1
|
Bajgai J, Lee M, Jang YG, Lee K, Sharma S, Jeong YJ, Park HJ, Goh SH, Kim CS, Kim HI, Lee KJ. Effects of Drinking Electrolyzed Alkaline-Reduced Water on Functional Dyspepsia: A Randomized, Double-Blind, Controlled Prospective Trial. Processes (Basel) 2023; 11:968. [DOI: 10.3390/pr11030968] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
A well-known functional gastrointestinal disorder called functional dyspepsia (FD) is defined by dyspeptic symptoms without any structural abnormalities. In alternative intervention, electrolyzed alkaline-reduced water (EARW) consumption is regarded as a treatment modality for gastrointestinal symptoms despite its mechanism not yet fully understood. The present clinical study aimed to investigate the effects of EARW on gastrointestinal symptoms of patients with FD. Forty-eight participants with FD were screened, and 42 were enrolled. Participants were randomly allocated to the EARW (n = 21) and purified water (PW) (n = 21) groups. The EARW group ingested EARW (10 mL/kg body weight/day) for 6 weeks. The gastrointestinal symptom rating scale (GSRS), functional dyspepsia-related quality of life (FD-QoL), the Korean version of the Nepean Dyspepsia Index (NDI-K) were used as primary outcome measures at baseline and at 6 weeks, and inflammatory markers were measured as the secondary outcome. Two participants dropped out, and 40 participants (EARW = 20 and PW = 20) completed the trial. Total GSRS score was significantly lower in the EARW group (34.27%, p < 0.01) than in the PW (18.16%) group. In the five subcategories of GSRS, the decreased score between baseline and post-intervention for the EARW and PW groups were 43.59% and 21.33% in abdominal pain score, respectively; 38.98% and 18.92% in reflux syndrome, respectively; 25.42% and 20.90% in diarrhea, respectively; 35.87% and 21.48% in indigestion, respectively; and 32.81% and 10.71% in constipation, respectively, and all the parameters were significantly different in the EARW group compared with those in the PW group. The NDI-K score was also lower in the EARW group (p < 0.01) than in the PW group. FD-QoL score decreased significantly more in the EARW group after intervention than in the PW group (p < 0.05). Additionally, inflammatory cytokines (TNF-α and IFN-γ) levels significantly suppressed in the EARW group after 6 weeks of drinking compared with the levels at the baseline. Our clinical study suggests that long-term drinking of EARW (pH 9.5) may improve FD-related symptoms and the quality of life of FD patients through home-based administration.
Collapse
Affiliation(s)
- Johny Bajgai
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Mihyun Lee
- Clinical Research Institute, Ceragem Clinical Inc. Asia Media Center Bldg, 27 Eonju-ro 93-gil, Gangnam-gu, Seoul 06142, Republic of Korea
| | - Yeon-Gyu Jang
- Department of Neurosurgery, Wonju Severance Christian Hospital, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Kiwon Lee
- Clinical Research Institute, Ceragem Clinical Inc. Asia Media Center Bldg, 27 Eonju-ro 93-gil, Gangnam-gu, Seoul 06142, Republic of Korea
| | - Subham Sharma
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Yun Ju Jeong
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Hong Jun Park
- Department of Gastroenterology, Yonsei University, Wonju 26426, Republic of Korea
| | - Seong Hoon Goh
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Cheol-Su Kim
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Hyun Il Kim
- Department of Gastroenterology, Yonsei University, Wonju 26426, Republic of Korea
| | - Kyu-Jae Lee
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| |
Collapse
|
2
|
MacNicol JL, Pearson W. Gastrin and Nitric Oxide Production in Cultured Gastric Antral Mucosa Are Altered in Response to a Gastric Digest of a Dietary Supplement. Front Vet Sci 2021; 8:684203. [PMID: 34671658 PMCID: PMC8520902 DOI: 10.3389/fvets.2021.684203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/31/2021] [Indexed: 12/04/2022] Open
Abstract
In vitro organ culture can provide insight into isolated mucosal responses to particular environmental stimuli. The objective of the present study was to investigate the impact of a prolonged culturing time as well as the addition of acidic gastric fluid into the in vitro environment of cultured gastric antral tissue to evaluate how altering the commonly used neutral environment impacted tissue. Furthermore, we aimed to investigate the impact of G's Formula, a dietary supplement for horses, on the secretion of gastrin, interleukin1-beta (IL-1β), and nitric oxide (NO). These biomarkers are of interest due to their effects on gastric motility and mucosal activity. Gastric mucosal tissue explants from porcine stomachs were cultured in the presence of a simulated gastric fluid (BL, n = 14), simulated gastric fluid containing the dietary supplement G's Formula (DF, n = 12), or an equal volume of phosphate buffered saline (CO, n = 14). At 48 and 60 h, 10−5 M carbachol was used to stimulate gastrin secretion. Cell viability was assessed at 72 h using calcein and ethidium-homodimer 1 staining. Media was analyzed for gastrin, IL-1β, and NO at 48, 60, and 72 h. There were no effects of treatment or carbachol stimulation on explant cell viability. Carbachol resulted in a significant increase in gastrin concentration in CO and DF treatments, but not in BL. NO was higher in CO than in BL, and NO increased in the CO and DF treatments but not in BL. In conclusion, the addition of carbachol and gastric digests to culture media did not impact cell viability. The use of an acidic gastric digest (BL) reduced the effect of cholinergic stimulation with carbachol at a concentration of 10−5 M and reduced NO secretion. The addition of the dietary supplement to the gastric digest (DF) appeared to mediate these effects within this model. Further research is required to evaluate the specific effects of this dietary supplement on direct markers of mucosal activity and the functional relevance of these results in vivo.
Collapse
Affiliation(s)
- Jennifer L MacNicol
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
| | - Wendy Pearson
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
3
|
Goswami SK, Rand AA, Wan D, Yang J, Inceoglu B, Thomas M, Morisseau C, Yang GY, Hammock BD. Pharmacological inhibition of soluble epoxide hydrolase or genetic deletion reduces diclofenac-induced gastric ulcers. Life Sci 2017; 180:114-122. [PMID: 28522175 DOI: 10.1016/j.lfs.2017.05.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 05/08/2017] [Accepted: 05/14/2017] [Indexed: 12/13/2022]
Abstract
AIMS This research was conducted to evaluate the hypothesis that gastric ulcers caused by the NSAID diclofenac sodium (DCF) can be prevented by the soluble epoxide hydrolase inhibitor TPPU. MAIN METHODS Mice were administered a single dose of 10, 30 or 100mg/kg of DCF. Once an ulcerative dose of DCF was chosen, mice were pretreated with TPPU for 7days at 0.1mg/kg to evaluate anti-ulcer effects of the sEH inhibitor on anatomy, histopathology, pH, inflammatory markers and epithelial apoptosis of stomachs. KEY FINDINGS Diclofenac caused ulceration of the stomach at a dose of 100mg/kg and a time post dose of 6h. Ulcers generated under these conditions were associated with a significant increase in the levels of TNF-α and IL-6 in serum and increased apoptosis compared to control mice. Pretreatment with TPPU resulted in a decrease of ulceration in mice treated with DCF with a significant decrease in the level of apoptosis, TNF-α and IL-6 in the serum in comparison to diclofenac-treated mice. TPPU did not affect the pH of the stomach, whereas omeprazole elevated the pH of the stomach as expected. A similar anti-ulcer effect was observed in sEH gene knockout mice treated with DCF. SIGNIFICANCE The sEH inhibitor TPPU decreases the NSAID-induced stomach ulcers.
Collapse
Affiliation(s)
- Sumanta Kumar Goswami
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Amelia Ann Rand
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Debin Wan
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Jun Yang
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Bora Inceoglu
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Melany Thomas
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Christophe Morisseau
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Guang-Yu Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA.
| |
Collapse
|
4
|
Pereira-Leite C, Nunes C, Jamal SK, Cuccovia IM, Reis S. Nonsteroidal Anti-Inflammatory Therapy: A Journey Toward Safety. Med Res Rev 2016; 37:802-859. [PMID: 28005273 DOI: 10.1002/med.21424] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 01/01/2023]
Abstract
The efficacy of nonsteroidal anti-inflammatory drugs (NSAIDs) against inflammation, pain, and fever has been supporting their worldwide use in the treatment of painful conditions and chronic inflammatory diseases until today. However, the long-term therapy with NSAIDs was soon associated with high incidences of adverse events in the gastrointestinal tract. Therefore, the search for novel drugs with improved safety has begun with COX-2 selective inhibitors (coxibs) being straightaway developed and commercialized. Nevertheless, the excitement has fast turned to disappointment when diverse coxibs were withdrawn from the market due to cardiovascular toxicity. Such events have once again triggered the emergence of different strategies to overcome NSAIDs toxicity. Here, an integrative review is provided to address the breakthroughs of two main approaches: (i) the association of NSAIDs with protective mediators and (ii) the design of novel compounds to target downstream and/or multiple enzymes of the arachidonic acid cascade. To date, just one phosphatidylcholine-associated NSAID has already been approved for commercialization. Nevertheless, the preclinical and clinical data obtained so far indicate that both strategies may improve the safety of nonsteroidal anti-inflammatory therapy.
Collapse
Affiliation(s)
- Catarina Pereira-Leite
- UCIBIO, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Cláudia Nunes
- UCIBIO, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Sarah K Jamal
- UCIBIO, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Iolanda M Cuccovia
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Salette Reis
- UCIBIO, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
5
|
Prosperi C, Scali C, Barba M, Bellucci A, Giovannini MG, Pepeu G, Casamenti F. Comparison between Flurbiprofen and its Nitric Oxide-Releasing Derivatives HCT-1026 and NCX-2216 on Aβ(1-42)-Induced Brain Inflammation and Neuronal Damage in the Rat. Int J Immunopathol Pharmacol 2016; 17:317-30. [PMID: 15461866 DOI: 10.1177/039463200401700312] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Brain inflammation is an underlying factor in the pathogenesis of Alzheimer's disease (AD) and epidemiological studies indicate that a sustained use of non-steroidal anti-inflammatory drugs (NSAIDs) has a protective effect on the disease. We investigated, in vivo, whether differences exist in the antiinflammatory and neuroprotective actions of flurbiprofen and its two nitric oxide-donor derivatives, HCT-1026 and NCX-2216, and the ability of these two derivatives to release nitric oxide in the brain. In adult rats injected into the nucleus basalis with preaggregated Aβ(1-42) we investigated by immunohistochemical methods glia reaction, the induction of inducible nitric oxide synthase (iNOS), the activation of p38 mitogen-activated protein kinase (p38MAPK) pathway and the number of choline acetyltransferase (ChAT)-positive neurons and, in naïve rats we investigated, by microdialysis, cortical extracellular levels of nitrite. Injection of Aβ(1-42) induced iNOS at the injection site, activated p38MAPK 7 days after injection and brought about an intense microglia and astrocyte reaction along with a marked reduction in the number of ChAT-positive neurones, persisting up to at least up to 21 days. Flurbiprofen, HCT-1026 and NCX-2216 (15 mg/kg) significantly attenuated the Aβ(1-42)-induced glia reaction, iNOS induction and p38MAPK activation 7 days after treatment and astrocytes reaction 21 days after treatment. On an equimolar basis, HCT-1026 resulted the most active agent in reducing the Aβ(1-42)-induced microglia reaction. The cholinergic cell loss was also significantly reduced by 21 days of HCT-1026 treatment. No differences in the body weight were found between the animals treated for 21 days with 15 mg/kg of either HCT-1026 or NCX-2216 and the controls. An oral administration of HCT-1026 (15 mg/kg) or NCX-2216 (100 mg/kg) to naiïve rats was followed by significant and long long -lasting increases in cortical nitrite levels. These findings indicate that the addition of a nitric oxide donor potentiates the anti-inflammatory activity of flurbiprofen in a model of brain inflammation.
