glycobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Aleksandra Grudniewska
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland
| | - Bartłomiej Kocbach
- Laboratory of Glycobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Gabriela Maciejewska
- Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| | - Grzegorz Kiełbowicz
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland
| | - Maciej Ugorski
- Laboratory of Glycobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
- Department of Biochemistry and Molecular Biology, Wrocław University of Environmental and Life Sciences, Norwida 31, 50-375, Wrocław, Poland
| | - Czesław Wawrzeńczyk
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland.
| |
Collapse
|
13
|
Wallace JL. Nitric oxide in the gastrointestinal tract: opportunities for drug development. Br J Pharmacol 2018; 176:147-154. [PMID: 30357812 DOI: 10.1111/bph.14527] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/12/2022] Open
Abstract
Nitric oxide (NO) plays important roles in gastrointestinal mucosal defence, as well as in the pathogenesis of several gastrointestinal diseases (e.g. irritable bowel syndrome and inflammatory bowel disease). The potent cytoprotective effects of NO have been demonstrated in a range of animal models. However, in some disease states, inhibition of NO synthesis is beneficial. Several attempts have been made to develop drugs for ulcerative and/or inflammatory disorders of the gastrointestinal tract, with varying degrees of success. Covalently linking a NO-releasing group to non-steroidal anti-inflammatory drugs or to drugs used in the treatment of inflammatory bowel disease and irritable bowel syndrome has shown some benefit, although no drug of this type has yet been fully developed. LINKED ARTICLES: This article is part of a themed section on Nitric Oxide 20 Years from the 1998 Nobel Prize. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.2/issuetoc.
Collapse
Affiliation(s)
- John L Wallace
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Aisa MC, Datti A, Orlacchio A, Di Renzo GC. COX inhibitors and bone: A safer impact on osteoblasts by NO-releasing NSAIDs. Life Sci 2018; 208:10-19. [PMID: 29990484 DOI: 10.1016/j.lfs.2018.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/03/2018] [Accepted: 07/06/2018] [Indexed: 12/27/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are commonly prescribed for the treatment of pain and inflammation. Although it is well known that NSAIDs can suppress bone growth, remodelling and repair, they are largely used post-operatively and post-traumatically to achieve analgesia and reduce inflammation in bone tissue. AIMS The impact of two NO-releasing, non-selective NSAIDs, NCX-4016 and HCT-3012 (NO-derivatives of Aspirin and Naproxen, respectively) on osteoblasts were evaluated and compared to the non-selective, parent chemicals and to the COX-2-selective inhibitor Celecoxib. MAIN METHODS Using MG-63 osteoblast-like cells, we considered proliferation, the early and late stage of differentiation, and the activity of proteinases thought to be involved in osteoid degradation, a preliminary fundamental event of bone remodelling. KEY FINDINGS Unlike Aspirin, Naproxen and Celecoxib, the two NO-NSAIDs did not alter proliferation and differentiation of osteoblasts. They also reduced the activity of plasminogen activator, metalloproteinases, and cathepsin B. Similar inhibitory effects against these proteinases were recapitulated by the NO-donor sodium nitroprusside, thereby suggesting a NO-mediated mechanism. SIGNIFICANCE Due to a differential effect on cell proliferation and differentiation, the two NO-NSAIDs exhibit a safer impact on osteoblast metabolism compared to Celecoxib and their parent compounds. This suggests an advantageous option for these drugs in individuals with a need of COX-inhibiting treatment, in general. In addition, their capability of modulating the proteinases involved in osteoid degradation may specifically suggest an additional safer use in comorbidity conditions of inflammation or pain with bone disorders characterized by high rate of remodelling, such as high-turnover osteoporosis in post-menopausal women.
Collapse
Affiliation(s)
- Maria Cristina Aisa
- Department of Surgery and Biomedical Sciences, Section of Obstetrics and Gynecology, University of Perugia, Perugia, Italy.
| | - Alessandro Datti
- Department of Agricultural, Food and Environmental Sciences, Section of Biochemistry and Molecular Biology, University of Perugia, Perugia, Italy
| | - Antonio Orlacchio
- Laboratory of Neurogenetics, European Centre for Brain Research (CERC) - Institute for Research and Health Care (IRCCS) Santa Lucia, Rome, Italy; Department of Surgery and Biomedical Sciences, Section of Medical Genetics, University of Perugia, Perugia, Italy
| | - Gian Carlo Di Renzo
- Department of Surgery and Biomedical Sciences, Section of Obstetrics and Gynecology, University of Perugia, Perugia, Italy
| |
Collapse
|
15
|
Kashfi K. The dichotomous role of H 2S in cancer cell biology? Déjà vu all over again. Biochem Pharmacol 2018; 149:205-223. [PMID: 29397935 PMCID: PMC5866221 DOI: 10.1016/j.bcp.2018.01.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/17/2018] [Indexed: 02/09/2023]
Abstract
Nitric oxide (NO) a gaseous free radical is one of the ten smallest molecules found in nature, while hydrogen sulfide (H2S) is a gas that bears the pungent smell of rotten eggs. Both are toxic yet they are gasotransmitters of physiological relevance. There appears to be an uncanny resemblance between the general actions of these two gasotransmitters in health and disease. The role of NO and H2S in cancer has been quite perplexing, as both tumor promotion and inflammatory activities as well as anti-tumor and antiinflammatory properties have been described. These paradoxes have been explained for both gasotransmitters in terms of each having a dual or biphasic effect that is dependent on the local flux of each gas. In this review/commentary, I have discussed the major roles of NO and H2S in carcinogenesis, evaluating their dual nature, focusing on the enzymes that contribute to this paradox and evaluate the pros and cons of inhibiting or inducing each of these enzymes.
Collapse
Affiliation(s)
- Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA.
| |
Collapse
|
16
|
Srinivasan A, De Cruz P. Review article: a practical approach to the clinical management of NSAID enteropathy. Scand J Gastroenterol 2017; 52:941-947. [PMID: 28587496 DOI: 10.1080/00365521.2017.1335769] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Co-prescription of acid suppressive therapy, together with advances in small bowel imaging techniques, have shifted the burden of NSAID-related toxicity from gastro-duodenal to more distal small bowel injury. Due to predominantly subclinical disease, NSAID enteropathy remains under-recognised, with an incidence of 53-80% amongst healthy short-term users, and a prevalence of 50-71% following long-term (>3 months) use. Despite their distinct pathogenesis, those at risk of NSAID-related gastro-duodenal and small bowel complications share several risk factors. Clinical complications of NSAID enteropathy such as protein-losing enteropathy, small bowel strictures and diaphragm disease, confer significant morbidity, and are often irreversible. Small bowel prophylaxis has proven of modest efficacy after short-term, high-dose NSAID use in asymptomatic patients. While selective COX-2 inhibitors are associated with fewer gastro-duodenal complications relative to non-selective NSAIDs, their comparative benefit in protecting against small bowel enteropathy remains unclear. Prophylaxis should be considered in those at high risk of small bowel complications, as treatment options for established disease remain limited; however, the optimal agent remains unclear. We propose a clinical algorithm that may help prevent, monitor, investigate, and manage the sequelae of NSAID-induced small bowel toxicity.
