EMAN RESEARCH PUBLISHING | <p>Targeting inducible Nitric Oxide Synthase (iNOS) in the prevention of vascular damage and cardiac inflammation in CVD</p>
Inflammation Cancer Angiogenesis Biology and Therapeutics | Impact 0.1 (CiteScore) | Online ISSN  2207-872X
REVIEWS   (Open Access)

Targeting inducible Nitric Oxide Synthase (iNOS) in the prevention of vascular damage and cardiac inflammation in CVD

Nafees Uddin Chowdhurya, Abida Tishab, Juthika Sarkerb, Pulak Dev Nathb, Nowshin Ahmedb, Shahanshah Abdullahb, Tasdik Farooqb, Waich Mahmudb, Md. Mohabbulla Mohibb,c, Md. Abu Taher Sagorb

+ Author Affiliations

Journal of Angiotherapy 2 (1) 067-077 https://doi.org/10.25163/angiotherapy.1200032116160818

Submitted: 01 June 2018 Revised: 15 August 2018  Published: 16 August 2018 


Abstract

Vascular damage and subsequent cardiac failure due to immune cytokines is steadily increasing the morbidity of cardiovascular diseases (CVD), making them the leading cause of death globally. Nevertheless, improper diagnosis is an important hurdle in case management. Free radicals are one of major factors in producing vascular abnormalities in CVD. Additionally, some other factors such as high blood pressure, myocardial injuries, vascular damage, ventricular hypertrophy, cardiac fibrosis and myocardial necrosis are also the results of free radical-mediated abnormal cardiac activity/function. Of note, Nitric Oxide Synthase (iNOS) is produced in the body at an elevated rate in the presence of free radicals which has been shown to lead to cardiac issues. Furthermore, iNOS is also increased due to cytoplasmic stress, faulty DNA replication and membrane potential impairment. Moreover, abnormal cardiac rhythm has also been reported to be linked to the presence of iNOS. iNOS can also lead to abnormal cardiac function through inhibition of mitochondrial activity and production of pro-inflammatory cytokines. The molecular mechanism of iNOS that leads to CVD is comprehensively summarized in this review so as to aid in drug discovery strategies for the treatment of CVD.

Key words: iNOS, Immunity, Inflammation, Hypertension, vascular damage

Introduction

GO

Hepatic disabilities and renal insufficiency have often been linked with vascular dysfunctions. In addition, excess circulating fat accumulation may potentiate the development of arterial constriction. Of note, systemic hypertension seems to play a central role in myocardium damage and, as a result, several vascular diseases such as angina, myocardial infarction and congestive heart failure have been reported (Abu Taher Sagor; Mohib, Rabby, Paran, Hasan, Ahmed, Hasan, Sagor, Mohiuddin, et al., 2016; Md. Abu Taher Sagor, Nabila Tabassum, Md. Abdullah Potol, & Md. Ashraful Alam, 2015). Taken together, cardiac insufficiencies and vascular dysfunctions have been the leading cause of death globally. According to the Department of Health and Human Services (DHHS), roughly 596,577 people have died in 2011-14 due to heart diseases in the USA only. Although a massive development in medical sector has achieved in the recent times, the death rate is still high to minimize. It is also reported that only 3% mortality rate has been increased in 2010’s while compared to the 1980’s (Heron & Anderson, 2016).

Inducible Nitric Oxide Synthase (iNOS), is a critical enzyme involved in cellular growth, migration and fibrosis. It is also implicated in inflammation, which uncontrolled, can lead to cardiac sepsis, dilated cardiomyopathy, myocarditis, ischemia, uncontrolled hypertension and transplant rejection (Alam, Chowdhury, Jain, Sagor, & Reza, 2015; Mungrue et al., 2002b). Risk factors such as arterial fat deposition, smoking, chronic alcoholism, and genetic predispositions are considered to be inducers of iNOS, thereby correlating them with cardiac disabilities (Crump, Sundquist, Winkleby, & Sundquist, 2017; Eckel, 1997). Multiple studies have suggested that iNOS-mediated pathways play the central role in cardiac cell dysfunction. In animal models, overexpression of iNOS in the myocardium leads to peroxynitrite over-production which further progresses edema, increases heart volume and can lead to heart block and sudden death (Mungrue et al., 2002b; Md Abu Taher Sagor, Nabila Tabassum, Md Abdullah Potol, & Md Ashraful Alam, 2015). However, a contra indicatory experiment showed that many cardiac damages in the animal model were not associated with iNOS expression (Heger et al., 2002). Along with the animal models, various clinical investigations have linked iNOS to heart failure (Haywood et al., 1996). Some studies have suggested that iNOS or NOS2 may act as a longer-lasting nitric oxide, thus indicating cytotoxic abilities, which can lead to reduced myocyte contractility and eventual cardiac death (P. Alam et al., 2018; Vejlstrup et al., 1998). On top of this, iNOS is currently being considered as potent inflammatory cytokine that is often associated with a series of phenomenon that ultimately stop the heartbeat (Csont et al., 2005; Mungrue et al., 2002a). In addition to these, several other factors such as transforming growth factor-ß, plasminogen activator factor, platelet-derived growth factor, vascular endothelial growth factor, tumor necrosis factor-a, monocyte chemoattractant protein-1 (MCP), mast cell accumulation, C-reactive protein and lipoprotein-associated phospholipase A2 also contribute to myocyte damage and can lead to cardiac mortality (Ballantyne & Nambi, 2005; Eggers, Oldgren, Nordenskjöld, & Lindahl, 2004; Fukumura et al., 2001). Furthermore, higher Aangiotensin production, over-expression of Xanthine Oxidase (XO), Uric Acid (UA), Creatine kinase, Adiponectin, Creatine Kinase-MB and a-Troponin have also been linked to iNOS signaling pathways (Mogensen et al., 2004). As iNOS generally acts as a free radical, many studies have suggested that other free radicals like NOX-4, malondialdehyde, superoxide, peroxide, singlet oxide and peroxynitrite may be correlated with iNOS while interacting with each other, which could be more damageable for the heart  (Heymes et al., 2003; Mohib, Rabby, Paran, Hasan, Ahmed, Hasan, Sagor, & Mohiuddin, 2016; Md Abu Taher Sagor et al., 2015).