Collapse
Affiliation(s)
- C Prosperi
- Department of Pharmacology, University of Florence, Florence, Italy
| | | | | | | | | | | | | |
Collapse
|
6
|
Cossentini LA, Da Silva RV, Yamada-Ogatta SF, Yamauchi LM, De Almeida Araújo EJ, Pinge-Filho P. Aspirin treatment exacerbates oral infections by Trypanosoma cruzi. Exp Parasitol 2016; 164:64-70. [PMID: 26826555 DOI: 10.1016/j.exppara.2016.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 01/08/2016] [Accepted: 01/20/2016] [Indexed: 01/05/2023]
Abstract
Oral transmission of the protozoan parasite Trypanosoma cruzi, the etiological agent of Chagas disease, has been documented in Latin American countries. The reported cases of infection were due to the ingestion of contaminated fresh fruit, juices, or sugar cane juice. There have been few studies on the physiopathology of the disease in oral transmission cases. Gastritis is a common ailment that can be caused by poor dietary habits, intake of alcohol or other gastric irritants, bacterial infection, or by the widespread use of non-steroidal anti-inflammatory drugs (NSAIDs). This study investigated in a mouse model whether gastric mucosal injury, induced by aspirin, would affect the course of disease in animals infected with T. cruzi by the oral route. The CL14 and G strains of T. cruzi, both of low infectivity, were used. To this end, groups of BALB/c mice were treated during 5 days with aspirin (100 mg kg(-1)) before oral infection with T. cruzi metacyclic forms (4 × 10(5) or 5 × 10(7) parasites/mouse). Histological analysis and determination of nitric oxide and TNF-α were performed in gastric samples obtained 5 days after infection. Parasitemia was monitored from the thirteenth day after infection. The results indicate that aspirin treatment of mice injured their gastric mucosa and facilitated invasion by both CL14 and G strains of T. cruzi. Strain CL14 caused more severe infection compared to the G strain, as larger numbers of amastigote nests were found in the stomach and parasitemia levels were higher. Our study is novel in that it shows that gastric mucosal damage caused by aspirin, a commonly used NSAID, facilitates T. cruzi infection by the oral route.
Collapse
Affiliation(s)
- Luana Aparecida Cossentini
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | | | | | - Lucy Megumi Yamauchi
- Departamento de Microbiologia, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Eduardo José De Almeida Araújo
- Laboratório de Neurogastroenterologia, Departamento de Histologia, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Phileno Pinge-Filho
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Universidade Estadual de Londrina, Londrina, Paraná, Brazil.
| |
Collapse
|
7
|
Shum AMY, Fung DCY, Corley SM, McGill MC, Bentley NL, Tan TC, Wilkins MR, Polly P. Cardiac and skeletal muscles show molecularly distinct responses to cancer cachexia. Physiol Genomics 2015; 47:588-99. [PMID: 26395599 DOI: 10.1152/physiolgenomics.00128.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 09/18/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer cachexia is a systemic, paraneoplastic syndrome seen in patients with advanced cancer. There is growing interest in the altered muscle pathophysiology experienced by cachectic patients. This study reports the microarray analysis of gene expression in cardiac and skeletal muscle in the colon 26 (C26) carcinoma mouse model of cancer cachexia. A total of 268 genes were found to be differentially expressed in cardiac muscle tissue, compared with nontumor-bearing controls. This was fewer than the 1,533 genes that changed in cachectic skeletal muscle. In addition to different numbers of genes changing, different cellular functions were seen to change in each tissue. The cachectic heart showed signs of inflammation, similar to cachectic skeletal muscle, but did not show the upregulation of ubiquitin-dependent protein catabolic processes or downregulation of genes involved in cellular energetics and muscle regeneration that characterizes skeletal muscle cachexia. Quantitative PCR was used to investigate a subset of inflammatory genes in the cardiac and skeletal muscle of independent cachectic samples; this revealed that B4galt1, C1s, Serpina3n, and Vsig4 were significantly upregulated in cardiac tissue, whereas C1s and Serpina3n were significantly upregulated in skeletal tissue. Our skeletal muscle microarray results were also compared with those from three published microarray studies and found to be consistent in terms of the genes differentially expressed and the functional processes affected. Our study highlights that skeletal and cardiac muscles are affected differently in the C26 mouse model of cachexia and that therapeutic strategies cannot assume that both muscle types will show a similar response.
Collapse
Affiliation(s)
- Angie M Y Shum
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, New South Wales, Australia
| | - David C Y Fung
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales Australia, New South Wales, Australia
| | - Susan M Corley
- New South Wales Systems Biology Initiative, University of New South Wales Australia, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales Australia, New South Wales, Australia
| | - Max C McGill
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, New South Wales, Australia
| | - Nicholas L Bentley
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Mitochondrial Bioenergetics Group, Department of Pharmacology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, New South Wales, Australia
| | - Timothy C Tan
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Blacktown Clinical School and Blacktown Hospital, Blacktown, New South Wales, Australia; and Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Boston, Massachusetts
| | - Marc R Wilkins
- New South Wales Systems Biology Initiative, University of New South Wales Australia, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales Australia, New South Wales, Australia
| | - Patsie Polly
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, New South Wales, Australia;
| |
Collapse
|
8
|
Magierowski M, Magierowska K, Kwiecien S, Brzozowski T. Gaseous mediators nitric oxide and hydrogen sulfide in the mechanism of gastrointestinal integrity, protection and ulcer healing. Molecules 2015; 20:9099-123. [PMID: 25996214 PMCID: PMC6272495 DOI: 10.3390/molecules20059099] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 04/29/2015] [Accepted: 05/13/2015] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are known as biological messengers; they play an important role in human organism and contribute to many physiological and pathophysiological processes. NO is produced from l-arginine by constitutive NO synthase (NOS) and inducible NOS enzymatic pathways. This gaseous mediator inhibits platelet aggregation, leukocyte adhesion and contributes to the vessel homeostasis. NO is known as a vasodilatory molecule involved in control of the gastric blood flow (GBF) and the maintenance of gastric mucosal barrier integrity in either healthy gastric mucosa or that damaged by strong irritants. Biosynthesis of H2S in mammals depends upon two enzymes cystathionine-β-synthase and cystathionine γ-lyase. This gaseous mediator, similarly to NO and carbon monoxide, is involved in neuromodulation, vascular contractility and anti-inflammatory activities. For decades, H2S has been known to inhibit cytochrome c oxidase and reduce cell energy production. Nowadays it is generally considered to act through vascular smooth muscle ATP-dependent K+ channels, interacting with intracellular transcription factors and promote sulfhydration of protein cysteine moieties within the cell, but the mechanism of potential gastroprotective and ulcer healing properties of H2S has not been fully explained. The aim of this review is to compare current results of the studies concerning the role of H2S and NO in gastric mucosa protection and outline areas that may pose new opportunities for further development of novel therapeutic targets.
Collapse
Affiliation(s)
- Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, Cracow 31-531, Poland.
| | - Katarzyna Magierowska
- Department of Physiology, Jagiellonian University Medical College, Cracow 31-531, Poland.
| | - Slawomir Kwiecien
- Department of Physiology, Jagiellonian University Medical College, Cracow 31-531, Poland.
| | - Tomasz Brzozowski
- Department of Physiology, Jagiellonian University Medical College, Cracow 31-531, Poland.
| |
Collapse
|
9
|
Li X, Zhang J, Bao Y, Ye Y, Li W, Yin W, Guo F, Chen A. WITHDRAWN: Efficacy and safety of short-term treatment of naproxcinod in patients with osteoarthritis (OA) of the hip: A prospective, randomized study. Arch Gerontol Geriatr 2015:S0167-4943(15)00013-8. [PMID: 25912890 DOI: 10.1016/j.archger.2015.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 01/13/2015] [Accepted: 01/14/2015] [Indexed: 11/15/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Xingyan Li
- Joint Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Second Department of Limb Bones, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Jinming Zhang
- Joint Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuan Bao
- Joint Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yaping Ye
- Joint Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenkai Li
- Joint Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weifeng Yin
- Joint Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fengjin Guo
- Joint Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anmin Chen
- Joint Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
10
|
Xue J, Shang G, Tanaka Y, Saihara Y, Hou L, Velasquez N, Liu W, Lu Y. Dose-dependent inhibition of gastric injury by hydrogen in alkaline electrolyzed drinking water. Altern Ther Health Med 2014; 14:81. [PMID: 24589018 PMCID: PMC3944674 DOI: 10.1186/1472-6882-14-81] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 02/25/2014] [Indexed: 12/20/2022]
Abstract
Background Hydrogen has been reported to relieve damage in many disease models, and is a potential additive in drinking water to provide protective effects for patients as several clinical studies revealed. However, the absence of a dose–response relationship in the application of hydrogen is puzzling. We attempted to identify the dose–response relationship of hydrogen in alkaline electrolyzed drinking water through the aspirin induced gastric injury model. Methods In this study, hydrogen-rich alkaline water was obtained by adding H2 to electrolyzed water at one atmosphere pressure. After 2 weeks of drinking, we detected the gastric mucosal damage together with MPO, MDA and 8-OHdG in rat aspirin induced gastric injury model. Results Hydrogen-dose dependent inhibition was observed in stomach mucosal. Under pH 8.5, 0.07, 0.22 and 0.84 ppm hydrogen exhibited a high correlation with inhibitory effects showed by erosion area, MPO activity and MDA content in the stomach. Gastric histology also demonstrated the inhibition of damage by hydrogen-rich alkaline water. However, 8-OHdG level in serum did not have significant hydrogen-dose dependent effect. pH 9.5 showed higher but not significant inhibitory response compared with pH 8.5. Conclusions Hydrogen is effective in relieving the gastric injury induced by aspirin-HCl, and the inhibitory effect is dose-dependent. The reason behind this may be that hydrogen-rich water directly interacted with the target tissue, while the hydrogen concentration in blood was buffered by liver glycogen, evoking a suppressed dose–response effect. Drinking hydrogen-rich water may protect healthy individuals from gastric damage caused by oxidative stress.