Collapse
Affiliation(s)
- Ashish Srinivasan
- a Department of Gastroenterology , Austin Health , Melbourne , Australia
| | - Peter De Cruz
- a Department of Gastroenterology , Austin Health , Melbourne , Australia.,b Department of Medicine , University of Melbourne , Melbourne , Australia
| |
Collapse
|
17
|
Bilani N, Bahmad H, Abou-Kheir W. Prostate Cancer and Aspirin Use: Synopsis of the Proposed Molecular Mechanisms. Front Pharmacol 2017; 8:145. [PMID: 28377721 PMCID: PMC5359278 DOI: 10.3389/fphar.2017.00145] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/07/2017] [Indexed: 12/18/2022] Open
Abstract
Background: Prostate cancer (PCa) is a critical health burden, impacting the morbidity and mortality of millions of men around the world. Most of the patients with PCa have their disease at first sensitive to androgen deprivation treatments, but later they develop resistance to therapy and eventually die of metastatic castration-resistant prostate cancer (CRPC). Although the newly developed anti-androgen therapies are effectively alleviating symptoms and prolonging lives of patients, there are still no curable treatments for CRPC. Recently, statistical studies have shown that the chronic use of aspirin might be significantly associated with better outcomes in PCa patients. Through this review, we aim to identify the different proposed molecular mechanisms relating aspirin to the pathobiology of PCa neoplasms, with a major focus on basic research done in this context. Methods: Articles were retrieved via online database searching of PubMed and MEDLINE between 1946 and September 2016. Keywords and combinations related to PCa and aspirin were used to perform the search. Abstracts of the articles were studied by two independent reviewers and then data extraction was performed on the relevant articles that met our review objectives. Results: Aspirin, a non-steroidal anti-inflammatory drug (NSAID), affects the proliferation, apoptosis, resistance and metastasis of PCa cell lines, through both COX-dependent and COX-independent mechanisms. It also lowers levels of the PCa diagnostic marker prostate specific antigen (PSA), suggesting that clinicians need to at least be aware if their patients are using Aspirin chronically. Conclusion: This review strongly warrants further consideration of the signaling cascades activated by aspirin, which may lead to new knowledge that might be applied to improve diagnosis, prognosis and treatment of PCa.
Collapse
Affiliation(s)
- Nadeem Bilani
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Hisham Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| |
Collapse
|
18
|
Tachecí I, Kopáčová M, Rejchrt S, Bureš J. Non-steroidal Anti-inflammatory Drug Induced Injury to the Small Intestine. ACTA MEDICA (HRADEC KRÁLOVÉ) 2016. [DOI: 10.14712/18059694.2016.56] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Non-steroidal anti-inflammatory drug (NSAIDs) induced enteropathy represents an important complication of one of the most commonly used drugs worldwide. Due to previous diagnostics difficulties the real prevalence of this disease was underestimated for a long time. The pathogenesis of NSAID-enteropathy is more multifactorial and complex than formerly assumed but has still not been fully uncovered. A combination of the local and systemic effect plays an important role in pathogenesis. Thanks to novel enteroscopy methods (wireless capsule endoscopy, double balloon enteroscopy), small bowel lesions are described in a substantial section of NSAID users although most are clinically asymptomatic. The other non-invasive tests (small bowel permeability, faecal calprotectin, scintigraphy using faecal excretion of 111-indium-labelled leukocytes etc.) proposed for diagnostics are not generally used in clinical practice, mainly because of their non-specificity. Despite intensive research into possible treatment, the main measure for patients with NSAID-enteropathy is still withdrawal of NSAIDs. Double balloon enteroscopy plays an important role in the treatment of complications (bleeding, strictures).
Collapse
|
19
|
Amir M, Akhter MW, Alam O. Synthesis, characterization, and biological evaluation of furoxan coupled ibuprofen derivatives as anti-inflammatory agents. MONATSHEFTE FUR CHEMIE 2015. [DOI: 10.1007/s00706-015-1557-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Ragot T, Provost C, Prignon A, Cohen R, Lepoivre M, Lausson S. Apoptosis induction by combination of drugs or a conjugated molecule associating non-steroidal anti-inflammatory and nitric oxide donor effects in medullary thyroid cancer models: implication of the tumor suppressor p73. Thyroid Res 2015; 8:13. [PMID: 26273323 PMCID: PMC4535850 DOI: 10.1186/s13044-015-0025-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 08/02/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Medullary thyroid cancer (MTC) is a C-cell neoplasm. Surgery remains its main treatment. Promising therapies based on tyrosine kinase inhibitors demand careful patient selection. We previously observed that two non-steroidal anti-inflammatory drugs (NSAID), indomethacin, celecoxib, and nitric oxide (NO) prevented tumor growth in a model of human MTC cell line (TT) in nude mice. METHODS In the present study, we tested the NO donor: glyceryl trinitrate (GTN), at pharmacological dose, alone and in combination with each of the two NSAIDs on TT cells. We also assessed the anti-proliferative potential of NO-indomethacin, an indomethacin molecule chemically conjugated with a NO moiety (NCX 530, Nicox SA) on TT cells and indomethacin/GTN association in rMTC 6-23 cells. The anti-tumoral action of the combined sc. injections of GTN with oral delivery of indomethacin was also studied on subcutaneous TT tumors in nude mice. Apoptosis mechanisms were assessed by expression of caspase-3, TAp73α, TAp73α inhibition by siRNA or Annexin V externalisation. RESULTS The two NSAIDs and GTN reduced mitotic activity in TT cells versus control (cell number and PCNA protein expression). The combined treatments amplified the anti-tumor effect of single agents in the two tested cell lines and promoted cell death. Moreover, indomethacin/GTN association stopped the growth of established TT tumors in nude mice. We observed a significant cleavage of full length PARP, a caspase-3 substrate. The cell death appearance was correlated with a two-fold increase in TAp73α expression, with inhibition of apoptosis after TAp73α siRNA addition, demonstrating its crucial role in apoptosis. CONCLUSION Association of NO with NSAID exhibited amplified anti-tumoral effects on in vitro and in vivo MTC models by inducing p73-dependent apoptotic cell death.