iNOS expression was observed in several target areas including hepatocytes, macrophages, neutrophils, the colonic epithelium, chondrocytes, Kupffer cells, the pulmonary epithelium, the vasculature, and in diverse neoplastic diseases  (Asano et al., 1994; Charles et al., 1993; MacMicking, Xie, & Nathan, 1997). A study suggested that preventing iNOS may enhance the activity of ß-adrenergic inotropic responsiveness in an animal model (Funakoshi, Kubota, Kawamura, et al., 2002). Similar studies also showed that blocking iNOS by using ONO-1714 improved ventricular performance and ß-adrenergic inotropic responsiveness to tumor necrosis factor (TNF)-athus developing dilated cardiomyopathy with myocardial inflammation in Transgenic Mice (Funakoshi, Kubota, Machida, et al., 2002). However, another study suggested that the iNOS/NO/cGMP pathway is more aggressive in senior subjects compared to the younger, thus suggesting that preventing iNOS could be an alternative strategy to protect the cardio-vascular system (Yang, Larson, & Watson, 2004). The study also suggested that the acetylation of SIRT by iNOS may induce NF-?B and thereby triggers unnecessary immune response. Along with stimulating inflammation, iNOS also prevents p53 gene and initiates apoptosis in normal functioning cells (Csiszar et al., 2009). Hence, a clear understanding of all the iNOS-related signaling pathways has yet to be established. Therefore, this study will attempt to explain some possible patho-physiology related to iNOS and give numerous treatment approach ideas that could prevent cardio vascular dysfunctions.

iNOS and its regulations

GO

Nitric oxide (NO) is an important component that works in both intracellular and extracellular pathways. At the same time, it monitors and maintains several signaling pathways for patho-physiological, pharmacological and physiological responses by regulating neuronal activities, cardiovascular responses, cytokine production, anti-microbial and anti-tumor activities (Abu Taher Sagor et al., 2016; Aktan, 2004; A. T. Sagor et al., 2015). In the living system, NO often plays an essential role in during differentiation process especially on sustaining and surviving conditions (Kengatharan, De Kimpe, Robson, Foster, & Thiemermann, 1998; J. D. MacMicking et al., 1997). The production of NO is very complex and is maintained and regulated by proteins of the Nitric oxide synthase (NOS) family.  Nitric Oxide however, was found to have a very short half-life and is therefore produced as needed. For example, organs wherein eNOS is produced use it for vasodilatation  (M. R. H. Chowdhury et al., 2015). nNOS, on the other hand, produced in neuronal cells, helps in cell communication. (Reza, Sagor, & Alam, 2015). In addition, iNOS, is generally expressed in the heart and immune cells where it has been linked to pathogenesis progression.  bNOS is mainly expressed in bacterial cells and plays several roles antibiotic resistance, immunity and oxidative stress.  (Alderton, Cooper, & Knowles, 2001). On top of these, Osteoarthritic NOS has controversially gained some attentions and created some interesting contents though it correlates with iNOS and interleukin (IL)-1 (Charles et al., 1993). Another groundbreaking discovery with Guinea-pig iNOS has drawn some interesting correlations in cell and protein cloning for novel discoveries. Guinea-pig iNOS was further correlated with nNOS and bNOS; while others correlated it with   iNOS. Hence, correlations have yet to be made clear (Daff, Sagami, & Shimizu, 1999; Salerno et al., 1997).

On the other hand, nitric oxide is a gas in nature and a very small compound that can freely diffuse into cells wherever it may be needed.  The formation and biosynthesis of this product is quite complex and regulated through several factor and co-factors but it has been shown to be biosynthesized from L-arginine. To complete the whole, oxygen is used with NADPH as electron donor, a Heme molecule, FAD, tetrahydrobiopterin (H4B) and FMN. The entire process uses other cofactors and five electrons (Alderton et al., 2001). This process is conducted in two phases i) oxidative exchange of L-arginine to nitric oxide and ii) L-citrulline via N-hydroxy -L-arginine (NOHarginine) which is considered the intermediate product that has been shown to interfere with monooxygenase I and monooxygenase II as a mixed-function oxidation(Mayer & Hemmens, 1997).

Among the all other NOS genes, iNOS is the most studied as it negatively impacts human health. Since the last three decades, the structure, molecular docking and functions have been investigated to explore new avenues related to disease pathology or physiological function. The iNOS gene is situated on chromosome 17. Mouse and Human iNOS are 80% similar, while that between eNOS and iNOS is51% and nNOS and iNOS is 53% (Taylor & Geller, 2000; Xu et al., 1994). Despite of its Ca2+ independent functions, the  iNOS complex cannon-covalently bind Calmodulin and thus representing a necessary subunit of the isoform (Cho et al., 1992). iNOS genes are highly transcribed following several stimulations such as TNF-a, interleukin-1ß, interferon-? and nuclear factor-?B (Lowenstein & Padalko, 2004; Müller-Werdan et al., 1997). Besides this, it can also be triggered by bacterial toxins such as the LPS-MyDD-88 pathway (Nathan, 1992). On top of that, the iNOS gene also contains a special region for binding under hypoxic conditions thereby interacts with phorbol esters and lipoarabinomannan to translate various regulatory proteins (Chan, Chan, & Schluger, 2001; Ferreiro et al., 2001). Other important signaling regulator such as Janus Tyrosine Kinase (JAK) and Signal Transducers and Activators of Transcription (STAT) play significant roles in iNOS gene expression (Xuan, Guo, Han, Zhu, & Bolli, 2001). iNOS gene regulation and transcription are highly dependent on the stability of iNOS mRNA stability and potent catalytic activity. Moreover, NF-?B, Activator Protein 1 (AP1), Activating Transcription Factor 2 (ATF2), cAMP-responsive elements, and other transcription factors may participate in regulating the transcription of the iNOS gene (Janssen-Heininger, Macara, & Mossman, 1999; Lechner, Lirk, & Rieder, 2005).

angiotherapy-1200032116160818 (1)

Figure 1: Multiple signaling pathways are responsible for the biosynthesis of iNOS. L-Arginin plays the prime role to initiates the synthesis, L-Arginine eventually releases NO-free radicals and L-Citrulline into the cell. On the other hand, activation of endothelin receptors causes iNOS gene expression by activating NF-?B and p38. Stimulation of Toll-Like receptor (TLR)-4 and interferon-? receptor (INFGR) also participates in the expression of iNOS gene.