Collapse
|
11
|
Anti-inflammatory and vasoprotective activity of a retroviral-derived peptide, homologous to human endogenous retroviruses: endothelial cell effects. PLoS One 2012; 7:e52693. [PMID: 23285152 PMCID: PMC3527569 DOI: 10.1371/journal.pone.0052693] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 11/21/2012] [Indexed: 01/13/2023] Open
Abstract
Malignant and inflammatory tissues sometimes express endogenous retroviruses or their proteins. A highly-conserved sequence from retroviral transmembrane (TM) proteins, termed the “immunosuppressive domain (ID)”, is associated with inhibition of immune and inflammatory functions. An octadecapeptide (MN10021) from the ID of retroviral TM protein p15E inhibits in vitro release of pro-inflammatory cytokines and increases synthesis of anti-inflammatory IL-10. We sought to determine if MN10021 has significant in vivo effects. MN10021, prepared by solid-phase synthesis, was dimerized through a naturally-occurring, carboxy-terminal cysteine. In vivo anti-inflammatory activity was determined using a murine model of sodium periodate (NaIO4)-induced peritonitis. In vivo vasoprotective effects were determined using: (1) a carrageenan-induced model of disseminated intravascular coagulation (DIC) in mice; (2) a reverse passive Arthus model in guinea pigs; and (3) vasoregulatory effects in spontaneously hypertensive rats (SHR). In vitro studies included: (1) binding/uptake of MN10021 using human monocytes, cultured fibroblasts, and vascular endothelial cells (VEC); (2) gene expression by RT-PCR of MN10021-treated VEC; and (3) apoptosis of MN10021-treated VEC exposed to staurosporine or TNF-α. One-tenth nmol MN10021 inhibits 50 percent of the inflammatory response in the mouse peritonitis model. Furthermore, 73 nmol MN10021 completely protects mice in a lethal model of carrageenan-induced DIC and inhibits vascular leak in both the mouse DIC model and a guinea pig reverse passive Arthus reaction. MN10021 binds to and is taken up in a specific manner by both human monocytes and VEC but not by cultured human fibroblasts. Surprisingly, orally-administered MN10021 lowers blood pressure in SHR rats by 10–15% within 1 h suggesting a direct or indirect effect on the vascular endothelium. MN10021 and derived octapeptides induce iNOS (inducible nitric oxide synthase) mRNA in VEC and nitrate in VEC cell culture supernatants and protect VEC from induced apoptosis or necrosis. However, pretreatment of VEC with nitro-L-arginine methyl ester (L-NAME), while inhibiting the release of nitrate, does not block the anti-apoptotic effect of MN10021 and derived octapeptides suggesting that their potent vasoprotective and anti-inflammatory activity is not nitric oxide dependent.
Collapse
|
12
|
Romero C, Benedí J, Villar A, Martín-Aragón S. Involvement of Hsp70, a stress protein, in the resistance of long-term culture of PC12 cells against sodium nitroprusside (SNP)-induced cell death. Arch Toxicol 2010; 84:699-708. [PMID: 20559625 DOI: 10.1007/s00204-010-0565-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 05/31/2010] [Indexed: 11/30/2022]
Abstract
Sodium nitroprusside (SNP)-treated PC12 cell line is being used in our laboratory as a cell model of nitric oxide (NO)-mediated damage for in vitro evaluation of potential neuroprotective compounds, thus cell response to SNP must be standardized to gain reproducible data. The NO-donor SNP has been shown to induce cell death at high concentrations in undifferentiated PC12 cells. Differences were found in sensitivity to SNP between cells from short- and long-term cultured cells. After 24-h exposure to 100-500 microM SNP, a decrease of cell viability was observed in both short- (17, 21 and 23rd passages) and long-term cultures (46, 49 and 50th passages), with IC(50) values of 312.72 and 462.90 microM, respectively. In cells from early passages, SNP-induced cell death was accompanied by significant increases of LDH leakage, nitrite production, malondialdehyde (MDA) levels, catalase (CAT) activity, cleavage of poly(ADP-ribose)polymerase (PARP) and caspase-3 activation in comparison with those from late passages. Furthermore, untreated and SNP-treated cells from long-term cultures displayed an increase of the stress protein Hsp70 levels when compared with those from short-term cultures. Up-regulated levels of Hsp70 may be associated with cell survival. Therefore, cells may acquire a certain resistance to SNP-induced toxicity associated with an increase in cell passage-dependent Hsp70. The protein Hsp70 might modulate the cellular response to the toxic insult by increasing CAT and GSH-Px activities and decreasing caspase-3 activation. Finally, it is crucial for the standardization of this cell model of neurotoxicity, at least in part, the use of PC12 cells in an optimum and reliable range of passages.
Collapse
Affiliation(s)
- Carmen Romero
- Pharmacology Department, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | | | | | | |
Collapse
|
13
|
Maity P, Bindu S, Dey S, Goyal M, Alam A, Pal C, Reiter R, Bandyopadhyay U. Melatonin reduces indomethacin-induced gastric mucosal cell apoptosis by preventing mitochondrial oxidative stress and the activation of mitochondrial pathway of apoptosis. J Pineal Res 2009; 46:314-23. [PMID: 19220725 DOI: 10.1111/j.1600-079x.2009.00663.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Augmentation of gastric mucosal cell apoptosis due to development of oxidative stress is one of the main pathogenic events in the development of nonsteroidal anti-inflammatory drug (NSAID)-induced gastropathy. Identification of a nontoxic, anti-apoptotic molecule is warranted for therapy against NSAID-induced gastropathy. The objective of the present study was to define the mechanism of the anti-apoptotic effect of melatonin, a nontoxic molecule which scavenges reactive oxygen species. Using an array of experimental approaches, we have shown that melatonin prevents the development of mitochondrial oxidative stress and activation of mitochondrial pathway of apoptosis induced by indomethacin (a NSAID) in the gastric mucosa. Melatonin inhibits the important steps of indomethacin-induced activation of mitochondrial pathway of apoptosis such as upregulation of the expression of Bax and Bak, and the downregulation of Bcl-2 and BclxL. Melatonin also prevents indomethacin-induced mitochondrial translocation of Bax and prevents the collapse of mitochondrial membrane potential. Moreover, melatonin reduces indomethacin-mediated activation of caspase-9 and caspase-3 by blocking the release of cytochrome c and finally rescues gastric mucosal cells from indomethacin-induced apoptosis as measured by the TUNEL assay. Histologic studies of gastric mucosa further document that melatonin almost completely protects against gastric damage induced by indomethacin. Thus, melatonin has significant anti-apoptotic effects to protect gastric mucosa from NSAID-induced apoptosis and gastropathy, which makes its use as potential therapy against gastric damage during NSAID treatment.
Collapse
Affiliation(s)
- Pallab Maity
- Department of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Kolkata, India
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
The gastrointestinal adverse effects of nonsteroidal anti-inflammatory drugs (NSAIDs) have been recognized since shortly after the introduction of aspirin to the marketplace over a century ago. However, the underlying pathogenesis of NSAID-induced gastropathy remains incompletely understood. Advances in understanding some of the factors that contribute to the mucosal injury have provided clues for the development of safer NSAIDs. The inhibitory effects of nitric oxide (NO) on NSAID-induced leukocyte adherence were exploited in the development of NO-releasing NSAIDs. As well as eliciting less gastrointestinal damage than conventional NSAIDs, these drugs do not elevate blood pressure and show anti-inflammatory effects, additional to those of the parent drugs. Modification of other drugs in a similar manner (i.e., NO-releasing derivatives) has similarly resulted in more effective drugs. More recently, hydrogen sulphide-releasing derivatives of NSAIDs and of other drugs, have been developed, based on the observed ability of H(2)S to reduce inflammation and pain in experimental models. H(2)S-releasing NSAIDs produce negligible gastric damage and exhibit enhanced anti-inflammatory potency as compared to the parent drugs. The NO-NSAIDs and H(2)S-releasing NSAIDs represent examples of new anti-inflammatory drugs with greatly reduced toxicity and improved therapeutic activity, both created through the concept of exploiting the beneficial effects of endogenous gaseous mediators.
Collapse
Affiliation(s)
- J L Wallace
- Inflammation Research Network, Department of Pharmacology and Therapeutics, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
15
|
Mamuk S, Melli M. Effect of aspirin, paracetamol and their nitric oxide donating derivatives on exudate cytokine and PGE2 production in zymosan-induced air pouch inflammation in rats. Eur J Pharmacol 2007; 561:220-5. [PMID: 17320862 DOI: 10.1016/j.ejphar.2007.01.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Revised: 01/11/2007] [Accepted: 01/16/2007] [Indexed: 10/23/2022]
Abstract
Effects of different doses of aspirin, compared to equimolar doses of nitric oxide (NO)-donating aspirin (NCX 4016), and of a single dose of paracetamol, compared to an equimolar dose of NO-donating paracetamol (NCX 701) were investigated in acute zymosan-induced air pouch inflammation in rats. Treatments were administered by orogastric route, and interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha) and prostaglandin E(2) (PGE(2)) levels in the exudates were analysed 4 h after zymosan injection by enzyme immunoassay (EIA). Aspirin, at 10, 30 and 100 mg/kg doses, increased IL-1beta levels in exudates, however, only the highest dose lead to a significant increase when compared to control, whereas a significant increase in TNF-alpha level was observed at all doses tested. NCX 4016, at equimolar doses for aspirin, i.e., 18.6, 55.8 and 186 mg/kg, respectively, did not cause any changes in exudate IL-1beta or TNF-alpha levels. These effects were significantly different, when aspirin was compared with the corresponding NCX 4016 group. Nevertheless, the ability of aspirin and NCX 4016 to inhibit PGE(2) synthesis in the exudate where comparable. Although paracetamol significantly increased exudate TNF-alpha level compared to the control group and NCX 701 group, neither paracetamol, nor NCX701 treatments changed the levels of exudate IL-1beta significantly. As expected, paracetamol and NCX 701 showed poor PGE(2) inhibition. At high doses, aspirin and NCX 4016 decreased the number of polymorphonuclear leukocytes in the exudate. However, this inhibition was not significantly different from the control group. Paracetamol and NO-paracetamol did not cause any change in the number of polymorphonuclear leukocytes in exudate. These results indicated that aspirin and NCX 4016 possessed different effects on cytokine production or release, despite the fact that both drugs inhibited the synthesis of PGE(2) in a similar way. Unlike paracetamol, which increased exudate TNF-alpha level, NCX 701 had no effect on TNF-alpha level in the exudates.
Collapse
Affiliation(s)
- Soner Mamuk
- Ankara University, School of Medicine, Department of Pharmacology and Clinical Pharmacology, Morfoloji Binasi, Sihhiye 06100, Ankara, Turkey
| | | |
Collapse
|
16
|
Gresele P, Momi S. Pharmacologic Profile and Therapeutic Potential of NCX 4016, a Nitric Oxide-releasing Aspirin, for Cardiovascular Disorders. ACTA ACUST UNITED AC 2006; 24:148-68. [PMID: 16961726 DOI: 10.1111/j.1527-3466.2006.00148.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
NCX 4016, 2-(acetyloxy)benzoic acid 3-[(nitrooxy)methyl]phenyl ester, is a new molecule in which a nitric oxide (NO)-releasing moiety is covalently linked to aspirin. After enzymatic metabolism, NCX 4016 releases both components. In vitro and in some animal models, these components exert their pharmacologic effects simultaneously. Nitric oxide (NO) is a small gaseous molecule that exerts several activities which may prevent atherothrombotic disorders. Moreover, it displays a protective activity on the gastric mucosa. NCX 4016 has been shown to inhibit platelet activation in vitro more effectively than aspirin, to inhibit smooth muscle cell proliferation, to exert an endothelial cell protective activity and to suppress the function of several inflammatory cells potentially involved in atherothrombosis. In animal models, NCX 4016 protected from platelet thromboembolism, prevented restenosis in atherosclerosis-prone animals, protected the heart from ischemia/reperfusion injury, and induced neoangiogenesis in critically ischemic limbs. Moreover, it displayed little or no gastric toxicity and appeared to protect stomach from noxious stimuli, including aspirin. NCX 4016 has been evaluated in healthy volunteers and found to inhibit platelet cyclo-oxygenase-1 (COX-1) similarly to or slightly less than aspirin, to raise the circulating levels of NO-degradation products, and to have little or no gastric toxicity in short term studies. In particular, in phase II studies, NCX 4016 had favorable effects on effort-induced endothelial dysfunction in intermittent claudication and on platelet-activation parameters elicited by short-term hyperglycemia in type II diabetics. In patients with type II diabetes the effects of NCX 4016 on microalbuminuria and on some hemodynamic parameters were promising. The pharmacokinetics of in vivo aspirin- and NO- released by NCX 4016, as well as the bioavailability of the two molecules, were not yet adequately studied. Also, the long-term tolerability of NCX 4016, as well as its possible effectiveness in preventing ischemic cardiovascular events and progression of atherosclerosis, should be explored.