Collapse
Affiliation(s)
- Thierry Ragot
- UMR 8203, Gustave Roussy, Laboratoire de Vectorologie et de Thérapeutiques Anticancéreuses, Villejuif, 94805 France ; UMR 8203, CNRS, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, Villejuif, 94805 France ; UMR 8203, Univ Paris-Sud, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, Villejuif, 94805 France
| | - Claire Provost
- Sorbonne Universités, UPMC University Paris 06, plateforme LIMP, Laboratoire d'Imagerie Médicale Positonique, Hôpital Tenon, Paris, 75020 France
| | - Aurélie Prignon
- Sorbonne Universités, UPMC University Paris 06, plateforme LIMP, Laboratoire d'Imagerie Médicale Positonique, Hôpital Tenon, Paris, 75020 France
| | - Régis Cohen
- Hopital Delafontaine, Endocrinology Unit, Saint Denis, France
| | - Michel Lepoivre
- IBBMC, CNRS 8619, bat 430, Université Paris Sud XI, Orsay, Paris, 91405 France
| | - Sylvie Lausson
- Sorbonne Universités, UPMC University Paris 06, plateforme LIMP, Laboratoire d'Imagerie Médicale Positonique, Hôpital Tenon, Paris, 75020 France
| |
Collapse
|
21
|
Wang X, Diao Y, Liu Y, Gao N, Gao D, Wan Y, Zhong J, Jin G. Synergistic apoptosis-inducing effect of aspirin and isosorbide mononitrate on human colon cancer cells. Mol Med Rep 2015; 12:4750-4758. [PMID: 26094902 DOI: 10.3892/mmr.2015.3963] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 05/27/2015] [Indexed: 11/05/2022] Open
Abstract
Aspirin and isosorbide mononitrate (ISMN) are two commonly used drugs, which are clinically applied for the treatment of inflammatory and cardiovascular diseases, respectively. Recently, aspirin has attracted interest due to its potential application for the treatment of cancer, particularly colon cancer. NO-aspirin, an aspirin derivative containing a covalently bound NO-donating moiety, has been proven to be an effective anti‑tumor agent with apoptosis-inducing ability. In the present study, ISMN was used as an NO donor and its synergic effect with aspirin was assessed in human colon cancer cells. In vitro, an MTT assay demonstrated that ISMN had a synergistic effect on the growth inhibitory effects of aspirin on HCT116 and SW620 colon cancer cells, while the growth of EA.hy926 normal endothelial cells was unaffected. This synergistic anti‑tumor effect was further validated in vivo using nude mouse HCT116 cell xenograft model. Observation of nuclear morphology, Annexin V-fluorescein isothiocyanate/propidium iodide double staining and a caspase-3 activity assay suggested that the combination of the two drugs induced apoptosis in HCT116 cells. Furthermore, the molecular mechanisms of the apoptotic effect of the drugs was assessed using an NO release assay, reverse transcription quantitative polymerase chain reaction analysis, western blot analysis and a luciferase reporter assay. It was certified that the increase in the amount of NO release, the decrease in the luciferase promoter activity and the expression of cyclin D1 and c-myc in HCT116 cells were affected by aspirin and ISMN in a synergistic manner. In conclusion, the present study was the first, to the best of our knowledge, to report on the synergistic apoptosis-inducing effects of aspirin and ISMN in human colon cancer cells, which were mediated via Wnt and NO signaling pathways. The results of the present study will facilitate the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Xiaodong Wang
- Shenzhen Engineering Laboratory of Synthetic Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Yuwen Diao
- Shenzhen Engineering Laboratory of Synthetic Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Yu Liu
- Shenzhen Engineering Laboratory of Synthetic Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Ningning Gao
- Shenzhen Engineering Laboratory of Synthetic Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Dong Gao
- Shenzhen Engineering Laboratory of Synthetic Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Yanyan Wan
- Shenzhen Engineering Laboratory of Synthetic Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Jingjing Zhong
- Shenzhen Engineering Laboratory of Synthetic Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Guangyi Jin
- Shenzhen Engineering Laboratory of Synthetic Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
22
|
Basudhar D, Cheng RC, Bharadwaj G, Ridnour LA, Wink DA, Miranda KM. Chemotherapeutic potential of diazeniumdiolate-based aspirin prodrugs in breast cancer. Free Radic Biol Med 2015; 83:101-14. [PMID: 25659932 PMCID: PMC4441830 DOI: 10.1016/j.freeradbiomed.2015.01.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/05/2015] [Accepted: 01/13/2015] [Indexed: 12/12/2022]
Abstract
Diazeniumdiolate-based aspirin prodrugs have previously been shown to retain the anti-inflammatory properties of aspirin while protecting against the common side effect of stomach ulceration. Initial analysis of two new prodrugs of aspirin that also release either nitroxyl (HNO) or nitric oxide (NO) demonstrated increased cytotoxicity toward human lung carcinoma cells compared to either aspirin or the parent nitrogen oxide donor. In addition, cytotoxicity was significantly lower in endothelial cells, suggesting cancer-specific sensitivity. To assess the chemotherapeutic potential of these new prodrugs in treatment of breast cancer, we studied their effect both in cultured cells and in a nude mouse model. Both prodrugs reduced growth of breast adenocarcinoma cells more effectively than the parent compounds while not being appreciably cytotoxic in a related nontumorigenic cell line (MCF-10A). The HNO donor also was more cytotoxic than the related NO donor. The basis for the observed specificity was investigated in terms of impact on metabolism, DNA damage and repair, apoptosis, angiogenesis and metastasis. The results suggest a significant pharmacological potential for treatment of breast cancer.
Collapse
Affiliation(s)
- Debashree Basudhar
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Robert C Cheng
- Radiation Biology Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gaurav Bharadwaj
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Lisa A Ridnour
- Radiation Biology Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A Wink
- Radiation Biology Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katrina M Miranda
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
23
|
Sahoo NK, Sahu M, Rao PS, Ghosh G. Solid phase extraction and quantification of diclofenac sodium in human plasma by liquid chromatography-tandem mass spectrometry. JOURNAL OF ANALYTICAL CHEMISTRY 2015. [DOI: 10.1134/s1061934815040115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Abstract
Although Andre Robert's historic article on "gastric cytoprotection" in 1979 introduced this new name and concept, gastroprotective drugs (e.g. sofalcone, sucralfate), which prevent and/or accelerate healing of gastric ulcers without inhibiting acid secretion, were known in Japan before or around that time. But since Robert's studies were solely focused on prostaglandins (PG), they became the center of gastrointestinal research for more than 30 years. As endogenous products, PG were implicated in mediating the gastroprotective effect of other drugs such as sofalcone and sucralfate, despite that the cyclooxygenase inhibitor indomethacin diminished but never abolished gastroprotection by other drugs. Another group of endogenous substances, that is, sulfhydryls (SH), investigated in parallel with PG, also seem to play a mechanistic role in gastroprotection, especially since SH alkylators like N-ethylmaleimide counteract virtually any form of gastroprotection. In Robert's terms of "prevention of chemically induced acute mucosal lesions," so far no single mechanism could explain the beneficial effects of diverse protective agents, but I argue that these two endogenous substances (i.e. PG, SH), in addition to histamine, are the main mechanistic mediators of acute gastroprotection: PG and histamine, because as mediators of acute inflammation, they increase vascular permeability (VP), and SH scavenge free radicals. This is contrary to the search for a single mechanism of action, long focused on enhanced secretion of mucus and/or bicarbonate that may contribute but cannot explain all forms of gastroprotection. Nevertheless, based on research work of the last 30 years, in part from our lab, a new mechanistic explanation of gastroprotection may be formulated: it's a complex but orderly and evolution-based physiologic response of the gastric mucosa under pathologic conditions. Namely, one of the first physiologic defense responses of any organ is inflammation that starts with rapid vascular changes (e.g. increased VP and blood flow), followed by cellular events (e.g. infiltration by acute and chronic inflammatory cells). Thus, PG and histamine, by increasing VP create a perivascular edema that dilutes and delays toxic agents reaching the subepithelial capillaries. Otherwise, damaging chemicals may induce severe early vascular injury resulting in blood flow stasis, hypoxia, and necrosis of surrounding epithelial and mesenchymal cells. In this complex response, increased mucus and/or bicarbonate secretion seem to cause luminal dilution of gastrotoxic chemicals that is further reinforced by a perivascular, histodilutional component. This mechanistic explanation would encompass the protective actions of diverse agents as PG, small doses of histamine, motility stimulants, and dilute irritants (i.e. "adaptive cytoprotection"). Thus, although markedly increased VP is pathologic, slight increase in VP seems to be protective, that is, a key element in the complex pathophysiologic response during acute gastroprotection. Over the years, "gastroprotection" was also applied to accelerated healing of chronic gastroduodenal ulcers without reduction of acid secretion. The likely main mechanism here is the binding of angiogenic growth factors (e.g. basic fibroblast growth factor, vascular endothelial growth factor) to the heparin-like structures of sucralfate and sofalcone. Thus, despite intensive research of the last 30 years, gastroprotection is incompletely understood, and we are still far away from effectively treating Helicobacter pylori-negative ulcers and preventing nonsteroidal anti-inflammatory drugs-caused erosions and ulcers in the upper and lower gastrointestinal tract; hence "gastric cytoprotection" research is still relevant.