Hypertension and Vascular Dysfunctions

GO

Hypertension is often defined as a slow killing process to humans (Md Abu Taher Sagor et al., 2015). It is very common in the older population, but recently it is has been reported in the middle aged population as well (Abu Taher Sagor et al., 2016; Alam et al., 2015). The exact reason for developing hypertension is yet to be unraveled but has been suggested that multiple factors such as smoking, diet, obesity and low energy expenditure are the main culprits (Mohib, Rabby, Paran, Hasan, Ahmed, Hasan, Sagor, & Mohiuddin, 2016; Sagor, Mohib, Tabassum, Ahmed, & Reza, 2016). Also, co-morbid conditions such as diabetes and pre-genetic dispositions are also correlated with hypertension (N. U. Chowdhury et al., 2016; Mohib et al., 2017). In cardiac cells, higher levels of sodium and calcium lead to hypertension. Similarly, abnormal levels of potassium and magnesium also play definitive roles in hypertension (Loh, Giribabu, & Salleh, 2017; G. Yu, Cheng, Wang, Zhao, & Liu, 2017). Inside the heart, damage to small vessels and constriction of arteries, unwanted inflammatory cell migration, excess uric acid and Ang-II and diseases like atherosclerosis may eventually lead to hypertension  (AT Sagor et al., 2016; Mohib, Hasan, et al., 2016; Md Abu Taher Sagor et al., 2015). Moreover, less endothelial nitric oxide production can also be the major contributor in the development of arterial pressure (Reza et al., 2016; Shabeeh et al., 2017). Left untreated, uncontrolled blood pressure can cause several macrovascular and microvascular diseases like nephropathy, retinopathy, stroke and heart attack by blocking small vessels (Sagor et al., 2016). Several correlations have been drawn so far to connect the possible mechanism for hypertension in response to iNOS. In recent literature, two major possibilities have been tied to increased vasoreactivity and thrombogenesis i) faulty action of peroxynitrite-mediated endothelial NO release, thereby impairing normal vasoreactivity (Seccombe, Pearson, & Schaff, 1994) and ii) superoxide stimulate platelet adhesion and aggregation binding with peroxynitrite interactions (Salvemini, Nucci, Sneddon, & Vane, 1989; Seccombe et al., 1994). These two possible theories make change in thrombogenesis and vasoreactivity thus progressing the aggregation of unstable plaque.This leads to  iNOS being blamed for thrombus, uncontrolled hypertension and chest pain by increasing vascular hyper-reactivity (Depré, Wijns, Robert, Renkin, & Havaux, 1997).

Effect of iNOS on vascular damage

GO

The heart is known as most vascular organ in the body and includes various arteries, vessels, aortas and veins. Blockage or stiffness in the vascular system may reduce blood flow which leads to uncontrolled hypertension leading the way for sudden cardiac failure. (Alam et al., 2015; Sagor, Reza, Tabassum, Rahman, & Alam, 2018). Endothelial cells play the most prominent role in the production of nitric oxide (NO) which relaxes arteries for smooth permeability. Besides this, several other factors such as vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), thromboxane and plasminogen activator factor (PAF) maintain and regulate normal blood flow (Figure 2). However, excess production of these growth factors may lead to angiogenesis and even cancers (Zhang, Peng, & Chen, 2005). Of note, VEGFR-1 and VEGFR-2 are very selective to stimulate endothelial cells. The Angiogenic properties of VEGF in human endothelial cells have been associated with nitric oxide production that further leads to angiogenesis (Papapetropoulos, García-Cardeña, Madri, & Sessa, 1997). In addition, VEGF can influence endothelial cells to produce NO by iNOS signaling (Fukumura et al., 2001). On the other hand, chronic calcification and non-calcified atherosclerotic plaque may cause luminal narrowing in the proximal Right Carotid Artery (RCA) and Left Carotid Artery (LCA) that might further block total cardiac functions. Several animal studies and clinical investigations have been linked many correlations among iNOS, vascular growth and atherosclerosis (M. Xia, Ling, Ma, Kitts, & Zawistowski, 2003; X. Xia et al., 2006). Furthermore, iNOS, is being connected with several cardiac inflammatory markers such as Interleukins, Troponin  and Creatinin kinase which are further responsible for the formation of atherosclerotic plaque. Similarly, Macrophages and Foam cells in atherosclerotic plaque may express the inducible isoform of NO synthase (iNOS) which complicates matters as the formation of peroxynitrite from NO can favor vasospasm, stiffness and thrombogenesis (Depre, Havaux, Renkin, Vanoverschelde, & Wijns, 1999; Luoma et al., 1998). On the contrary, it is of notethat NO production by EcNOS impedes against platelet aggregation, leukocyte adhesion and vasoconstriction. However, higher NO production in response to iNOS can positively affect plaque stability through different potential mechanisms thereby triggering numerous vascular dysfunctions  (Wever, Lüscher, Cosentino, & Rabelink, 1998). Other vascular system such as the retina can be affected by iNOS leading to permanent vision loss (D.-H. Lee, Cho, Kim, Choi, & Joo, 2001). iNOS has also been correlated with acute migraine inhuman subjects (Palmer et al., 2009).

angiotherapy-1200032116160818 (2)

Figure 2: Nitric Oxide, a free radical processed by iNOS that activates the expression of Cyclooxygenase (Cox) and c-fos transcription factor. It also participates in the activation of collagen and production of inflammatory cytokines which further initiates plaque formation and synthesis of Xanthine oxidase (XA) to produce uric acid.