Collapse
Affiliation(s)
- Paolo Gresele
- Department of Internal Medicine, Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy.
| | | |
Collapse
|
17
|
Ekong U, Zeng S, Dun H, Feirt N, Guo J, Ippagunta N, Guarrera JV, Lu Y, Weinberg A, Qu W, Ramasamy R, Schmidt AM, Emond JC. Blockade of the receptor for advanced glycation end products attenuates acetaminophen-induced hepatotoxicity in mice. J Gastroenterol Hepatol 2006; 21:682-8. [PMID: 16677153 DOI: 10.1111/j.1440-1746.2006.04225.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIM Severe injury to the liver, such as that induced by toxic doses of acetaminophen, triggers a cascade of events leading to hepatocyte death. It is hypothesized that activation of the receptor for advanced glycation end products (RAGE) might contribute to acetaminophen-induced liver toxicity by virtue of its ability to generate reactive oxygen species, at least in part via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and thereby activate downstream signaling pathways leading to cellular injury. METHODS A model was employed in which toxic doses of acetaminophen (1125 mg/kg) were administered to C57BL/6 mice. To block RAGE, mice received murine soluble (s) RAGE, the extracellular ligand binding domain of the receptor that acts as a decoy to interrupt ligand-RAGE signaling. RESULTS Animals treated with sRAGE displayed increased survival compared with vehicle treatment, and markedly decreased hepatic necrosis. Consistent with an important role for RAGE-triggered oxidant stress in acetaminophen-induced injury, a significant reduction of nitrotyrosine protein adducts was observed in hepatic tissue in sRAGE-treated versus vehicle-treated mice receiving acetaminophen, in parallel with significantly increased levels of glutathione. In addition, pro-regenerative cytokines tumor necrosis factor-alpha and interleukin-6 were increased in sRAGE-treated versus vehicle-treated mice. CONCLUSION These findings implicate RAGE-dependent mechanisms in acetaminophen-induced liver damage and suggest that blockade of this pathway may impart beneficial effects in toxin-induced liver injury.
Collapse
Affiliation(s)
- Udeme Ekong
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Bolla M, Momi S, Gresele P, Del Soldato P. Nitric oxide-donating aspirin (NCX 4016): an overview of its pharmacological properties and clinical perspectives. Eur J Clin Pharmacol 2005. [DOI: 10.1007/s00228-005-0026-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
19
|
Tomisato W, Tanaka KI, Tsutsumi S, Hoshino T, Yokomizo K, Suzuki K, Katsu T, Mizushima T, Mizushima T. Low direct cytotoxicity and cytoprotective effects of nitric oxide releasing indomethacin. Dig Dis Sci 2005; 50:1927-37. [PMID: 16187199 DOI: 10.1007/s10620-005-2963-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Accepted: 01/21/2005] [Indexed: 01/06/2023]
Abstract
Nitric oxide (NO) releasing non-steroidal anti-inflammatory drugs (NSAIDs) have shown a marked reduction of gastrointestinal side effects and we here examined the cytotoxicity of NCX 530 (NO-indomethacin). Under conditions where indomethacin clearly induced both necrosis and apoptosis, NCX 530 induced neither. NCX 530 protected cells from celecoxib-induced necrosis and apoptosis. NCX 530 partially suppressed celecoxib-dependent membrane permeabilization and an inhibitor for guanylate cyclase suppressed the cytoprotective effect of NCX 530 against celecoxib. In vivo, NCX 530 alone produced fewer gastric lesions in rats than did indomethacin. A combination of the oral administration of celecoxib together with the intraperitoneal administration of indomethacin, but not of NCX 530, clearly resulted in the production of gastric lesions. The low direct cytotoxicity and the cytoprotective effect of NCX 530 observed in vitro may also act in vivo, thus ensuring that NCX 530 is safe for use on the gastric mucosa.
Collapse
Affiliation(s)
- Wataru Tomisato
- Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Furlan R, Kurne A, Bergami A, Brambilla E, Maucci R, Gasparini L, Butti E, Comi G, Ongini E, Martino G. A nitric oxide releasing derivative of flurbiprofen inhibits experimental autoimmune encephalomyelitis. J Neuroimmunol 2004; 150:10-9. [PMID: 15081244 DOI: 10.1016/j.jneuroim.2004.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2003] [Revised: 01/05/2004] [Accepted: 01/05/2004] [Indexed: 11/20/2022]
Abstract
Nitric oxide (NO)-releasing non-steroidal anti-inflammatory drugs (NSAIDs) have been reported to have a safer profile and additional anti-inflammatory and immuno-modulatory properties compared to parent compounds. Preventive treatment of experimental autoimmune encephalomyelitis (EAE)-induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein (MOG) peptide 35-55-with the NO-releasing derivative of flurbiprofen HCT1026 delayed disease onset and significantly decreased disease severity. HCT1026 treatment was associated to (i) decreased mRNA levels of pro-inflammatory cytokines, caspase-1, and iNOS in blood cells; (ii) decreased ability of encephalitogenic T cells to proliferate; (iii) reduced number of central nervous system (CNS)-infiltrating T cells; (iv) decreased axonal loss and demyelination; (v) increased CD4(+) CD69(-) CD25(+) regulatory T cells in the spleen.
Collapse
MESH Headings
- Administration, Oral
- Adoptive Transfer
- Animals
- Antigens, CD/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- Cell Movement/drug effects
- Cell Movement/immunology
- Central Nervous System/drug effects
- Central Nervous System/pathology
- Cytokines/antagonists & inhibitors
- Cytokines/genetics
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Epitopes, T-Lymphocyte/immunology
- Female
- Flurbiprofen/administration & dosage
- Flurbiprofen/analogs & derivatives
- Glycoproteins/administration & dosage
- Glycoproteins/immunology
- Growth Inhibitors/administration & dosage
- Immunosuppressive Agents/administration & dosage
- Interleukin-10/deficiency
- Interleukin-10/genetics
- Lectins, C-Type
- Lymphocyte Count
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein
- Nitric Oxide/metabolism
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- RNA, Messenger/antagonists & inhibitors
- Receptors, Interleukin-2/biosynthesis
- Spleen/immunology
- Spleen/pathology
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- Roberto Furlan
- Neuroimmunology Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Dutta S, Padhye S, Mckee V. Structural characterization and SOD activity of copper–oxaprozinate. INORG CHEM COMMUN 2004. [DOI: 10.1016/j.inoche.2004.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
22
|
Lamarque D. Physiopathologie des lésions gastro-duodénales induites par les anti-inflammatoires non stéroïdiens. ACTA ACUST UNITED AC 2004; 28 Spec No 3:C18-26. [PMID: 15366671 DOI: 10.1016/s0399-8320(04)95275-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The pathogenesis of the gastroduodenal lesions induced by non-steroidal anti-inflammatory drugs and aspirin is primarily caused by a reduction in mucosal blood flow, which is the consequence of inhibition of cyclooxygenase-producing vasodilator prostaglandins. The subsequent phase is adherence of leukocytes to the endothelium, which may depend on cyclooxygenase-2. Endothelial lesions accentuate the fall of mucosal blood flow and promote the inflammatory process in the gastric mucosa. The inflammatory process is amplified by expression of TNFalpha in polymorphonuclears induced by non-steroidal anti-inflammatory drugs. A few days after starting treatment, epithelial proliferation and increased mucosal blood flow, partly dependent on cyclooxygenase-2 and nitric oxide expression, compensates for the damaging process. Selective inhibitors of inducible cyclooxygenase-2 have reduced gastrointestinal toxicity, which could partially be explained by the protection effect of cyclooxygenase-2 on the gastrointestinal mucosa during inflammation or epithelial repair. Selective inhibitors may worsen inflammatory bowel disease. Non-steroidal inflammatory drugs and aspirin, but perhaps not selective inhibitors, increase the mucosal lesions associated with Helicobacter pylori-induced gastritis. Co-administration of selective inhibitors and aspirin leads to gastrointestinal toxicity equivalent to that of non-specific anti-inflammatory drugs.
Collapse
|
23
|
Salgueiro-Pagadigorria CL, Kelmer-Bracht AM, Bracht A, Ishii-Iwamoto EL. Naproxen affects Ca2+ fluxes in mitochondria, microsomes and plasma membrane vesicles. Chem Biol Interact 2004; 147:49-63. [PMID: 14726152 DOI: 10.1016/j.cbi.2003.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
There is substantial evidence that nonsteroidal anti-inflammatory drugs (NSAIDs) affect cellular processes regulated by Ca(2+) ions, including the metabolic responses of the liver to Ca(2+)-dependent hormones. The aim of the present study was to determine whether the effects of naproxen are mediated by a direct action on cellular Ca(2+) fluxes. The effects of naproxen on 45Ca(2+) fluxes in mitochondria, microsomes and inside-out plasma membrane vesicles were examined. Naproxen strongly impaired the mitochondrial capacity to retain 45Ca(2+) and inhibited also ATP-dependent 45Ca(2+) uptake by microsomes. Naproxen did not modify 45Ca(2+) uptake by inside-out plasma membrane vesicles, but it inhibited the hexokinase/glucose-induced Ca(2+) efflux from preloaded vesicles. Additional assays performed in isolated mitochondria revealed that naproxen causes mitochondrial uncoupling and swelling in the presence of Ca(2+) ions. These effects were prevented by EGTA, ruthenium red and cyclosporin A, indicating that naproxen acts synergistically with Ca(2+) ions by promoting the mitochondrial permeability transition. The experimental results suggest that naproxen may impair the metabolic responses to Ca(2+)-dependent hormones acting by at least two mechanisms: (1) by interfering with the supply of external Ca(2+) through a direct action on the plasma membrane Ca(2+) influx, and (2) by affecting the refilling of the agonist-sensitive internal stores, including endoplasmic reticulum and mitochondria.
Collapse
|
24
|
Walley M, Hotz-Behofsits C, Simpson R, Bjarnason I. Nitric oxide: potential role for reducing gastro-enteropathy. Inflammopharmacology 2003; 11:429-36. [PMID: 15035796 DOI: 10.1163/156856003322699609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The pathogenesis of non-steroidal anti-inflammatory drug (NSAID)-induced gastroenteropathy may involve a number of key events leading to increased intestinal permeability and inflammation (topical effect) and the development of ulcers (micro-vascular effects of COX-1 inhibition and prostaglandin deficiency). Many strategies have been employed in an attempt to reduce the toxic effects of NSAIDs and these have been targeted at the different pathogenic stages of lesion development. One of the latest in this long chain of damage limitation has been the development of nitric oxide (NO) sequestering NSAIDs (NO-NSAIDs). It is suggested that the NO, which is released as the compounds are broken down, may counteract the consequences of the NSAID-induced decrease in mucosal prostaglandins. Here we examine the proposed mechanisms for NSAID-induced gastrointestinal damage together with some of the methods employed to address these mechanisms. We also consider the physiologic roles of NO in the gut together with how it may be potentially employed as an agent for limiting the side effects of NSAIDs in the gastrointestinal tract.