Collapse
Affiliation(s)
- Sandor Szabo
- Departments of Pathology and Pharmacology, University of California-Irvine and VA Medical Center, Long Beach, California, USA
| |
Collapse
|
25
|
Kim HS, Kim T, Kim MK, Suh DH, Chung HH, Song YS. Cyclooxygenase-1 and -2: molecular targets for cervical neoplasia. J Cancer Prev 2014; 18:123-34. [PMID: 25337538 PMCID: PMC4189449 DOI: 10.15430/jcp.2013.18.2.123] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 06/18/2013] [Accepted: 06/20/2013] [Indexed: 01/29/2023] Open
Abstract
Cyclooxygenase (COX) is a key enzyme responsible for inflammation, converting arachidonic acid to prostaglandin and thromboxane. COX has at least two isoforms, COX-1 and COX-2. While COX-1 is constitutively expressed in most tissues for maintaining physiologic homeostasis, COX-2 is induced by inflammatory stimuli including cytokines and growth factors. Many studies have shown that COX-2 contributes to cancer development and progression in various types of malignancy including cervical cancer. Human papillomavirus, a necessary cause of cervical cancer, induces COX-2 expression via E5, E6 and E7 oncoproteins, which leads to prostaglandin E2 increase and the loss of E-cadherin, promotes cell proliferation and production of vascular endothelial growth factor. It is strongly suggested that COX-2 is associated with cancer development and progression such as lymph node metastasis. Many studies have suggested that non-selective COX-2 inhibitors such as non-steroidal anti-inflammatory drugs (NSAIDs), and selective COX-2 inhibitors might show anti-cancer activity in COX-2 -dependent and -independent manners. Two phase II trials for patients with locally advanced cervical cancer showed that celecoxib increased toxicities associated with radiotherapy. Contrary to these discouraging results, two phase II clinical trials, using rofecoxib and celecoxib, demonstrated the promising chemopreventive effect for patients with cervical intraepithelial neoplasia 2 or 3. However, these agents cause a rare, but serious, cardiovascular complication in spite of gastrointestinal protection in comparison with NSAIDs. Recent pharmacogenomic studies have showed that the new strategy for overcoming the limitation in clinical application of COX-2 inhibitors shed light on the use of them as a chemopreventive method.
Collapse
Affiliation(s)
- Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul
| | - Taehun Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul
| | - Mi-Kyung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul
| | - Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam
| | - Hyun Hoon Chung
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul ; Cancer Research Institute, Seoul National University College of Medicine ; Major in Biomodulation, World Class University, Seoul National University, Seoul, Korea
| |
Collapse
|
26
|
Arora RK, Kaur N, Bansal Y, Bansal G. Novel coumarin-benzimidazole derivatives as antioxidants and safer anti-inflammatory agents. Acta Pharm Sin B 2014; 4:368-75. [PMID: 26579406 PMCID: PMC4629095 DOI: 10.1016/j.apsb.2014.07.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/26/2014] [Accepted: 07/02/2014] [Indexed: 12/19/2022] Open
Abstract
Inspired from occurrence of anti-inflammatory activity of 3-substituted coumarins and antiulcer activity of various 2-substituted benzimidazoles, novel compounds have been designed by coupling coumarin derivatives at 3-position directly or through amide linkage with benzimidazole nucleus at 2-position. The resultant compounds are expected to exhibit both anti-inflammatory and antioxidant activities along with less gastric toxicity profile. Two series of coumarin-benzimidazole derivatives (4a-e and 5a-e) were synthesized and evaluated for anti-inflammatory activity and antioxidant activity. Compounds 4c, 4d and 5a displayed good anti-inflammatory (45.45%, 46.75% and 42.85% inhibition, respectively, versus 54.54% inhibition by indomethacin) and antioxidant (IC50 of 19.7, 13.9 and 1.2 µmol/L, respectively, versus 23.4 µmol/L for butylatedhydroxytoluene) activities. Evaluation of ulcer index and in vivo biochemical estimations for oxidative stress revealed that compounds 4d and 5a remain safe on gastric mucosa and did not induce oxidative stress in tissues. Calculation of various molecular properties suggests the compounds to be sufficiently bioavailable.
Collapse
Affiliation(s)
| | | | - Yogita Bansal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
| | | |
Collapse
|
27
|
Zamani Taghizadeh Rabe S, Mousavi SH, Tabasi N, Rastin M, Zamani Taghizadeh Rabe S, Siadat Z, Mahmoudi M. Rose Bengal suppresses gastric cancer cell proliferation via apoptosis and inhibits nitric oxide formation in macrophages. J Immunotoxicol 2014; 11:367-75. [PMID: 24575814 DOI: 10.3109/1547691x.2013.853715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Rose Bengal (RB) has been used as a safe agent in clinical diagnosis. In addition, it is used as a photodynamic sensitizer for removing microorganisms and cancer cells. Recently, its preferential toxicity after direct exposure to cancer cells was proven. The present study focuses on anti-cancer and anti-inflammatory activities of RB. The toxicity of RB against AGS gastric cancer and NIH 3T3 fibroblast cell lines was studied using an MTT assay. Patterns of any cell death among the AGS cells were defined using Annexin-V and PI staining. In addition, the effect of RB on nitric oxide (NO) and prostaglandin E(2) (PGE(2)) production induced by lipopolysaccha-ride in J774A.1 macrophages was determined. Modulation of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 expressions in the macrophages was also evaluated by Western blots. The results showed that AGS cells exhibited significant concentration-dependent decreases in growth in response to RB; these cells showed a greater growth inhibition than did non-malignant 3T3 cells, suggesting that anti-growth activity of RB could be cell-specific. Moreover, AGS cells exposed to RB exhibited a significant increase in apoptosis; only at high RB doses did the cells display significant levels of necrosis. While RB also caused a modest decrease in the growth of J774A.1 macrophages, the cells displayed remarkable decreases in NO production and iNOS expression without significant concurrent modulation in PGE(2) production or COX-2 expression. The data from this study appears to suggest that RB differentially impacts on transformed cell lines, preferentially suppresses growth of a gastric cancer cell line through induction of apoptosis, and induces changes in cells that could reflect potential anti-inflammatory effects that might be induced in situ.