Role of iNOS on other organs

GO

The damaging effects of iNOS are not only observed in the vascular systems but it is also found in the intestine (Wong et al., 1996), kidney (Gupta et al., 2007), liver (M. A. Alam et al., 2018), pancreas (Shimabukuro, Ohneda, Lee, & Unger, 1997), brain (Wong et al., 1996), adipose tissues (Kapur, Marcotte, & Marette, 1999), lung (Sarady et al., 2004) and smooth muscles (Kibbe et al., 2000). Several mechanisms and destructive pathways have been explained so far depending on the iNOS nature. In the lung, iNOS has been linked with inceased production of prostaglandins E2 and NO that could further play a major role in the lung’s inflammatory or fibrogenic response (J. K. Lee et al., 2012). In the smooth muscles, iNOS upregulates p21 and blocks vascular smooth muscle cell proliferation via p42/44 MAPK activation independently of p53 (Kibbe et al., 2000). iNOS expression in the adipose tissue has been shown to be elevated in adipocytes as they act as artificial production houses of iNOS. The mRNA of iNOS has detectedin both white and brown adipocytes and has been further linked with TNF-a and INF-? (Kapur et al., 1999). iNOS expression in the kidney has been investigated, especially in diabetic and fibrosis subjects. Animal model showed higher iNOS expression in diabetic kidneys and that its expression corelates with a-SMA, collagen I, fibronectin, TGFß-1, NF-?B, apoptosis and DNA damage (S. Khan & Jena, 2014). Liver fibrosis has been a bigger target due to having a very complex patho-physiology involvement (Fathy, Khalifa, & Fawzy, 2018). iNOS plays a vital role in the production and activation of kuffer cells, which are responsible for extracellular matrix, collagen, MMPs and a-SMA production (Anavi et al., 2015; Rodrigues et al., 2017). The NOS2 gene encodes iNOS synthesis and often contributes to antimicrobial and antipathogenic activities. Excess production of iNOS may accelerate in inflammatory bowel disease (IBD) and NOS2 may be implicated in a specific subset of IBD patients with severe and/or extensive colitis (Dhillon et al., 2014). In the brain, iNOS has been correlatedwith MPO, neutrophil accumulation, ROS generation and NF-?B signalling due to excess peroxynitrite production (Abdelsalam & Safar, 2015). In the pancreas, iNOS often induces free radical,inflammatory cytokines and killer cells which ultimately destroys pancreatic ß-cells, resulting in type-1 diabetes mellitus. Study have shown a possible correlation between the iNOS-2087A>G polymorphism and chronic pancreatitis, which further links it with human diabetes (Padureanu et al., 2017). Furthermore, iNOS has also been implicatedwith several types of cancers (Vannini, Kashfi, & Nath, 2015). Several relationships have been drawn between iNOS and cancer biology.Of those,most of them were identified with inflammatory pathways (Ying & Hofseth, 2007). Selective iNOS inhibitors have proven to be effective cancer markers and may help in the progression stages of various abnormal cell replication (Janakiram & Rao, 2012).

Table 1: Role of iNOS in the cardiovascular dysfunctions on various models

Subjects

Outcomes of the study

References

Model: Mouse Ventricular Myocytes

Induced By: Genetic

Target: Nitric Oxide Signaling

Cardiac necrosis and apoptosis were reported.

(Das, Xi, & Kukreja, 2005)

Model: Rats

Induced By: Post-ischemic myocardial surgery

Target: iNOS –VEGF –KDR – eNOS signaling

Superior post-ischemic ventricular damages, increased myocardial infarct size and cardiac apoptosis were found, and Vascular endothelial growth factor noticed higher.

(S. Das et al., 2005)

Model: Vascular Smooth Muscle Cells

Induced By:IL-1a and TNF-a

Target: Natriuretic peptides in vascular remodeling

Vascular remodeling was noticed due to iNOS signaling.

(Marumo et al., 1995)

Model: Male SD rats

Induced By: Worm Ischemic Precondition

Target: iNOS and eNOS

Endothelial dysfunction and ischemia/reperfusion injury were observed.

(Wang, Fang, Stepheson, Khiabani, & Zamboni, 2004)

Model: Rats

Induced By: Endotoxemia

Target: MEK1/ERK1/2/iNOS/sGC/PKG pathway

Hypotension and vascular hyporeactivity were noticed.

(Korkmaz et al., 2011)

Model: Wistar rats

Induced By: Hyperbaric oxygen mitigates ischemia-reperfusion

Target: NOS signaling

Ischemia-reperfusion injuries along with higher NOS were determined.

 

(Baynosa et al., 2013)

Model: Mice

Induced By: High fat diet

Target: Atherosclerotic Plaque

Pro-inflammatory cytokines such as tumor necrosis factor-a, arginase-2, Interleukin (IL)-1ß, IL-6 and Interferon-? noticed to be increased.

(Simsekyilmaz et al., 2013)

Model: RAW264.7 macrophages

Induced By: Already Induced

Target: NF-?B and AP-1

NF-?B, AP-1 activation and related inflammatory cytokines found to be increased.

(Ryu et al., 2013)

Model: OLP oral tissues samples

Induced By: Induced

Target: VEGF and iNOS

-Expression of VEGF was observed higher in all epithelial layers,

-Angiogenesis and inflammatory cell infiltrates noticed, and

-Number of CD68+ cells was significantly increased.

(Metwaly, Ebrahem, & Saku, 2014)

Model: Vascular Smooth Muscle Cells

Induced By: Genetic

Target: Inflammatory cytokines and HO-1 pathways

-An increased number of harmful cytokine was reported, and

-Decreased HO-1 was noticed.

(X.-m. Liu, Peyton, Wang, & Durante, 2012)

Model: Spontaneously Hypertensive Rats

Induced By: Genetic

Target: Arterial natriuretic peptide and arterial pressure

-Heart, valve and aortic eNOS activity decreased, and

-Abnormal BP was evaluated.

(Costa et al., 2010)

Model: Human

Induced By: Acute Global I/R during Cardiac bypass Surgery

Target: RNase1

-Cardiac markers were found to be increased, and

-Inflammatory cytokines production (TNF-a) noticed higher.