Collapse
Affiliation(s)
- Matthew Walley
- Department of Medicine, Guys, Kings and St Thomas' School of Medicine, Bessemer Road, London SE5 9PJ, UK.
| | | | | | | |
Collapse
|
25
|
Wallace JL. Nitric oxide-releasing mesalamine: potential utility for treatment of inflammatory bowel disease. Dig Liver Dis 2003; 35 Suppl 2:S35-40. [PMID: 12846442 DOI: 10.1016/s1590-8658(03)00050-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nitric oxide can accelerate ulcer healing and exert anti-inflammatory effects. Addition of a nitric oxide-releasing moiety to mesalamine significantly boosts its anti-inflammatory activity. NO-releasing mesalamine suppresses inflammatory cytokine production and reduces leukocyte infiltration.
Collapse
Affiliation(s)
- J L Wallace
- Mucosal Inflammation Research Group, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
26
|
Abstract
Nitric oxide-releasing aspirins are new chemical entities obtained by adding a nitric oxide-releasing moiety to aspirin. NCX-4016 is the prototype of this family of molecules. NCX-4016 consists of the parent molecule (aspirin) linked to a 'spacer' via an ester linkage, which is in turn connected to a nitric oxide-releasing moiety. Both aspirin and nitric oxide moieties of NCX-4016 contribute to its effectiveness, the latter occurring via both cyclic guanosyl monophosphate-dependent and -independent mechanisms. In vitro studies have shown that NCX-4016 inhibits platelet aggregation induced by aspirin-sensitive (arachidonic acid) and aspirin-insensitive (thrombin) agonist. In contrast to aspirin, NCX-4016 exerts a multilevel regulation of inflammatory target, including caspase-1 and NF-kappaB. This broad spectrum of activities translates to an increased potency of this drug in modulating cardiovascular inflammation. Human studies have shown, that while nitric oxide-aspirin maintains its anti-thrombotic activity, it spares the gastrointestinal tract. Indeed, a 7-day course of NCX-4016 results in 90% reduction of gastric damage caused by equimolar doses of aspirin. Further studies are ongoing to define whether this superior anti-inflammatory and anti-thrombotic profile translates in clinical benefits in patients with cardiovascular diseases.
Collapse
Affiliation(s)
- S Fiorucci
- Gastrointestinal and Liver Unit, Department of Internal Medicine, University of Perugia, Perugia, Italy.
| | | |
Collapse
|
27
|
Weder JE, Dillon CT, Hambley TW, Kennedy BJ, Lay PA, Biffin J, Regtop HL, Davies NM. Copper complexes of non-steroidal anti-inflammatory drugs: an opportunity yet to be realized. Coord Chem Rev 2002. [DOI: 10.1016/s0010-8545(02)00086-3] [Citation(s) in RCA: 360] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Keeble JE, Moore PK. Pharmacology and potential therapeutic applications of nitric oxide-releasing non-steroidal anti-inflammatory and related nitric oxide-donating drugs. Br J Pharmacol 2002; 137:295-310. [PMID: 12237248 PMCID: PMC1573498 DOI: 10.1038/sj.bjp.0704876] [Citation(s) in RCA: 132] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2001] [Revised: 03/03/2002] [Accepted: 03/15/2002] [Indexed: 01/25/2023] Open
Abstract
This review examines the biological significance, therapeutic potential and mechanism(s) of action of a range of nitric oxide-releasing non-steroidal anti-inflammatory drugs (NO-NSAID) and related nitric oxide-releasing donating drugs (NODD). The slow release of nitric oxide (NO) from these compounds leads to subtle changes in the profile of pharmacological activity of the parent, non-steroidal anti-inflammatory drugs (NSAID). For example, compared with NSAID, NO-NSAID cause markedly diminished gastrointestinal toxicity and improved anti-inflammatory and anti-nociceptive efficacy. In addition, nitroparacetamol exhibits hepatoprotection as opposed to the hepatotoxic activity of paracetamol. The possibility that NO-NSAID or NODD may be of therapeutic benefit in a wide variety of disease states including pain and inflammation, thrombosis and restenosis, neurodegenerative diseases of the central nervous system, colitis, cancer, urinary incontinence, liver disease, impotence, bronchial asthma and osteoporosis is discussed.
Collapse
Affiliation(s)
- J E Keeble
- Centre for Cardiovascular Biology and Medicine, King's College, University of London, Guy's Campus, London SE1 9RT
| | - P K Moore
- Centre for Cardiovascular Biology and Medicine, King's College, University of London, Guy's Campus, London SE1 9RT
| |
Collapse
|
29
|
Wallace JL, Ignarro LJ, Fiorucci S. Potential cardioprotective actions of no-releasing aspirin. Nat Rev Drug Discov 2002; 1:375-82. [PMID: 12120413 DOI: 10.1038/nrd794] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The use of low doses of aspirin on a daily basis has increased greatly in the past 20 years, based on observations that it can significantly reduce the risk of heart attacks and strokes. However, aspirin can also cause severe damage to the stomach. A modified version of aspirin that releases nitric oxide has been developed that seems to offer important advantages over its 103-year-old parent--namely, improved protection for the heart without the unwanted effects on the stomach.
Collapse
Affiliation(s)
- John L Wallace
- Department of Pharmacology & Therapeutics, University of Calgary, Calgary, Alberta, T2N 4N1 Canada.
| | | | | |
Collapse
|
30
|
Naito Y, Handa O, Takagi T, Ishikawa T, Imamoto E, Nakagawa S, Yamaguchi T, Yoshida N, Matsui H, Yoshikawa T. Ubiquitin-proteasome inhibitor enhances tumour necrosis factor-alpha-induced apoptosis in rat gastric epithelial cells. Aliment Pharmacol Ther 2002; 16 Suppl 2:59-66. [PMID: 11966525 DOI: 10.1046/j.1365-2036.16.s2.30.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
BACKGROUND Tumour necrosis factor (TNF-alpha) is a candidate factor for involvement in inflammation-mediated gastric mucosal injury. However, the effect of this cytokine on gastric epithelial cells has been poorly investigated. In the present study, we examined whether gastric epithelial cells are resistant to TNF-alpha-induced apoptosis, and whether this resistance is related to ubiquitin-proteasome-associated nuclear factor-kappaB (NF-kappaB) activation. METHODS The rat gastric mucosal cell line RGM-1 was grown in DMEM/F12 medium supplemented with 10% FCS. Confluent monolayers of cells were pretreated or not for 60 min with PSI, a peptide aldehyde known to specifically inhibit the chymotrypsin-like activity of 26S proteasome. Cells were subsequently stimulated with recombinant rat TNF-alpha and their viability was determined by WST-1 assay. Apoptosis was confirmed by fluorescence microscopy after staining with Hoechst 33342 and propidium iodide, and DNA fragmentation was determined by flow cytometry using an APO-BRDU kit. IkappaB-alpha and the p65 binding subunit of NF-kappaB were detected by Western blots. RESULTS Twenty-four-hour incubation with TNF-alpha alone or PSI alone did not affect the cell viability of RGM-1 cells. Pretreatment with PSI significantly enhanced the level of apoptosis induced by TNF-alpha. In RGM-1 cells treated with TNF-alpha, cytoplasmic IkappaB-alpha decreased and p65 in nuclear extracts increased markedly 30 min after cytokine stimulation. Pretreatment with PSI at 12.5 micromol/L blocked these TNF-alpha-induced changes. CONCLUSION PSI enhances TNF-alpha-induced apoptosis through inhibition of NF-kappaB activation in RGM-1 cells.
Collapse
Affiliation(s)
- Y Naito
- First Department of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fiorucci S, Antonelli E, Mencarelli A, Palazzetti B, Alvarez-Miller L, Muscara M, del Soldato P, Sanpaolo L, Wallace JL, Morelli A. A NO-releasing derivative of acetaminophen spares the liver by acting at several checkpoints in the Fas pathway. Br J Pharmacol 2002; 135:589-99. [PMID: 11834606 PMCID: PMC1573169 DOI: 10.1038/sj.bjp.0704500] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
NCX-701 is a nitric oxide (NO)-releasing acetaminophen (APAP) derivative. In the present study we demonstrated that NCX-701 is as effective as APAP in controlling body temperature in a rat model of endotoxin-induced fever. Liver toxicity is a major complication of APAP overdosing. To investigate whether NCX-701 is hepatotoxic, BALB/C mice were injected with 100 - 500 mg kg(-1) APAP or NCX-701 alone or in combination (i.e. 500 mg kg(-1) of both compounds). Our results demonstrated that although APAP caused a dose-dependent liver injury, NCX-701 was completely devoid of liver toxicity. At the dose of 500 mg kg(-1) APAP caused an approximately 40 fold increase of AST plasma levels and extensive centrilobular necrosis. APAP and NCX-701 share the same metabolic pathway as demonstrated by the time-course of APAP-glucuronide concentrations in plasma and liver. NCX-701 was safe in mice with pre-existing chronic liver disease. Indeed, while C57BL6 transgenic mice expressing the hepatitis B virus (HBV) at the age of 8 months were significantly more susceptible to liver damage induced by APAP (500 mg kg(-1)) than their congenic littermates, treating HBV-transgenic mice with NCX-701, 500 mg kg(-1), caused no damage. Co-administration of NCX-701 at the dose 500 mg kg(-1) to mice treated with APAP, 500 mg kg(-1), completely protected against liver damage induced by APAP. APAP, but not NCX-701, upregulated liver Fas and Fas Ligand mRNA expression in vivo. Incubating mouse hepatocytes with APAP, but not with NCX-701, increased cell surface Fas expression and sensitized hepatocytes to death induced by challenge with a Fas-agonistic antibody. Collectively, these observations suggest that APAP toxicity is Fas mediated and that NCX-701 spares the liver by acting at several checkpoints in the Fas pathway.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina Clinica e Sperimentale, Clinica di Gastroenterologia ed Epatologia, Università degli Studi di Perugia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kono Y, Sawada S, Kawahara T, Tsuda Y, Higaki T, Yamasaki S, Imamura H, Tada Y, Sato T, Hiranuma O, Akamatsu N, Komatsu S, Tamagaki T, Nakagawa K, Tsuji H, Nakagawa M. Bradykinin inhibits serum-depletion-induced apoptosis of human vascular endothelial cells by inducing nitric oxide via calcium ion kinetics. J Cardiovasc Pharmacol 2002; 39:251-61. [PMID: 11791011 DOI: 10.1097/00005344-200202000-00012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Vascular endothelial cells play important roles in atherogenesis, and bradykinin is associated with atherosclerosis. The effect of bradykinin on apoptosis in human umbilical vein endothelial cells (HUVECs) was investigated, with a focus on Ca2+ kinetics and nitric oxide production. In serum-free conditions, the number of apoptotic cells increased in a time-dependent manner, but this increase was inhibited by bradykinin in a dose-dependent manner. The apoptosis inhibited by bradykinin was reduced by nitric oxide inhibitor N(G)-monomethyl-L-arginine (L-NMMA) and consequently restored by combined treatment with L-NMMA and L-arginine. Bradykinin increased influx of extracellular Ca2+, generation of inositol 1,4,5-trisphosphate, and release of Ca2+ from intracellular storage sites, thus increasing the total intracellular Ca2+ concentration ([Ca2+]i). Bradykinin increased nitric oxide production, which was inhibited by L-NMMA and restored by combined treatment with L-NMMA and L-arginine. Sodium nitroprusside (SNP) dose-dependently increased nitric oxide production and inhibited apoptosis; however, 10(-5) M SNP did not inhibit apoptosis. Caspase-3 inhibitor, acetyl-Asp-Met-Gln-Asp-aldehyde, enhanced bradykinin-induced inhibition of apoptosis but did not effect bradykinin-induced nitric oxide production. These findings suggest that bradykinin inhibits serum-depletion-induced apoptosis in HUVECs by enhancing nitric oxide production via an increase in [Ca2+]i.