Collapse
|
28
|
Martelli A, Testai L, Anzini M, Cappelli A, Di Capua A, Biava M, Poce G, Consalvi S, Giordani A, Caselli G, Rovati L, Ghelardini C, Patrignani P, Sautebin L, Breschi M, Calderone V. The novel anti-inflammatory agent VA694, endowed with both NO-releasing and COX2-selective inhibiting properties, exhibits NO-mediated positive effects on blood pressure, coronary flow and endothelium in an experimental model of hypertension and endothelial dysfunction. Pharmacol Res 2013; 78:1-9. [DOI: 10.1016/j.phrs.2013.09.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/18/2013] [Accepted: 09/20/2013] [Indexed: 11/29/2022]
|
29
|
Satoh H, Amagase K, Takeuchi K. Mucosal protective agents prevent exacerbation of NSAID-induced small intestinal lesions caused by antisecretory drugs in rats. J Pharmacol Exp Ther 2013; 348:227-35. [PMID: 24254524 DOI: 10.1124/jpet.113.208991] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Antisecretory drugs such as histamine H₂-receptor antagonists and proton pump inhibitors are commonly used for the treatment of upper gastrointestinal mucosal lesions induced by nonsteroidal anti-inflammatory drugs (NSAIDs). However, it has recently been reported that these drugs exacerbate NSAID-induced small intestinal lesions in rats. Unfortunately, there are few effective agents for the treatment of this complication. We examined the effects of mucosal protective agents (MPAs) (misoprostol, irsogladine, and rebamipide) and mucin of porcine stomach on diclofenac-induced intestinal lesions and the exacerbation of the lesions by ranitidine or omeprazole. The effects of the drugs on intestinal motility and mucus distribution/content were also examined. Male Wistar rats (180-220 g) were used. Each drug was administered orally under fed conditions. Diclofenac (1-10 mg/kg) produced multiple lesions in the small intestine dose-dependently. Both ranitidine (30 mg/kg) and omeprazole (100 mg/kg) significantly increased the intestinal lesions induced by low doses (3 and 6 mg/kg) of diclofenac. Misoprostol (0.03-0.3 mg/kg), irsogladine (3-30 mg/kg), and rebamipide (30-300 mg/kg), as well as mucin (30-300 mg/kg) inhibited the formation of intestinal lesions caused by a high dose (10 mg/kg) of diclofenac alone and prevented the exacerbation of diclofenac-induced lesions by antisecretory drugs. Diclofenac (10 mg/kg) markedly increased the intestinal motility and decreased the mucosal mucus, and the decrease of mucus was significantly inhibited by the MPAs. These results indicate the usefulness of the MPAs for the treatment of intestinal lesions induced by NSAIDs alone or by coadministration with antisecretory drugs, and suggest that mucus plays an important role in the protection of intestinal mucosa by the MPAs.
Collapse
Affiliation(s)
- Hiroshi Satoh
- Department of Pharmacology & Experimental Therapeutics, Division of Pathological Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | | | | |
Collapse
|
30
|
Cheng J, Du YF, Xiao ZY, Pan LL, Li W, Huan L, Gong ZN, Wei SH, Huang SQ, Xun W, Zhang Y, Chang LL, Xie MY, Ao GZ, Cai J, Qiu T, Wu H, Sun T, Xu GL. Growth inhibitory effect of KYKZL-1 on Hep G2 cells via inhibition of AA metabolites and caspase-3 pathway and cell cycle arrest. Toxicol Appl Pharmacol 2013; 274:96-106. [PMID: 24189224 DOI: 10.1016/j.taap.2013.10.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/19/2013] [Accepted: 10/22/2013] [Indexed: 10/26/2022]
Abstract
KYKZL-1, a newly synthesized compound with COX/5-LOX dual inhibition, was subjected to the inhibitory activity test on Hep G2 growth. We found that KYKZL-1 inhibited the growth of Hep G2 cells via inducing apoptosis. Further studies showed that KYKZL-1 activated caspase-3 through cytochrome c release from mitochondria and down regulation of Bcl-2/Bax ratio and reduced the high level of COX-2 and 5-LOX. As shown in its anti-inflammatory effect, KYKZL-1 also exhibited inhibitory effect on the PGE2 and LTB4 production in Hep G2 cells. Accordingly, exogenous addition of PGE2 or LTB4 reversed the decreases in cell viability. In addition, KYKZL-1 caused cell cycle arrest at the S-G2 checkpoint via the activation of p21(CIP1) protein and down-regulation of cyclin A expression. These data indicate that the growth inhibitory effect of KYKZL-1 is associated with inhibition of AA metabolites and caspase-3 pathway and cell cycle arrest. Combined with our previous findings, KYKZL-1 exhibiting COX/5-LOX inhibition may be a promising potential agent not only for inflammation control but also for cancer prevention/therapy with an enhanced gastric safety profile.
Collapse
Affiliation(s)
- Jing Cheng
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Yi-Fang Du
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Zhi-Yi Xiao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Li-Li Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Wei Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lin Huan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Zhu-Nan Gong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Shao-Hua Wei
- College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, China
| | - Shi-Qian Huang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Wei Xun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Yi Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lei-Lei Chang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Meng-Yu Xie
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Gui-Zhen Ao
- Department of Medicinal Chemistry, School of Pharmacy, Soochow University, Jiangsu, China
| | - Jie Cai
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ting Qiu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Hao Wu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ting Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Guang-Lin Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China; Department of Pharmacology, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
31
|
Inhibition of inflammatory mediators contributes to the anti-inflammatory activity of KYKZL-1 via MAPK and NF-κB pathway. Toxicol Appl Pharmacol 2013; 272:221-9. [DOI: 10.1016/j.taap.2013.05.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 04/11/2013] [Accepted: 05/10/2013] [Indexed: 11/19/2022]
|
32
|
The role of food for the formation and prevention of gastrointestinal lesions induced by aspirin in cats. Dig Dis Sci 2013; 58:2840-9. [PMID: 23765256 DOI: 10.1007/s10620-013-2725-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 05/15/2013] [Indexed: 01/19/2023]
Abstract
BACKGROUND/AIMS The effects of feeding conditions (fasted or fed) and dietary fiber (DF) in the diet on gastrointestinal (GI) damage induced by aspirin (ASA) were examined in cats. METHODS Plain ASA (P-ASA, 20 mg/kg) or one enteric-coated ASA tablet (EC-ASA, containing 100 mg ASA) was administered p.o. once daily for 3 or 7 days just after morning meal, 3 h after the evening meal, or in the morning without a morning meal (fasted). Several types of diet, dry food (DRY, DF: 2.8 %), canned food (CAN, DF: 0.4 %), and diets with added cellulose or pectin were provided twice daily. RESULTS P-ASA or EC-ASA administered just after feeding of DRY caused marked lesions in the GI tract, although EC-ASA did not produce any lesions in the stomach. GI damage was markedly decreased when ASA was administered 3 h after the evening meal. The induction of lesions by EC-ASA was markedly decreased in cats that ate CAN, but lesions were induced in cats fed CAN with added cellulose (6 %). The addition of pectin (6 %) to the DRY markedly decreased the induction of lesions by EC-ASA. CONCLUSIONS The results indicate that the induction of GI lesions by ASA was highly dependent on the feeding conditions and DF. To minimize the induction of GI damage, it would be better to take ASA 3 h after the evening meal, or after consuming diets that contain low amounts of insoluble DF and high amounts of soluble DF.
Collapse
|
33
|
Recent Advances in NSAIDs-Induced Enteropathy Therapeutics: New Options, New Challenges. Gastroenterol Res Pract 2013; 2013:761060. [PMID: 24159330 PMCID: PMC3789478 DOI: 10.1155/2013/761060] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/02/2013] [Accepted: 08/13/2013] [Indexed: 12/17/2022] Open
Abstract
The injurious effects of NSAIDs on the small intestine were not fully appreciated until the widespread use of capsule endoscopy. It is estimated that over two-thirds of regular NSAID users develop injury in the small intestinal injuries and that these injuries are more common than gastroduodenal mucosal injuries. Recently, chronic low-dose aspirin consumption was found to be associated with injury to the lower gut and to be a significant contributing factor in small bowel ulceration, hemorrhage, and strictures. The ability of aspirin and NSAIDs to inhibit the activities of cyclooxygenase (COX) contributes to the cytotoxicity of these drugs in the gastrointestinal tract. However, many studies found that, in the small intestine, COX-independent mechanisms are the main contributors to NSAID cytotoxicity. Bile and Gram-negative bacteria are important factors in the pathogenesis of NSAID enteropathy. Here, we focus on a promising strategy to prevent NSAID-induced small intestine injury. Selective COX-2 inhibitors, prostaglandin derivatives, mucoprotective drugs, phosphatidylcholine-NSAIDs, and probiotics have potential protective effects on NSAID enteropathy.