(Cabrera-Fuentes et al., 2014)

Model: RAW264.7 cells

Induced By: LPS-induced

Target: iNOS and COX-2

-Activation of ERK1/2 signaling, Expression of NF-?B p65 subunit, and up-regulated I?B kinase were found owing to iNOS.

(Jin et al., 2010)

Model: Sprague-Dawley rats

Induced By: Unilateral Ureteric Obstruction

Target: Human gene-2 (H2) relaxin, iNOS and Fibrosis

-Increased TGF-ß, Smad axis and collagen were found higher, and

-Matrix Metalloproteinase (MMP) noticed to be induced.

(Chow et al., 2012)

Model: Rats

Induced By: Ischemia Reperfusion Surgery

Target: Eicosapentaenoic acid (EPA) and Myocardial Infarction

-Phosphoinositide 3-kinase, caspase-3 dependent apoptosis, and calcium dependent iNOS activity found to be increased, and

-Myocardial infarct size was increased.

(Keyes et al., 2010)

Effect of iNOS on cardiac inflammation

GO

Cardiac inflammation is one of the major reasons for myocardial injuries and atherosclerosis, which often correlate with cardiac failure (Md Abu Taher Sagor et al., 2015). Several explanations have been put forward as to the exact the reason for cardiac injuries in western diets compared to others. A study reported that western diets can produce more advanced glycation end products (AGE), which interact with receptors for advanced glycation end products (RAGE), which in turn induce cardiac inflammation (Tikellis et al., 2008). Recent reports suggested that cardiac inflammation eventually alters the extracellular matrix in heart failure patients and that phages concurrently contribute to the Heart Failure with Normal Ejection Fraction (HFNEF) rate, ultimately increasing mortality (Westermann et al., 2010). Monocyte Chemoattractant Protein-1 (mcp-1) and its receptor CCR2, on the other hand, were found to be significantly responsible for vascular remodeling and inflammation in hypertension-suffering subjects (Ishibashi et al., 2004). The mRNA of pro-inflammatory cytokines and iNOS have generally been founded simultaneously which makes iNOS as a potent inflammatory marker during an investigation. A population-based study reported that  patients with dilated cardiomyopathy showed the presence of iNOS and TNF-a mRNA in their  cardiomyocytes (Satoh et al., 1997).

Treatment strategies against iNOS

GO

As cardio-vascular diseases become an ever increasing concern, the pressure on healthcare professionals to discover novel molecules to treat these issues is elevating.  Recent research is trying to focus on plant-based therapies as they are considered quite safe and rarely show any adverse drug reaction (ADR).For instance,  Terminalia arjuna bark can reduce multiple cardiac symptoms, complexities and often improve vascular activities (Dwivedi, 2007). On the contrary, several studies suggest that complementary therapies do not have any specific dose or dosage forms, and similarly, some researchers believe that using the whole plant, rather than a purified active compound, is unnecessary (Rasoanaivo, Wright, Willcox, & Gilbert, 2011). As iNOS is an enzyme, natural therapies must ensure to mitigate any possible secondary protein interactions. Several plant-based therapies have been investigated in the laboratory and found to have suppressive activities against iNOS expression. Indeed, umbelliferone 6-carboxylic acid isolated from Angelica decursiva showed inhibitory activity against iNOS and nitric oxide production in lip polysaccharide (LPS)-stimulated raw 264.7 macrophages (Islam et al., 2012). In the laboratory, macrophage or related cell culture has been extremely popular for mimicking and establishing iNOS expression and or inhibition. A study conducted with pomegranate (Punica granatum L.) mesocarp extract found suppressed activity of iNOS expression in the treated adipocytes. Along with the iNOS inhibition, the study also suggested IL-6 and NF-?B inhibition (Ramlagan et al., 2017). On the other hand, several studies reported that aggregation of inflammatory and pro-inflammatory cytokines interferes with normal cardiac function and disturbs vascular activity. An investigation into this phenomena noted suppressed iNOS activity and COX-2 mRNA production when LPS induced RAW264.7 were treated with Litsea japonica fruit-isolated litsenolide B2.Protein investigation also found NF-?B and MAPK pathways inhibition (Ham et al., 2015). Terminalia arjuna is very familiar all over the world and was shown to be very beneficial in several cardiac diseases including coronary arterial dysfunctions (Kapoor, Vijayvergiya, & Dhawan, 2014). Terminalia arjuna bark contains a new triterpene glycoside which is known as Terminoside which was found to be very protective against murine macrophages by blocking NO and iNOS pathways (Ali et al., 2003). Similarly, patho-physiology of atherosclerosis and neointima formation especially after angioplasty are associated with aggregation and proliferation of inflammatory cytokines which stimulated by macrophages and vascular smooth muscle cells in the neointima. Terminalia chebula is a very well-known traditional herb containing several phyto-nutrients including that was shown to suppress1,1-diphenyl-2-picryl hydrazyl (DPPH) scavenging activity and lipid peroxidation breakdown. This plant was also found to be very effective when applied to RAW 264.7 cells and showed inhibitory activity towards iNOS and NO production and platelet-derived growth factor (PDGF-BB) induced VSMC migration (H.-H. Lee, Paudel, & Kim, 2015). However, cardiac functions largely depend on many electrolytes such as sodium, potassium, magnesium and most importantly calcium. Abnormalities in the ions may negatively affect cardiac function. On the other hand, sarcoplasmic reticulum serves the most prominent role in storing calcium ions and disturbances in sarcoplasmic reticulum may release higher calcium thus increasing cardiac activity and fatiguing the heart within a shorter period. It was previously reported that IL-6 decreases contractility and increases inducible NOS in the heart of mice. Following that investigation; the same authors conducted another study on L-type Ca+ channel and noticed a negative inotropy role of iNOS on chronic IL-6 exposure in the adult ventricular myocytes. The investigation finally concluded that activation of iNOS mediates IL-6-induced inhibition of SR function. Hence, preventing iNOS can be an excellent way to protect cardiovascular system (X. W. Yu, Chen, Kennedy, & Liu, 2005).