Collapse
Affiliation(s)
- Yoshihito Kono
- Second Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Miller MJS, Angeles FM, Reuter BK, Bobrowski P, Sandoval M. Dietary antioxidants protect gut epithelial cells from oxidant-induced apoptosis. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2001; 1:11. [PMID: 11749672 PMCID: PMC61450 DOI: 10.1186/1472-6882-1-11] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2001] [Accepted: 12/10/2001] [Indexed: 02/07/2023]
Abstract
BACKGROUND The potential of ascorbic acid and two botanical decoctions, green tea and cat's claw, to limit cell death in response to oxidants were evaluated in vitro. METHODS Cultured human gastric epithelial cells (AGS) or murine small intestinal epithelial cells (IEC-18) were exposed to oxidants - DPPH (3 microM), H2O2 (50 microM), peroxynitrite (300 microM) - followed by incubation for 24 hours, with antioxidants (10 microg/ml) administered as a 1 hour pretreatment. Cell number (MTT assay) and death via apoptosis or necrosis (ELISA, LDH release) was determined. The direct interactions between antioxidants and DPPH (100 microM) or H2O2 (50 microM) were evaluated by spectroscopy. RESULTS The decoctions did not interact with H2O2, but quenched DPPH although less effectively than vitamin C. In contrast, vitamin C was significantly less effective in protecting human gastric epithelial cells (AGS) from apoptosis induced by DPPH, peroxynitrite and H2O2 (P < 0.001). Green tea and cat's claw were equally protective against peroxynitrite and H2O2, but green tea was more effective than cat's claw in reducing DPPH-induced apoptosis (P < 0.01). Necrotic cell death was marginally evident at these low concentrations of peroxynitrite and H2O2, and was attenuated both by cat's claw and green tea (P < 0.01). In IEC-18 cells, all antioxidants were equally effective as anti-apoptotic agents. CONCLUSIONS These results indicate that dietary antioxidants can limit epithelial cell death in response to oxidant stress. In the case of green tea and cat's claw, the cytoprotective response exceed their inherent ability to interact with the injurious oxidant, suggestive of actions on intracellular pathways regulating cell death.
Collapse
Affiliation(s)
- Mark JS Miller
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA
| | - Fausto M Angeles
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA
| | - Brian K Reuter
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA
| | | | - Manuel Sandoval
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA
| |
Collapse
|
34
|
Ajmone-Cat MA, Nicolini A, Minghetti L. Differential effects of the nonsteroidal antiinflammatory drug flurbiprofen and its nitric oxide-releasing derivative, nitroflurbiprofen, on prostaglandin E(2), interleukin-1beta, and nitric oxide synthesis by activated microglia. J Neurosci Res 2001; 66:715-22. [PMID: 11746392 DOI: 10.1002/jnr.10038] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Increasing experimental, clinical, and epidemiological studies point to the pivotal role of inflammation in the pathogenesis of acute and chronic neurodegenerative diseases and to the protective effects of nonsteroidal antiinflammatory drug (NSAID) therapies. Nonetheless, NSAID long-term therapies are limited by their significant adverse effects on gastrointestinal tract and kidneys. Nitroflurbiprofen (NO-flurbiprofen) belongs to a novel class of antiinflammatory agents obtained by derivatization of conventional NSAIDs with a nitric oxide (NO)-releasing moiety, which strongly reduces their untoward side effects without altering the antiinflammatory effectiveness. The recent evidence of neuroprotective effects of NO-NSAIDs in animal models of chronic brain inflammation prompted us to investigate the activities of NO-flurbiprofen and its parent molecule flurbiprofen on activated rat microglia, the brain resident macrophages. We found that NO-flurbiprofen was as potent as flurbiprofen in preventing prostaglandin E(2) synthesis in lipopolysaccharide-activated microglial cultures. At variance with previous observations on peripheral macrophages is that NO-flurbiprofen did not show any additional capacity to inhibit interleukin-1beta synthesis compared with flurbiprofen. Moreover, NO enhanced the expression of the inducible NO synthase; this effect was most likely attributable to the NO released from the drug, as suggested by experiments performed in the presence of the NO donor Deta-NONOate, which similarly to NO-flurbiprofen is characterised by a slow and long-lasting release. Our findings indicate that NO-NSAIDs may differently affect peripheral and brain macrophages. Given their potential therapeutic role in brain inflammation, further in vivo and in vitro studies are required to understand fully their mechanism of action in the CNS.
Collapse
Affiliation(s)
- M A Ajmone-Cat
- Laboratory of Pathophysiology, Istituto Superiore di Sanità, Rome, Italy
| | | | | |
Collapse
|
35
|
Janero DR. Nutritional aspects of nitric oxide: human health implications and therapeutic opportunities. Nutrition 2001; 17:896-903. [PMID: 11684398 DOI: 10.1016/s0899-9007(01)00647-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Nitric oxide (NO), the most potent natural vasorelaxant known, has close historical ties to cardiovascular physiology, despite NO's rich physiologic chemistry as an ubiquitous, signal-transducing radical. Aspects of NO biology critical to gastrointestinal health and, consequently, nutritional status are increasingly being recognized. Attempts are underway to exploit the gastrointestinal actions of NO for therapeutic gain. Cross-talk between NO and micronutrients within and outside the gastrointestinal system affects the establishment or progression of several diseases with pressing medical needs. These concepts imply that NO biology can influence nutrition and be nutritionally modulated to affect mammalian (patho)physiology. At least four nutritional facets of NO biology are at the forefront of contemporary biomedical research: 1) NO as modulator of feeding behavior and mediator of gastrointestinal homeostasis; 2) NO supplementation as a therapeutic modality for preserving gastrointestinal health; 3) interactions among elemental micronutrients (e.g., zinc), NO, and inflammation as potential contributors to diarrheal disease; and 4) vitamin micronutrients (e.g., vitamins E and C) as protectors of NO-dependent vascular function. Discussion of extant data on these topics prompts speculation that future research will broaden NO's nutritional role as an integrative signaling molecule supporting gastrointestinal and nutritional well-being.
Collapse
Affiliation(s)
- D R Janero
- NitroMed, Inc., 12 Oak Park Drive, Bedford, MA 01730, USA.
| |
Collapse
|
36
|
Minuz P, Degan M, Gaino S, Meneguzzi A, Zuliani V, Santonastaso CL, Soldato PD, Lechi A. NCX4016 (NO-Aspirin) has multiple inhibitory effects in LPS-stimulated human monocytes. Br J Pharmacol 2001; 134:905-11. [PMID: 11606332 PMCID: PMC1573016 DOI: 10.1038/sj.bjp.0704326] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
NCX4016 (2 acetoxy-benzoate 2-(2-nitroxymethyl)-phenyl ester, NicOx S.A., France) is an anti-thrombotic agent, chemically related to acetylsalicylic acid (ASA) and able to release NO. We tested the effects of NCX4016 and ASA on the release of the thromboxane (TX) A(2) metabolite TXB(2), tumour necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), expression and activity of tissue factor (TF) in stimulated, adherent human monocytes. Both ASA and NCX4016 1 - 1000 micromol l(-1) dose-dependently reduced TXB(2) concentration, measured by RIA in the supernatant of 10 microg ml(-1) LPS-stimulated cells. NCX4016 activity was comparable to that of equimolar ASA when incubation lasted 6 h (NCX4016 30 micromol l(-1): -86.0+/-10.1%, NCX4016 300 micromol l(-1): -92.2+/-9.0%, ASA 30 micromol l(-1): -92.3+/-7.5%, ASA 300 micromol l(-1): -97.3+/-1.0%, n=6, M+/-s.d.). Most of the activity of NCX4016 up to 100 micromol l(-1) was prevented by 10 micromol l(-1) ODQ, inhibitor of cyclic GMP. NCX4016 100 - 300 micromol l(-1) reduced TNF-alpha (NCX4016 300 micromol l(-1)=-77.2+/-19.9%, n=6) and IL-6 (NCX4016 300 micromol l(-1): -61.9+/-15.2%, n=6) in LPS stimulated monocytes while ASA had no significant effects. TF activity (NCX4016 300 micromol l(-1): 53.7+/-39.9%, n=4) and immunoreactive TF (NCX4016 300 micromol l(-1): -93.9+/-7.9%, n=7), measured in the supernatant of stimulated cells, were also dose-dependently inhibited by NCX4016 but not by ASA. The present results indicate that NCX4016 inhibits TXA(2) generation as well as cytokine release and TF in human monocytes partly via NO-dependent mechanisms. NCX4016 may have a favourable profile of activities in the clinical setting of athero-thrombosis.
Collapse
Affiliation(s)
- P Minuz
- Department of Biomedical and Surgical Sciences, University of Verona, Medicina Interna C, Policlinico G.B. Rossi, Piazzale L.A. Scuro, 10, 37134 Verona, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Calatayud S, Barrachina D, Esplugues JV. Nitric oxide: relation to integrity, injury, and healing of the gastric mucosa. Microsc Res Tech 2001; 53:325-35. [PMID: 11376493 DOI: 10.1002/jemt.1100] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nitric oxide (NO) plays a multifaceted role in mucosal integrity. The numerous functions of NO and the double-edged role played by NO in most of them provide a great complexity to the NO action. The three enzymatic sources of NO, neuronal NO-synthase (nNOS), endothelial NOS (eNOS), and inducible NOS (iNOS), have been characterised in the gastrointestinal tract. The protective properties of the NO derived from constitutive NO-synthases (eNOS and nNOS) have already been well established. Less clear is the role assigned to iNOS. The simplistic initial view of low levels of NO synthesised by constitutive NOS being protective while exaggerated NO levels after iNOS induction leading irremediably to cytotoxicity is being questioned by new evidence. As initially reported for constitutive NOS, iNOS activity may be associated to reduced leukocyte-endothelium interaction and platelet aggregation as well as protection of mucosal microcirculation. Moreover, iNOS activity may be important to resolve inflammation by increasing apoptosis in inflammatory cells. It is entirely possible that a low level of expression of iNOS will reflect a positive host-defense response to challenge, but that exaggerated or uncontrolled expression of iNOS itself becomes detrimental. There is no doubt about the protective role of NO in physiological conditions. However, when the mucosa is threatened, the role of NO becomes multiple and the final effect will probably depend on the nature of the insult, the environment involved, and the interaction with other mediators.