Collapse
|
34
|
Wallace JL. Mechanisms, prevention and clinical implications of nonsteroidal anti-inflammatory drug-enteropathy. World J Gastroenterol 2013; 19:1861-1876. [PMID: 23569332 PMCID: PMC3613102 DOI: 10.3748/wjg.v19.i12.1861] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 02/23/2013] [Accepted: 03/08/2013] [Indexed: 02/06/2023] Open
Abstract
This article reviews the latest developments in understanding the pathogenesis, detection and treatment of small intestinal damage and bleeding caused by nonsteroidal anti-inflammatory drugs (NSAIDs). With improvements in the detection of NSAID-induced damage in the small intestine, it is now clear that this injury and the associated bleeding occurs more frequently than that occurring in the stomach and duodenum, and can also be regarded as more dangerous. However, there are no proven-effective therapies for NSAID-enteropathy, and detection remains a challenge, particularly because of the poor correlation between tissue injury and symptoms. Moreover, recent studies suggest that commonly used drugs for protecting the upper gastrointestinal tract (i.e., proton pump inhibitors) can significantly worsen NSAID-induced damage in the small intestine. The pathogenesis of NSAID-enteropathy is complex, but studies in animal models are shedding light on the key factors that contribute to ulceration and bleeding, and are providing clues to the development of effective therapies and prevention strategies. Novel NSAIDs that do not cause small intestinal damage in animal models offer hope for a solution to this serious adverse effect of one of the most widely used classes of drugs.
Collapse
|
35
|
Bruce King S. Potential biological chemistry of hydrogen sulfide (H2S) with the nitrogen oxides. Free Radic Biol Med 2013; 55:1-7. [PMID: 23165065 PMCID: PMC3798156 DOI: 10.1016/j.freeradbiomed.2012.11.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 10/24/2012] [Accepted: 11/08/2012] [Indexed: 12/14/2022]
Abstract
Hydrogen sulfide, an important gaseous signaling agent generated in numerous biological tissues, influences many physiological processes. This biological profile seems reminiscent of nitric oxide, another important endogenously synthesized gaseous signaling molecule. Hydrogen sulfide reacts with nitric oxide or oxidized forms of nitric oxide and nitric oxide donors in vitro to form species that display distinct biology compared to both hydrogen sulfide and NO. The products of these interesting reactions may include small-molecule S-nitrosothiols or nitroxyl, the one-electron-reduced form of nitric oxide. In addition, thionitrous acid or thionitrite, compounds structurally analogous to nitrous acid and nitrite, may constitute a portion of the reaction products. Both the chemistry and the biology of thionitrous acid and thionitrite, compared to nitric oxide or hydrogen sulfide, remain poorly defined. General mechanisms for the formation of S-nitrosothiols, nitroxyl, and thionitrous acid based upon the ability of hydrogen sulfide to act as a nucleophile and a reducing agent with reactive nitric oxide-based intermediates are proposed. Hydrogen sulfide reactivity seems extensive and could have an impact on numerous areas of redox-controlled biology and chemistry, warranting more work in this exciting and developing area.
Collapse
Affiliation(s)
- S Bruce King
- Department of Chemistry, Wake Forest University, Winston-Salem, NC 27109, USA.
| |
Collapse
|
36
|
Boggara MB, Mihailescu M, Krishnamoorti R. Structural Association of Nonsteroidal Anti-Inflammatory Drugs with Lipid Membranes. J Am Chem Soc 2012; 134:19669-76. [DOI: 10.1021/ja3064342] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Mohan Babu Boggara
- Department of Chemical and Biomolecular
Engineering, University of Houston, Houston,
Texas 77204, United States
| | - Mihaela Mihailescu
- Institute for Bioscience and
Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States
- National Institute for Standard and Technology, Center for Neutron Research,
Gaithersburg, Maryland 20899, United States
| | - Ramanan Krishnamoorti
- Department of Chemical and Biomolecular
Engineering, University of Houston, Houston,
Texas 77204, United States
| |
Collapse
|
37
|
Synthesis and investigation of anti-inflammatory activity of novel nitric oxide donating hybrid drugs. Med Chem Res 2012. [DOI: 10.1007/s00044-012-0345-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
38
|
Lim YJ, Yang CH. Non-steroidal anti-inflammatory drug-induced enteropathy. Clin Endosc 2012; 45:138-44. [PMID: 22866254 PMCID: PMC3401617 DOI: 10.5946/ce.2012.45.2.138] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/18/2012] [Accepted: 04/19/2012] [Indexed: 12/13/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are one of the most commonly prescribed drugs in the world. NSAID-induced lower gastrointestinal (GI) complications are increasing while upper GI complications are decreasing. Lower GI events accounted for 40% of all serious GI events in patients on NSAIDs. Capsule endoscopy and device assisted enteroscopy are available for detection of small intestinal lesions. Capsule endoscopy studies have demonstrated that NSAIDs use in healthy volunteers raised the incidence (55% to 75%) of intestinal damage. It appears that selective cyclooxygenase-2 inhibitors (coxibs) improved upper and lower GI safety based on results of clinical trials. Selective coxibs are still capable of triggering GI adverse events and cardiovascular toxicity issues were the main focus of concerns. Unfortunately, definite strategies are not available to prevent or heal NSAID-induced intestinal injuries. Thus, there is still a strong clinical need for effective drugs with improved safety profiles than the existing NSAIDs.
Collapse
Affiliation(s)
- Yun Jeong Lim
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University Graduate School of Medicine, Seoul, Korea
| | | |
Collapse
|
39
|
Taguchi K, Chuang VTG, Maruyama T, Otagiri M. Pharmaceutical aspects of the recombinant human serum albumin dimer: structural characteristics, biological properties, and medical applications. J Pharm Sci 2012; 101:3033-46. [PMID: 22573538 DOI: 10.1002/jps.23181] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/12/2012] [Accepted: 04/18/2012] [Indexed: 12/15/2022]
Abstract
Human serum albumin is the most abundant protein in the blood. It is clinically used in the treatment of severe hypoalbuminemia and as a plasma expander. The use of albumins as a carrier for drugs is currently being developed, and some are now in the preclinical and clinical trial stages. The main technologies for utilizing an albumin as a drug carrier are protein fusion, polymerization and surface modification, and so on. Among these technologies, albumin dimerization has wide clinical applications as a plasma expander as well as a drug carrier. Despite the fact that many reports have appeared on drugs using an albumin dimer as a carrier, our knowledge of the characteristics of the albumin dimer itself is incomplete. In this review, we summarize the structural characteristics of recombinant albumin dimers produced by two methods, namely, chemical linkage with 1,6-bis(maleimido)hexane and genetically linked with an amino acid linker, and the physicochemical characteristics and biological properties of these preparations. Finally, the potential for pharmaceutical applications of albumin dimers in clinical situations is discussed.