Endothelial cells directly from animals or Raw endothelial cells are both popular cells that respond to iNOS production. As iNOS-mediated endothelial dysfunction has been highly correlated with the cardiovascular system. With this in mind, preventing or blocking iNOS or NO would be a good approach to protect from heart diseases. Several studies have explained that natural polyphenols-based treatment strategies can block iNOS expression, thereby preventing excess NO production. A study found that Arthrospira platensis  is a very potential source of natural polyphenol when applied on endothelial cells, as its  polyphenolic property significantly prevent the expression of endothelial Nitric Oxide Synthase, MMP-9,  iNOS  and Vascular Cell Adhesion Molecule 1 on TNFa-induced endothelial activation (Ferrari et al., 2015).

The heart is the most sophisticated organ in the entire body and any halting of cardiac activity due to however small the problem can have dramatic impacts on the body. It is thus primordial to take good care of the cardiovascular system to keep the other organs running optimally. iNOS, on the other hand, is an enzyme which has a very short biological half-life, meaning that targeting this molecule must be very specific as unwanted protein interaction may occur. As iNOS also interferes in other biochemical pathways, which further trigger other harmful downstream signaling pathways, proper blocking of iNOS might be very beneficial to prevent other diseases. In addition, iNOS also triggers free radical-mediated oxidative stress which damages cardiomyocytes. Researchers are working very hard to discover and design new molecules to inhibit the iNOS pathway.  A study suggested that Glucagon-like peptide 1 protects microvascular endothelial cells by interfering with intracellular signaling pathway that activates apoptosis. The study further explained that inactivating the PARP-1/iNOS/NO pathway by treating with Glucagon-like peptide 1 may restore islet microvascular endothelial cells (IMECs) (F.-q. Liu et al., 2011). In these studies, over expression of iNOS in animal cardiomyocytes triggers peroxynitrite generation, heart block, and sudden death (Mungrue et al., 2002a), further solidifying that need for preventing iNOS to counter cardiac dysfunctions. Expression of COX-2 mRNA is currently being interrelated with the production of iNOS in macrophages and epithelial cells as these mRNAs serve almost the same function. A study found that preventing COX-2 and iNOS by a-tocopherol can inhibit PGE2 production, resulting in protected vascular function (Jiang, Elson-Schwab, Courtemanche, & Ames, 2000).

Hypertension, on the other hand, has been highly correlated with activated iNOS in the systemic circulation, so preventing or blocking iNOS can play an important role in countering arterial pressure. A clinical trial with 25 subjects, 14 of which were hypertensive subjects given 4 mg/day of a Lacidipine treatment for six months found that this treatment greatly helped hypertensive subjects. The treatment controlled systemic blood pressure, decreased free radical production, reduced the oxidative damage to the endothelium and decrease iNOS in macrophages without interfering with NO level (Escames, Khaldy, León, González, & Acuña-Castroviejo, 2004). Nitric Oxide is essential for endothelial cells to relax the arteries, hence, absence of iNOS functions less oxidative stress and related-damages (Sun et al., 2005).

Table 2: The role of plant extracts against Cardiac and vascular iNOS on various animal model

Subjects

Outcomes of the study

References

Model: Male ICR mice, RAW 264.7 murine macrophages

Wt/Age of model: 25-30g

Plant extract: Angelica decursiva

Dose: 50, 100, 200 µg/ml

- Pretreatment with umbelliferone 6-carboxylic acid isolated from : Angelica decursiva down-regulated the expression of nitric oxide synthase and inhibited the production of nitric oxide

 

(Islam et al., 2012)

Model: 3T3-L1 pre-adipocyte

Plant extract: Punica granatum L.

Dose:0-15.6 µg/ml

-Pomegranate mesocarp extract from Punica granatum L down-regulated the over expression of iNOS.

(Ramlagan et al., 2017)

Model: RINm5F rat pancreatic ß-cell line

Plant extract: Rheum palmatum

Dose:15, 30 µg

-Emodin from Rheum palmatum attenuated iNOS mRNA and protein levels.

(Bae et al., 2015)

Model:EAhy926 endothelial cells

Plant extract: Arthrospira platensis

Dose: 0.05, .10mg/ml

-Both polyphenol from A. platensis and from UV-stressed A platensis down-regulated the expression of iNOS in a dose dependent manner.

(Ferrari et al., 2015)

Model: Murine macrophage cell line RAW 264.7

Plant extract: Litsea japonica

Dose: 2.5, 5 , 10 µM

-Litsenolide extracted from Litsea japonica down-regulated iNOS expression and suppressed nitric oxide release on the macrophage cell line.

 

(Ham et al., 2015)

Model: RAW 264.7 Murine macrophages

Plant extract: Saccharina japonica

Dose: Pheophorbide A 0.125–1.0 lM, pheophytin A 5–30 lM

-Pre-treatment with pheophorbide and pheophytina extracted from Saccharina japonica down-regulated the expression of iNOS protein in a dose dependent manner.

(Islam et al., 2013)

Model: RAW 264.7 murine macrophages

Plant extract: Eisenia bicyclis

Dose: 5, 10 µM

-Fucosterol from Eisenia bicyclis suppressed iNOS in lipopolysaccharide induced macrophage cell line and reduced nitric oxide production.

 

(Jung, Jin, Ahn, Lee, & Choi, 2013)

Model:PC12 cell line          

Plant extract: Rhodiolarosea L.

Dose: 10, 50, 100 µM

-Salidroside extracted from Rhodiolarosea L. counteracted the over-expression of iNOS protein and reduced the production of nitric oxide level.

(X. Li et al., 2011)

Model: BV-2 microglial cells

Plant extract: Acorus gramineus

Dose: ß-asarone: 10, 50, 100 µM

-ß-asarone from Acorus gramineus attenuated iNOS expression in LPS activated BV-2 microglia cells to exhibit anti-inflammatory effects.

(Lim et al.)

Model- hCB1/hCB2-CHO cells

Plant extract: Cannabis sativa

Dose: Tetrahydrocannabivarin: 1 µM

-Tetrahydrocannabivarin of Cannabis sativa attenuated the iNOS expression in the lipopolysaccharide induced peritoneal macrophage.