Collapse
Affiliation(s)
- S Calatayud
- Departmento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | | | | |
Collapse
|
38
|
Takeuchi K, Tanaka A. [Gastrointestinal sparing anti-inflammatory drugs--COX-2 selective inhibitors and NO-releasing NSAIDs]. Nihon Yakurigaku Zasshi 2001; 117:138-48. [PMID: 11233305 DOI: 10.1254/fpj.117.138] [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: 10/26/2022]
Abstract
The use of NSAIDs is associated with a wide array of alterations in the gastrointestinal integrity and function. Various approaches have been taken to develop NSAIDs with reduced gastrointestinal toxicity, and few have successfully reduced the incidence of adverse reactions. These include COX-2 selective inhibitors and NO-releasing NSAIDs. Much has been written about the potential of COX-2 inhibitors as antiinflammatory agents that lack the gastrointestinal side effects of traditional NSAIDs. COX-2 expression is most evident at sites of inflammation, while COX-1 accounts for most of the PG synthesis in the normal gastrointestinal tract. However, there are distinct examples of circumstances in which COX-2-derived PGs play a role in the maintenance of the mucosal integrity, and the differentiation of COX-1 and COX-2 is not quite as clear as has been suggested. On the other hand, the rational behind the NO-releasing NSAIDs is that NO released from the derivatives exerts beneficial effects on the gastrointestinal mucosa. The present article overviews the roles of COX and NO in housekeeping functions of the gastrointestinal mucosa in various circumstances and the effects of gastrointestinal sparing NSAIDs, such as COX-2 selective inhibitors and NO-releasing NSAIDs, on the ulcerogenic and healing responses in the gastrointestinal mucosa.
Collapse
Affiliation(s)
- K Takeuchi
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607-8414, Japan.
| | | |
Collapse
|
39
|
Mizoguchi H, Hase S, Tanaka A, Takeuchi K. Lack of small intestinal ulcerogenecity of nitric oxide-releasing indomethacin, NCX-530, in rats. Aliment Pharmacol Ther 2001; 15:257-67. [PMID: 11148446 DOI: 10.1046/j.1365-2036.2001.00916.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
AIM To evaluate the intestinal ulcerogenic property of nitric oxide-releasing indomethacin (NCX-530) in the rat, in comparison with indomethacin. METHODS Animals were given indomethacin or NCX-530 subcutaneously and killed 24 h later for macroscopic examination of the small intestine. RESULTS A single administration of indomethacin (10 mg/kg) provoked damage, mainly in the jejunum and ileum, accompanied by an increase in myeloperoxidase and inducible nitric oxide synthase activities as well as bacterial translocation. NCX-530 at an equimolar dose (14.2 mg/kg) caused no gross damage in the small intestine, nor any significant change in inducible nitric oxide synthase and myeloperoxidase activities or bacterial translocation. NOR-3, the nitric oxide donor (6.0 mg/kg), when administered subcutaneously together with indomethacin, significantly prevented the occurrence of intestinal lesions and other mucosal changes. Indomethacin reduced mucus and fluid secretions in the small intestine, while both NCX-530 and NOR-3 enhanced these secretions. NCX-530 reduced the mucosal prostaglandin E2 contents and exhibited an anti-inflammatory action against carrageenan-induced paw oedema, with equal effectiveness to indomethacin. CONCLUSION NCX-530 does not cause intestinal damage, despite inhibiting cyclooxygenase activity. The reduced intestinal toxicity of NCX-530 may be attributable to inhibition of enterobacterial translocation, partly by increasing the mucus and fluid secretions mediated by nitric oxide released from this compound.
Collapse
Affiliation(s)
- H Mizoguchi
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto, Japan
| | | | | | | |
Collapse
|
40
|
Paul-Clark M, Del Soldato P, Fiorucci S, Flower RJ, Perretti M. 21-NO-prednisolone is a novel nitric oxide-releasing derivative of prednisolone with enhanced anti-inflammatory properties. Br J Pharmacol 2000; 131:1345-54. [PMID: 11090106 PMCID: PMC1572462 DOI: 10.1038/sj.bjp.0703704] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2000] [Revised: 07/26/2000] [Accepted: 09/11/2000] [Indexed: 11/08/2022] Open
Abstract
1. Anti-inflammatory effects of a novel derivative of the glucocorticoid prednisolone were investigated. NCX-1015 (prednisolone 21-[(4'-nitrooxymethyl)benzoate]) incubation in human platelet-rich plasma produced a time (0 - 60 min) and concentration (3 - 300 microM) dependent release of nitrite, that was mirrored by accumulation of cyclic guanosine monophosphate in the human platelets. Intraperitoneal injection of NCX-1015 to mice (up to 27.7 micromol kg(-1)) produced nitrite accumulation in the peritoneal cavity maximal at 60 min. 2. NCX-1015 dose-dependently induced the steroid sensitive cell surface marker CD163 in human peripheral blood mononuclear cells (PBMCs). NCX-1015 was more potent than prednisolone in inducing CD163. Similarly, lipopolysaccharide induced interleukin-1 beta release from these cells was inhibited by NCX-1015 with higher potency than prednisolone. 3. In the zymosan peritonitis model, NCX-1015 was more active than prednisolone in suppressing neutrophil extravasation (ED(50) of 5.5 and 25.8 micromol kg(-1), respectively), nitrite accumulation (ED(50) of 1.38 and 22.2 micromol kg(-1), respectively) and release of the chemokine KC (ED(50) of 5.5 and 27.7 micromol kg(-1), respectively) as determined at the 4 h time-point. No differences were measured for the levels of interleukin-1 beta or prostaglandin E(2). NCX-1015 administered orally was also found to be equally active. Co-administration of the nitric oxide donors NOC-18 ((z)-1-[(2-aminoethyl)-N-(2-aminoethyl)amino] diazen-1-ium-1, 2-diolate; 7.9 micromol kg(-1)) or sodium nitroprusside (13.8 micromol kg(-1)) with prednisolone resulted in an additive anti-migratory action. 4. In a chronic model of granulomatous tissue inflammation, administration of NCX-1015 (13.9 micromol kg(-1)) from day 1 (i.e. after induction of inflammation) was more effective than prednisolone in reducing the granuloma dry weight, and this was associated to a lower anti-angiogenic effect. 5. In conclusion we show that NCX-1015 is more potent than prednisolone in controlling several, though not all, parameters of acute and chronic inflammation, and propose that this effect may be due to a co-operation between the steroid moiety and nitric oxide or related species released in biological fluids. Whereas this aspect needs to be further clarified, we propose NCX-1015 as the first member of a novel class of anti-inflammatory compounds, the nitro-steroids.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Anti-Inflammatory Agents/pharmacology
- Antigens, CD
- Antigens, Differentiation, Myelomonocytic/biosynthesis
- Antigens, Differentiation, Myelomonocytic/drug effects
- Chronic Disease
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Female
- Granuloma/prevention & control
- Humans
- Inflammation/prevention & control
- Interleukin-1/metabolism
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Male
- Mice
- Nitric Oxide/metabolism
- Peritonitis/chemically induced
- Peritonitis/prevention & control
- Prednisolone/analogs & derivatives
- Prednisolone/metabolism
- Prednisolone/pharmacology
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/drug effects
- Treatment Outcome
- Zymosan/adverse effects
Collapse
Affiliation(s)
- Mark Paul-Clark
- Department of Biochemical Pharmacology, St Bartholomew's and the Royal London School of Medicine and Dentistry, London
| | | | - Stefano Fiorucci
- Dipartimento di Medicina Clinica e Sperimentale Clinica di Gastroenterologia ed Endoscopia Digestiva, Universitá degli Studi di Perugia, Italy
| | - Roderick J Flower
- Department of Biochemical Pharmacology, St Bartholomew's and the Royal London School of Medicine and Dentistry, London
| | - Mauro Perretti
- Department of Biochemical Pharmacology, St Bartholomew's and the Royal London School of Medicine and Dentistry, London
| |
Collapse
|
41
|
Fiorucci S, Santucci L, Cirino G, Mencarelli A, Familiari L, Soldato PD, Morelli A. IL-1 beta converting enzyme is a target for nitric oxide-releasing aspirin: new insights in the antiinflammatory mechanism of nitric oxide-releasing nonsteroidal antiinflammatory drugs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:5245-54. [PMID: 11046058 DOI: 10.4049/jimmunol.165.9.5245] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Caspase-1, the IL-1beta converting enzyme (ICE), is required for intracellular processing/maturation of IL-1beta and IL-18. NO releasing nonsteroidal antiinflammatory drugs (NSAIDs) are a new class of NSAID derivatives that spare the gastric mucosa. Here, we tested the hypothesis that NCX-4016, a NO-aspirin derivative, inhibits proinflammatory cytokine release from endotoxin (LPS)-challenged monocytes. Our results demonstrated that exposing LPS-stimulated human monocytes to NCX-4016 resulted in a 40-80% inhibition of IL-1beta, IL-8, IL-12, IL-18, IFN-gamma, and TNF-alpha release with an EC(50) of 10-20 microM for IL-1beta and IL-18. Incubating LPS-primed monocytes with NCX-4016 resulted in intracellular NO formation as assessed by measuring nitrite/nitrate, intracellular cGMP concentration, and intracellular NO formation. Exposing LPS-stimulated monocytes to aspirin or celecoxib caused a 90% inhibition of prostaglandin E(2) generation but had no effect on cytokine release. NCX-4016, similar to the NO donor S-nitroso-N-acetyl-D-L-penicillamine, inhibited caspase-1 activity with an EC(50) of approximately 20 microM. The inhibition of caspase-1 by NCX-4016 was reversible by the addition of DTT, which is consistent with S-nitrosylation as the mechanism of caspase-1 inhibition. NCX-4016, but not aspirin, prevented ICE activation as measured by assessing the release of ICE p20 subunit. IL-18 immunoneutralization resulted in a 60-80% reduction of IL-1beta, IL-8, IFN-gamma, and TNF-alpha release from LPS-stimulated monocytes. Taken together, these data indicate that incubating human monocytes with NCX-4016 causes intracellular NO formation and suppresses IL-1beta and IL-18 processing by inhibiting caspase-1 activity. Caspase-1 inhibition is a new, cycloxygenase-independent antiinflammatory mechanism of NO-aspirin.
Collapse
Affiliation(s)
- S Fiorucci
- Dipartimento di Medicina Clinica e Sperimentale, Clinica di Gastroenterologia ed Epatologia, Università degli Studi di Perugia.
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The gastroduodenal mucosa is a model system of defense with several structural levels and biologic strategies that are closely interrelated with each other to cope with the harmful ingredients of ingested food and the potentially deleterious effects of gastric acid and pepsin. Experimental and clinical research carried out during the review period added to the understanding of each component of the multiple mechanisms of gastroduodenal mucosal protection. In the first place, mucosal integrity is defended by the mucus gel barrier, the epithelial cell barrier, and the immune barrier. The properties of these barriers are maintained by adequate regulation of mucus production, bicarbonate secretion, mucosal microcirculation, and motor activity. These regulatory systems are alarmed by nociceptive neurons and the mucosal immune system which includes chemokine-secreting epithelial cells. The ultimate defense system is rapid repair of the injured mucosa under the control of several growth factors. Progressing insight into the network of mucosal defense not only will improve existing therapies of inflammation and ulceration but also will provide new leads for the management of functional diseases in the gastroduodenal region.