Collapse
Affiliation(s)
- Kazuaki Taguchi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | | | | | | |
Collapse
|
40
|
Chattopadhyay M, Kodela R, Olson KR, Kashfi K. NOSH-aspirin (NBS-1120), a novel nitric oxide- and hydrogen sulfide-releasing hybrid is a potent inhibitor of colon cancer cell growth in vitro and in a xenograft mouse model. Biochem Biophys Res Commun 2012; 419:523-8. [PMID: 22366248 DOI: 10.1016/j.bbrc.2012.02.051] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 02/08/2012] [Indexed: 02/06/2023]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are prototypical anti-cancer agents. However, their long-term use is associated with adverse gastrointestinal effects. Recognition that endogenous gaseous mediators, nitric oxide (NO) and hydrogen sulfide (H(2)S) can increase mucosal defense mechanisms has led to the development of NO- and H(2)S-releasing NSAIDs with increased safety profiles. Here we report on a new hybrid, NOSH-aspirin, which is an NO- and H(2)S-releasing agent. NOSH-aspirin inhibited HT-29 colon cancer growth with IC(50)s of 45.5 ± 2.5, 19.7 ± 3.3, and 7.7 ± 2.2 nM at 24, 48, and 72 h, respectively. This is the first NSAID based agent with such high degree of potency. NOSH-aspirin inhibited cell proliferation, induced apoptosis, and caused G(0)/G(1) cell cycle block. Reconstitution and structure-activity studies representing a fairly close approximation to the intact molecule showed that NOSH-aspirin was 9000-fold more potent than the sum of its parts towards growth inhibition. NOSH-aspirin inhibited ovine COX-1 more than ovine COX-2. NOSH-ASA treatment of mice bearing a human colon cancer xenograft caused a reduction in volume of 85%. Taken together, these results demonstrate that NOSH-aspirin has strong anti-cancer potential and merits further evaluation.
Collapse
Affiliation(s)
- Mitali Chattopadhyay
- Department of Physiology, Pharmacology, and Neuroscience, Sophie Davis School of Biomedical Education, City University of New York Medical School, NY 10031, USA
| | | | | | | |
Collapse
|
41
|
Ishima Y, Chen D, Fang J, Maeda H, Minomo A, Kragh-Hansen U, Kai T, Maruyama T, Otagiri M. S-Nitrosated Human Serum Albumin Dimer is not only a Novel Anti-Tumor Drug but also a Potentiator for Anti-Tumor Drugs with Augmented EPR Effects. Bioconjug Chem 2012; 23:264-71. [DOI: 10.1021/bc2005363] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | | | | | - Ulrich Kragh-Hansen
- Department
of Medical Biochemistry, University of Aarhus, DK-8000 Aarhus C, Denmark
| | - Toshiya Kai
- Tohoku Nipro Pharmaceutical Corporation, 428 Okanouchi, Kagamiishimachi, Iwasegun,
Fukushima 969-0401, Japan
| | | | | |
Collapse
|
42
|
Blackler R, Syer S, Bolla M, Ongini E, Wallace JL. Gastrointestinal-sparing effects of novel NSAIDs in rats with compromised mucosal defence. PLoS One 2012; 7:e35196. [PMID: 22496907 PMCID: PMC3322164 DOI: 10.1371/journal.pone.0035196] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 03/10/2012] [Indexed: 01/10/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs are among the most commonly used prescription and over-the-counter medications, but they often produce significant gastrointestinal ulceration and bleeding, particularly in elderly patients and patients with certain co-morbidities. Novel anti-inflammatory drugs are seldom tested in animal models that mimic the high risk human users, leading to an underestimate of the true toxicity of the drugs. In the present study we examined the effects of two novel NSAIDs and two commonly used NSAIDs in models in which mucosal defence was expected to be impaired. Naproxen, celecoxib, ATB-346 (a hydrogen sulfide- and naproxen-releasing compound) and NCX 429 (a nitric oxide- and naproxen-releasing compound) were evaluated in healthy, arthritic, obese, and hypertensive rats and in rats of advanced age (19 months) and rats co-administered low-dose aspirin and/or omeprazole. In all models except hypertension, greater gastric and/or intestinal damage was observed when naproxen was administered in these models than in healthy rats. Celecoxib-induced damage was significantly increased when co-administered with low-dose aspirin and/or omeprazole. In contrast, ATB-346 and NCX 429, when tested at doses that were as effective as naproxen and celecoxib in reducing inflammation and inhibiting cyclooxygenase activity, did not produce significant gastric or intestinal damage in any of the models. These results demonstrate that animal models of human co-morbidities display the same increased susceptibility to NSAID-induced gastrointestinal damage as observed in humans. Moreover, two novel NSAIDs that release mediators of mucosal defence (hydrogen sulfide and nitric oxide) do not induce significant gastrointestinal damage in these models of impaired mucosal defence.
Collapse
Affiliation(s)
- Rory Blackler
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Stephanie Syer
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | | | | | - John L. Wallace
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
43
|
Abstract
Nitric oxide (NO)-releasing agents such as JS-K and NO-releasing hybrids such as NO- and NONO-nonsteroidal anti-inflammatory drugs are novel agents with great potential for controlling cancer. Although studied extensively, a key question pertaining to their molecular targets and mechanism of action remains unclear: the role of NO in the overall biological effect of these agents. It has been shown that NO can directly modify sulfhydryl residues of proteins through S-nitrosylation and induce apoptosis. We showed that 3 structurally diverse NO-nonsteroidal anti-inflammatory drugs S-nitrosylated nuclear factor-κB p65 in vitro and in vivo and also showed that these agents S-nitrosylated caspase-3 in vivo. JS-K reduced nuclear β-catenin and cyclin D1 protein levels without affecting cytosolic β-catenin expression. On the basis of a time course study, S-nitrsolyation of nuclear β-catenin was determined to precede its degradation. These data provide a mechanistic role for NO and a rationale for the chemopreventive effects of these novel agents.
Collapse
Affiliation(s)
- Khosrow Kashfi
- Department of Physiology, Pharmacology and Neuroscience, City University of New York Medical School, 138th Street and Convent Avenue, New York, NY 10031; Tel.: (212) 650-6641; -7692
| |
Collapse
|
44
|
Abdul-Hay S, Schiefer IT, Chandrasena REP, Li M, Abdelhamid R, Wang YT, Tavassoli E, Michalsen B, Asghodom RT, Luo J, Thatcher GRJ. NO-SSRIs: Nitric Oxide Chimera Drugs Incorporating a Selective Serotonin Reuptake Inhibitor. ACS Med Chem Lett 2011; 2:656-661. [PMID: 21927645 DOI: 10.1021/ml2000033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hybrid nitrate drugs have been reported to provide NO bioactivity to ameliorate side effects or to provide ancillary therapeutic activity. Hybrid nitrate selective serotonin reuptake inhibitors (NO-SSRIs) were prepared to improve the therapeutic profile of this drug class. A synthetic strategy for use of a thiocarbamate linker was developed, which in the case of NO-fluoxetine facilitated hydrolysis to fluoxetine at pH 7.4 within 7 hours. In cell culture, NO-SSRIs were weak inhibitors of the serotonin transporter, however, in the forced swimming task (FST) in rats, NO-fluoxetine demonstrated classical antidepressant activity. Comparison of NO-fluoxetine, with fluoxetine, and an NO-chimera nitrate developed for Alzheimer's disease (GT-1061), was made in the step through passive avoidance (STPA) test of learning and memory in rats treated with scopolamine as an amnesic agent. Fluoxetine was inactive, whereas NO-fluoxetine and GT-1061 both restored long-term memory. GT-1061 also produced antidepressant behavior in FST. These data support the potential for NO-SSRIs to overcome the lag in onset of therapeutic action and provide co-therapy of neuropathologies concomitant with depression.