(Romano et al., 2016)

 

Table 3: The role of API or crude molecules against iNOS on several animal models

Subjects

Outcomes of the study

References

Model: Male ICR mice

Wt of model:28-30g

API used: Lactobacillus sakei K040706

Dose:108 CFU/day orally for12 days

-K040706 attenuated iNOS protein expression in the colon resulting in anti-inflammatory effects.

 

 

(Seo et al., 2017)

Model: Male Sprague Dawley rats

Wt of model: 220-240g

API used: 100 mg Lansoprazole/kg orally for 24 hours

-Lansoprazole reduced iNOS expression in the intestine of Indomethacin induced rats

(Yoda et al., 2010)

Model: T98G human Glioblastoma cell line             

API used: Thalidomide

Dose: 20 µM

-Thalidomide significantly reduced the overexpression of iNOS in T908G morphine dependent cell line.

(M. I. Khan et al., 2017)

Model: Adult male Wistar rats

Wt of model: 150-200g

API used: 9mg Oral Nimesulide/kg BW for 7 days

-Nimesulide reduced iNOS expression in the lung

(Khanduja, Sohi, Pathak, & Kaushik, 2006)

Model: Male C57BL/6 mice

Age of model: 6-8 weeks old

API used: 5, 20 and 50 mg oralLuteolin/kg for 18 days

-Lutelin protected mice from colitis through inhibition of iNOS

(Y. Li, Shen, & Luo, 2016)

Model: Female ICR mice

Wt of model: 25 g

API used: 10 and 100 mg oral Zerumbone /kg/BW for 8 days

-Dietary Zerumbone repressed iNOS expression.

(Chen et al., 2011)

Model: MS-1 cell line

API used: Glucagon like peptide-1

Dose: 100 nmol/L

-GLP-1 suppressed the expression of iNOS to inhibit the production of NO in microvascular endothelial cells.

(F.-q. Liu et al., 2011)

Model: Male adult Sprague-Dawley rat

Wt of model:150-200g

API used: Dolastatin 15, Celecoxib

Dose : Oral Dolastatin 15: 5 µg/day to each rat; Celecoxib: 6mg/kg BW for6 weeks               

-Dolastatin and Celecoxib inhibited iNOS by interacting with it directly.

(Piplani, Vaish, & Sanyal, 2012)

Model: Adult male ICR mice

Wt of model: 25-30 g

API used: 50 mg oral Tetrahydrocurcumin and Deferiprone kg/day for 8 weeks

-Tetrahydrocurcumin and Deferiprone inhibited iNOS expression to alleviate the deleterious cardiovascular effect of iron overload.                               

 

(Sangartit et al., 2016)

Model: 11 healthy subjects and 14 hypertensive patients

API used: 4mg oral Lacidipine/day for6 months

-Lacidipine reduced iNOS expression in the macrophage along with alleviation of blood pressure.

(Escames et al., 2004)

Conclusion

GO

Evidences suggest that heart dysfunctions are the prime reason for human mortality and morbidity. Studies also showed significant data on hypertension which has been the silent killer of mankind. Importantly, iNOS-induced cardiac dysfunctions are mainly through free radical-mediated myocardium injuries, pro-inflammatory and inflammatory-mediated destructions, vascular stiffness and arterial injuries. iNOS often leads to atherosclerosis as well, which further blocks the heart and eventually leads to heart failure. Several researchers now focus on targeting iNOS as a preventative measure against cardio-vascular diseases. Blocking iNOS and its mRNA proves to be highly beneficial in both animal and human models to decrease vascular damaging events. Targeting iNOS may be helpful in the near future to establish novel therapeutic approaches.

Author Contribution

GO

N.U.C. conceived the primary idea and searched all study materials. A.T. Prepared the pathways. J.S., P.D.N.,N.A., S.A., T.F. and W.M. Helped NUC to write the initial chapters and tables. M.M.M. proposed the pathways. N.U.C., M.M.M. and M.A.T.S. _nalized the manuscript and trained all group members.

Acknowledgement

GO

The authors are grateful to Dr. Sarif Mohiuddin, PhD student in Aichi Medical University, Japan to guide us on this manuscript.
 

Competing financial interest

GO

Authors have declared that no competing interest exists.

References


Abdelsalam, R. M., & Safar, M. M. (2015). Neuroprotective effects of vildagliptin in rat rotenone Parkinson's disease model: role of RAGE-NFκB and Nrf2-antioxidant signaling pathways. Journal of neurochemistry, 133(5), 700-707.
https://doi.org/10.1111/jnc.13087
PMid:25752913

Abu Taher Sagor, M. Angiotensin-II, a potent peptide, participates in the development of hepatic dysfunctions. Immunology, Endocrine & Metabolic Agents in Medicinal Chemistry, 16, 1-17.

Abu Taher Sagor, M., Mahmud Reza, H., Tabassum, N., Sikder, B., Ulla, A., Subhan, N., . . . Ashraful Alam, M. (2016). Supplementation of rosemary leaves (Rosmarinus officinalis) powder attenuates oxidative stress, inflammation and fibrosis in carbon tetrachloride (CCl4) treated rats. Current Nutrition & Food Science, 12(4), 288-295.
https://doi.org/10.2174/1573401312666160816154610

Aktan, F. (2004). iNOS-mediated nitric oxide production and its regulation. Life sciences, 75(6), 639-653.
https://doi.org/10.1016/j.lfs.2003.10.042
PMid:15172174

Alam, M. A., Chowdhury, M. R. H., Jain, P., Sagor, M. A. T., & Reza, H. M. (2015). DPP-4 inhibitor sitagliptin prevents inflammation and oxidative stress of heart and kidney in two kidney and one clip (2K1C) rats. Diabetology & metabolic syndrome, 7(1), 107.
https://doi.org/10.1186/s13098-015-0095-3
PMid:26609328 PMCid:PMC4658771

Alderton, W. K., Cooper, C. E., & Knowles, R. G. (2001). Nitric oxide synthases: structure, function and inhibition. Biochemical Journal, 357(3), 593-615.
https://doi.org/10.1042/0264-6021:3570593
https://doi.org/10.1042/bj3570593
PMid:11463332 PMCid:PMC1221991