Collapse
Affiliation(s)
- P Holzer
- Department of Experimental and Clinical Pharmacology, University of Graz, Austria.
| |
Collapse
|
43
|
Wenk GL, McGann K, Mencarelli A, Hauss-Wegrzyniak B, Del Soldato P, Fiorucci S. Mechanisms to prevent the toxicity of chronic neuroinflammation on forebrain cholinergic neurons. Eur J Pharmacol 2000; 402:77-85. [PMID: 10940360 DOI: 10.1016/s0014-2999(00)00523-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Inflammatory processes may play an important role in the degeneration of basal forebrain cholinergic cells Alzheimer's disease. We infused the proinflammagen lipopolysaccharide into the basal forebrain of young rats and determined whether the chronic administration of two novel non-steroidal anti-inflammatory drugs or a pan-caspase synthesis inhibitor, z-Val-Ala-Asp(OMe)-fluoromethyl ketone (zVAD), could provide neuroprotection from the cytotoxic effects of the neuroinflammation. Chronic lipopolysaccharide infusions decreased choline acetyltransferase activity and increased the number of activated microglia within the basal forebrain region. The level of caspases 3, 8 and 9 was increased in ventral caudate/putamen. Non-steroidal anti-inflammatory drug therapy attenuated the toxicity of the inflammation upon cholinergic cells and reduced caspases 3, 8 and 9 activity in the caudate/putamen. zVAD treatment significantly decreased the levels of caspases 3, 8 and 9 but did not provide neuroprotection for the cholinergic neurons. These results suggest that prostaglandins contribute to the degeneration of forebrain cholinergic neurons in Alzheimer's disease.
Collapse
Affiliation(s)
- G L Wenk
- Arizona Research Laboratories, Division of Neural Systems, Memory and Aging, University of Arizona, 384 Life Sciences North Building, Tucson, AZ 85724, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
Nitric oxide (NO) is now recognized as an important modulator of an enormous number of physiological processes, ranging from blood pressure regulation to neuronal transmission to penile erectile function. In the gastrointestinal tract, NO also participates in many physiological and pathophysiological processes. In this review, we summarize the contribution made by NO to the ability of the gastrointestinal mucosa to resist injury induced by luminal toxins and to defend against microbial invasion. We also review some of the main features of NO chemistry and the potential of NO as a target for new drugs to treat gastrointestinal disorders.
Collapse
Affiliation(s)
- J L Wallace
- Mucosal Inflammation Research Group, University of Calgary, Calgary, Alberta, Canada.
| | | |
Collapse
|
45
|
Johal K, Hanson PJ. Opposite effects of flurbiprofen and the nitroxybutyl ester of flurbiprofen on apoptosis in cultured guinea-pig gastric mucous cells. Br J Pharmacol 2000; 130:811-8. [PMID: 10864887 PMCID: PMC1572138 DOI: 10.1038/sj.bjp.0703383] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The nitric oxide (NO)-donating nitroxybutyl ester of flurbiprofen (NO-flurbiprofen), shows reduced gastro-intestinal toxicity relative to flurbiprofen. NO may exert either pro- or anti-apoptotic effects, while non-steroidal anti-inflammatory drugs may induce apoptosis. The aim of the present work was therefore to compare the effects of flurbiprofen and NO-flurbiprofen on apoptosis in guinea-pig gastric mucous cells. Apoptotic activity was assessed by assay of caspase activity and from the fragmentation and condensation of nuclei. Incubation with flurbiprofen for 24 h produced a concentration-dependent induction of apoptosis in cells attached to the culture plate (caspase 3-like activity increased by 257% at 500 microM), while NO-flurbiprofen inhibited basal apoptosis (caspase 3-like activity decreased by 71% at 500 microM). Caspase activity and nuclear fragmentation were substantially increased in cells that had spontaneously detached from the culture plate. NO-flurbiprofen inhibited caspase activity (55% at 500 microM) but not nuclear fragmentation in these detached cells. NO flurbiprofen inhibited the activation of apoptosis by 25 microM C(6)-ceramide in cells attached to the culture plate. Inhibition of caspase activity by NO-flurbiprofen was detectable after 6 h of incubation with intact cells, but by contrast with the NO-donor S-nitrosyl-N-acetyl-penicillamine, was not demonstrable with cell homogenates. Activation of caspase 3-like activity by flurbiprofen was slow (>6 h incubation needed) and was inhibited by cycloheximide. The presence of a nitroxybutyl ester moiety on flurbiprofen prevents the pro-apoptotic activity of the parent compound and may contribute to the reduced gastro-intestinal toxicity of NO-flurbiprofen.
Collapse
Affiliation(s)
- Kamaljit Johal
- Pharmaceutical Sciences Institute, Aston University, Aston Triangle, Birmingham B4 7ET
| | - Peter J Hanson
- Pharmaceutical Sciences Institute, Aston University, Aston Triangle, Birmingham B4 7ET
- Author for correspondence: P.J.
| |
Collapse
|
46
|
Abstract
The ability to elucidate the molecular mechanisms that modulate programmed cell death (PCD) may provide the crucial clues to unravel the cellular basis of neurodegenerative disorders. Employing both a novel assay to follow serially PCD in individual living neurons and the neuroprotective agent lubeluzole as an investigative tool, we examined the development of nitric oxide (NO)-induced PCD over time through the reversible annexin V labelling of membrane phosphatidylserine (PS) exposure and the electron microscopy of genomic DNA in primary rat hippocampal neurons. Exposure to the NO generators SNP (300 microM) or NOC-9 (300 microM) alone increased annexin V-positive neurons in the population from 7% +/- 4% in untreated cultures to 13% +/- 4% at 1 hr and to 61% +/- 5% at 24 hr. Administration of a neuroprotective concentration of lubeluzole (750 nM) at the time of NO exposure initially prevented the exposure of PS residues, but consistently maintained DNA integrity over a 24 hr period. During posttreatment paradigms of lubeluzole (750 nM) at 2, 4, and 6 hr following NO exposure, progression of membrane PS inversion was reversed and subsequently suppressed over a 24 hr course. Our work illustrates that neuronal PCD is composed of at least two physiologically distinct and separate pathways that consist of the externalization of membrane PS residues and the independent maintenance of genomic DNA integrity. In addition, neuronal injury is fluid and reversible in nature, suggesting a "window of opportunity" for the repair and reversal of neurons yet to be committed to PCD.
Collapse
Affiliation(s)
- K Maiese
- Departments of Neurology and Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | |
Collapse
|
47
|
Fiorucci S, Santucci L, Antonelli E, Distrutti E, Del Sero G, Morelli O, Romani L, Federici B, Del Soldato P, Morelli A. NO-aspirin protects from T cell-mediated liver injury by inhibiting caspase-dependent processing of Th1-like cytokines. Gastroenterology 2000; 118:404-21. [PMID: 10648469 DOI: 10.1016/s0016-5085(00)70223-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Concanavalin A (con A)-induced hepatitis is an immunomediated disease in which assembly of CD4(+) T cells and T helper (Th)1-like cytokines causes Fas-mediated liver cell death. Nitric oxide (NO) modulates Th1 response in vitro. NCX-4016 is an NO-aspirin derivative that spares the gastrointestinal tract and shares molecular targets with NO. The aim of this study was to investigate whether this NO-aspirin modulates Th1-like response induced by con A. METHODS BALB/c mice were injected with 0.3 mg con A per mouse alone or in combination with NO-aspirin (18-100 mg/kg) or aspirin (10-55 mg/kg). RESULTS NO-aspirin, but not aspirin, caused a dose-dependent protection against liver damage induced by con A. At a dose of 100 mg/kg, NO-aspirin caused a 40%-80% reduction of interleukin (IL)-1beta, IL-12, IL-18, interferon (IFN)-gamma, and tumor necrosis factor alpha production without affecting cytokine messenger RNA expression. NO-aspirin prevented Fas, Fas ligand, and IL-2 receptor up-regulation on spleen lymphocytes and Fas ligand on hepatocytes and caused the S-nitrosylation/inhibition of IL-1beta-converting enzyme-like cysteine proteases (caspases) involved in the processing and maturation of IL-1beta and IL-18. IL-18 immunoneutralization prevented IFN-gamma release and protected from liver injury induced by con A. In contrast to a selective caspase 1 inhibitor, zVAD.FMK, a pancaspase inhibitor, prevented IFN-gamma release and protected the liver from injury. CONCLUSIONS Th1-like response induced by con A is mediated by IL-18 and requires activation of multiple caspases. NCX-4016 causes the S-nitrosylation/inhibition of caspases involved in cytokine production. Inhibition of Th1-like response is a new anti-inflammatory mechanism of action of NO-aspirin.
Collapse
Affiliation(s)
- S Fiorucci
- Dipartimento di Medicina Clinica e Sperimentale, Clinica di Gastroenterologia ed Epatologia, Università degli Studi di Perugia, Perugia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
The importance of intracellular glutathione (GSH) in the pathology of disease, particularly cancer, has long been appreciated. However the ubiquitous nature of GSH has made it difficult to ascribe to a specific molecular mechanism in disease fulfillment. In addition, in all but a few cases, the underlying genetic regulation of the cellular redox state disrupted in disease has not been well described. Early identification of the importance of intracellular GSH to detoxification reactions has now led to investigating the potential importance that glutathione chemistry has on signal transduction and molecular regulation of cellular physiology. Here new relationships between the cellular redox state and the apoptotic regulatory protein BCL-2 will be described with emphasis on potential mechanisms by which GSH can alter cellular physiology in addition to its role in detoxification.
Collapse
Affiliation(s)
- D W Voehringer
- Department of Genetics, Stanford University Medical School, CA 94305-5318, USA.
| |
Collapse
|
49
|
Wallace JL, Vergnolle N, Muscará MN, Asfaha S, Chapman K, McKnight W, Del Soldato P, Morelli A, Fiorucci S. Enhanced anti-inflammatory effects of a nitric oxide-releasing derivative of mesalamine in rats. Gastroenterology 1999; 117:557-66. [PMID: 10464131 DOI: 10.1016/s0016-5085(99)70448-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Nitric oxide (NO)-releasing derivatives of cyclooxygenase inhibitors exhibit enhanced anti-inflammatory activity and greatly reduced gastrointestinal toxicity. We evaluated whether a similar derivatization of mesalamine (5-aminosalicylic acid) would improve its anti-inflammatory activity. METHODS Effects of an NO-releasing derivative of mesalamine (NCX-456; NO-mesalamine) were compared with those of mesalamine itself and 2 other NO donors in a rat model of colitis. These drugs were compared for their ability to inhibit leukocyte adherence to the vascular endothelium in vivo, interleukin (IL)-1beta and interferon (IFN)-gamma release in vitro (splenocytes and colon), and messenger RNA expression in the inflamed colon. RESULTS NO-mesalamine was significantly more effective than mesalamine in reducing the severity of colitis (damage and granulocyte infiltration). Unlike mesalamine, NO-mesalamine significantly suppressed leukocyte adherence to the vascular endothelium in vivo. NO-mesalamine inhibited IL-1beta and IFN-gamma release and caspase 1 activity in splenocytes; such effects were not found in the inflamed colon. CONCLUSIONS These studies show that an NO-releasing derivative of mesalamine has significantly enhanced anti-inflammatory activity, including improved efficacy in a rat model of colitis. The improved efficacy of this derivative is most likely caused by its enhanced ability to suppress leukocyte infiltration and possibly to scavenge peroxynitrite.
Collapse
Affiliation(s)
- J L Wallace
- Department of Pharmacology and Therapeutics, University of Calgary, Calgary, Alberta, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|