Collapse
Affiliation(s)
- Samer Abdul-Hay
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Isaac T. Schiefer
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - R. Esala P. Chandrasena
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Min Li
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Ramy Abdelhamid
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Yue-Ting Wang
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Ehsan Tavassoli
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Bradley Michalsen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Rezene T. Asghodom
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Jia Luo
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Gregory R. J. Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| |
Collapse
|
45
|
Abstract
INTRODUCTION It is becoming increasingly clear that many diseases are characterized or associated with perturbations in nitric oxide (NO) production/signaling. Therapeutics or strategies designed to restore normal NO homeostasis will likely have broad application and utility in human health. This highly complex and multi-step pathway for NO production and subsequent target activation provides many steps in the endogenous pathway that may be useful targets for drug development. Important therapeutic areas for NO-based therapies are inflammatory disorders, cardiovascular diseases, erectile dysfunction and metabolic disorders. AREAS COVERED The following review will discuss the endogenous NO pathway, highlight the current market and indications for NO-based therapeutics, as well as identify pathway targets currently under drug development. Each step along the NO pathway will be discussed including exogenous sources of NO, use of precursors to promote NO production and downstream pathways affected by NO production with advantages and disadvantages highlighted for each. EXPERT OPINION Development of NO-based therapeutics is and will continue to be a major focus of biotech and pharmaceutical companies. Understanding and utilizing dietary and nutritional strategies to restore NO homeostasis could allow for safer, quicker marketing of products that may be just as efficacious as drugs designed against specific targets.
Collapse
Affiliation(s)
- Nathan S Bryan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine , The University of Texas Health Science Center at Houston,The Graduate School of Biomedical Sciences at Houston , Department of Integrative Biology and Pharmacology , 1825 Pressler St. 530C, Houston, TX 77030 , USA +1 713 500 2439 ; +1 713 500 2447 ;
| |
Collapse
|
46
|
Huang Z, Velázquez CA, Abdellatif KRA, Chowdhury MA, Reisz JA, DuMond JF, King SB, Knaus EE. Ethanesulfohydroxamic acid ester prodrugs of nonsteroidal anti-inflammatory drugs (NSAIDs): synthesis, nitric oxide and nitroxyl release, cyclooxygenase inhibition, anti-inflammatory, and ulcerogenicity index studies. J Med Chem 2011; 54:1356-64. [PMID: 21280601 DOI: 10.1021/jm101403g] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The carboxylic acid group of the anti-inflammatory (AI) drugs indo-methacin, (S)-naproxen and ibuprofen was covalently linked via a two-carbon ethyl spacer to a sulfohydroxamic acid moiety (CH(2)CH(2)SO(2)NHOH) to furnish a group of hybrid ester prodrugs that release nitric oxide (NO) and nitroxyl (HNO). Biological data acquired for this hitherto unknown class of ethanesulfohydroxamic acid ester prodrugs showed (i) all compounds exhibited superior NO, but similar HNO, release properties relative to arylsulfohydroxamic acids, (ii) the (S)-naproxen and ibuprofen prodrug esters are more potent AI agents than their parent NSAID, (iii) the indomethacin prodrug ester, in contrast to indomethacin which is highly ulcerogenic, showed no visible stomach lesions [ulcer index (UI) = 0 for a 80 μmol/kg oral dose] while retaining potent AI activity, and iv) that the indomethacin prodrug ester, unlike indomethacin which is an ulcerogenic selective COX-1 inhibitor, is a selective COX-2 inhibitor (COX-2 selectivity index = 184) devoid of ulcerogenicity that is attributed to its high COX-2 SI and/or ability to release cytoprotective NO.
Collapse
Affiliation(s)
- Zhangjian Huang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton Alberta T6G 2N8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Wallace JL, Ferraz JGP. New pharmacologic therapies in gastrointestinal disease. Gastroenterol Clin North Am 2010; 39:709-20. [PMID: 20951926 DOI: 10.1016/j.gtc.2010.08.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Many gastrointestinal diseases remain poorly responsive to therapies, and even in the cases of conditions for which there are many effective drugs, there is still considerable room for improvement. This article is focused on drugs for digestive disorders that have entered the marketplace recently, or are expected to reach the marketplace within the next 1 to 2 years. Although advances have been made in understanding gastrointestinal motility, visceral pain, mucosal inflammation, and tissue repair, the major gastrointestinal diseases remain as significant therapeutic challenges.
Collapse
Affiliation(s)
- John L Wallace
- Farncombe Family Digestive Health Research Institute, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | | |
Collapse
|
48
|
Haeggström JZ, Rinaldo-Matthis A, Wheelock CE, Wetterholm A. Advances in eicosanoid research, novel therapeutic implications. Biochem Biophys Res Commun 2010; 396:135-9. [DOI: 10.1016/j.bbrc.2010.03.140] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 03/23/2010] [Indexed: 10/19/2022]
|
49
|
Wallace JL. Physiological and pathophysiological roles of hydrogen sulfide in the gastrointestinal tract. Antioxid Redox Signal 2010; 12:1125-33. [PMID: 19769457 DOI: 10.1089/ars.2009.2900] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Like nitric oxide and carbon monoxide, hydrogen sulfide has historically been recognized as an industrial pollutant, but in recent years, it has been shown to be an important mediator of many physiological processes. Hydrogen sulfide contributes to the maintenance of gastrointestinal mucosal defense and repair. It also exerts many antiinflammatory effects, including inhibition of leukocyte adherence to the vascular endothelium and leukocyte migration to sites of inflammation. Conversely, inhibition of endogenous hydrogen sulfide synthesis leads to a loss of mucosal integrity and to an increase in mucosal inflammation. Hydrogen sulfide therefore appears to have overlapping actions with nitric oxide and prostaglandins in terms of modulating mucosal defense and resolution of inflammation. Recent evidence suggests that these properties of hydrogen sulfide can be exploited in the design of novel therapies for ulcerative and inflammatory diseases of the gastrointestinal tract.
Collapse
Affiliation(s)
- John L Wallace
- Farncombe Family Institute for Digestive Health Research, Department of Medicine, McMaster University, 1200 Main Street West, Hamilton, Ontario, Canada.
| |
Collapse
|
50
|
Boggara MB, Krishnamoorti R. Small-angle neutron scattering studies of phospholipid-NSAID adducts. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2010; 26:5734-5745. [PMID: 20014785 DOI: 10.1021/la903854s] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are known to have strong interactions with lipid membranes. Using small-angle neutron scattering, the effect of ibuprofen, a prominent NSAID, on the radius of small unilamellar vesicles of 1,2-dimyristoyl-sn-glycero-3-phosphatidylcholine (DMPC) and their bilayer structure was studied systematically as a function of pH (ranging from 2 to 8) and drug-to-lipid mole ratio (from 0/1 to 0.62/1 mol/mol). Ibuprofen with a pK(a) of approximately 4.6 was found to significantly affect the bilayer structure at all pH values, irrespective of the charge state of the drug. At low pH values, the drug reduces the bilayer thickness, induces fluid-like behavior, and changes headgroup hydration. The incorporation of the drug in the lipid bilayer while affecting the local bilayer structure and hydration of the lipid does not affect the overall stability of the vesicle dispersions over the pH range studied.
Collapse
Affiliation(s)
- Mohan Babu Boggara
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, USA
| | | |
Collapse
|