Ali, A., Kaur, G., Hamid, H., Abdullah, T., Ali, M., Niwa, M., & Alam, M. (2003). Terminoside A, a new triterpene glycoside from the bark of Terminalia arjuna inhibits nitric oxide production in murine macrophages. Journal of Asian natural products research, 5(2), 137-142.
https://doi.org/10.1080/1028602031000066834
PMid:12765198

Anavi, S., Eisenberg-Bord, M., Hahn-Obercyger, M., Genin, O., Pines, M., & Tirosh, O. (2015). The role of iNOS in cholesterol-induced liver fibrosis. Laboratory investigation, 95(8), 914.
https://doi.org/10.1038/labinvest.2015.67
PMid:26097999

Asano, K., Chee, C., Gaston, B., Lilly, C. M., Gerard, C., Drazen, J. M., & Stamler, J. S. (1994). Constitutive and inducible nitric oxide synthase gene expression, regulation, and activity in human lung epithelial cells. Proceedings of the National Academy of Sciences, 91(21), 10089-10093.
https://doi.org/10.1073/pnas.91.21.10089

AT Sagor, M., M Mohib, M., S Azam, M., Rahman, A., T Tanmoy, F., K Chowdhury, W., . . . A Alam, M. (2016). Angiotensin-II, a Potent Peptide, Participates in the Development of Hepatic Dysfunctions. Immunology, Endocrine & Metabolic Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Immunology, Endocrine and Metabolic Agents), 16(3), 161-177.

Bae, U.-J., Song, M.-Y., Jang, H.-Y., Lim, J. M., Lee, S. Y., Ryu, J.-H., & Park, B.-H. (2015). Emodin isolated from Rheum palmatum prevents cytokine-induced β-cell damage and the development of type 1 diabetes. Journal of Functional Foods, 16, 9-19.
https://doi.org/10.1016/j.jff.2015.04.016

Ballantyne, C. M., & Nambi, V. (2005). Markers of inflammation and their clinical significance. Atherosclerosis Supplements, 6(2), 21-29.
https://doi.org/10.1016/j.atherosclerosissup.2005.02.005
PMid:15823493

Baynosa, R. C., Naig, A. L., Murphy, P. S., Fang, X. H., Stephenson, L. L., Khiabani, K. T., . . . Zamboni, W. A. (2013). The effect of hyperbaric oxygen on nitric oxide synthase activity and expression in ischemia-reperfusion injury. journal of surgical research, 183(1), 355-361.
https://doi.org/10.1016/j.jss.2013.01.004
PMid:23485074

Cabrera-Fuentes, H. A., Ruiz-Meana, M., Simsekyilmaz, S., Kostin, S., Inserte, J., Saffarzadeh, M., . . . Barreto, G. (2014). RNase1 prevents the damaging interplay between extracellular RNA and tumour necrosis factor-α in cardiac ischaemia/reperfusion injury. Thrombosis & Haemostasis, 112(6), 1110-1119.
https://doi.org/10.1160/th14-08-0703
PMid:25354936

Chan, E. D., Chan, J., & Schluger, N. W. (2001). What is the role of nitric oxide in murine and human host defense against tuberculosis? Current knowledge. American journal of respiratory cell and molecular biology, 25(5), 606-612.
https://doi.org/10.1165/ajrcmb.25.5.4487
PMid:11713103

Charles, I. G., Palmer, R., Hickery, M. S., Bayliss, M. T., Chubb, A. P., Hall, V. S., . . . Moncada, S. (1993). Cloning, characterization, and expression of a cDNA encoding an inducible nitric oxide synthase from the human chondrocyte. Proceedings of the National Academy of Sciences, 90(23), 11419-11423.
https://doi.org/10.1073/pnas.90.23.11419

Chen, B.-Y., Lin, D. P.-C., Wu, C.-Y., Teng, M.-C., Sun, C.-Y., Tsai, Y.-T., . . . Chang, H.-H. (2011). Dietary zerumbone prevents mouse cornea from UVB-induced photokeratitis through inhibition of NF-κB, iNOS, and TNF-α expression and reduction of MDA accumulation.

Cho, H. J., Xie, Q., Calaycay, J., Mumford, R. A., Swiderek, K. M., Lee, T. D., & Nathan, C. (1992). Calmodulin is a subunit of nitric oxide synthase from macrophages. Journal of experimental medicine, 176(2), 599-604.
https://doi.org/10.1084/jem.176.2.599
PMid:1380065

Chow, B. S. M., Chew, E. G. Y., Zhao, C., Bathgate, R. A., Hewitson, T. D., & Samuel, C. S. (2012). Relaxin signals through a RXFP1-pERK-nNOS-NO-cGMP-dependent pathway to up-regulate matrix metalloproteinases: the additional involvement of iNOS. PloS one, 7(8), e42714.
https://doi.org/10.1371/journal.pone.0042714
PMid:22936987 PMCid:PMC3425563

Chowdhury, N. U., Farooq, T., Abdullah, S., Mahadi, A. S., Hasan, M. M., Paran, T. Z., . . . Alam, M. A. (2016). Matrix Metalloproteinases (MMP), a Major Responsible Downstream Signaling Molecule for Cellular Damage-A Review. Molecular Enzymology and Drug Targets.
https://doi.org/10.21767/2572-5475.10019

Costa, M. A., Elesgaray, R., Caniffi, C., Fellet, A., Mac Laughlin, M., & Arranz, C. (2010). Role of nitric oxide as a key mediator on cardiovascular actions of atrial natriuretic peptide in spontaneously hypertensive rats. American Journal of Physiology-Heart and Circulatory Physiology, 298(3), H778-H786.
https://doi.org/10.1152/ajpheart.00488.2009
PMid:19783776

Crump, C., Sundquist, J., Winkleby, M. A., & Sundquist, K. (2017). Interactive effects of obesity and physical fitness on risk of ischemic heart disease. International journal of obesity (2005), 41(2), 255.
https://doi.org/10.1038/ijo.2016.209
PMid:27867205 PMCid:PMC5296285

Csont, T., Viappiani, S., Sawicka, J., Slee, S., Altarejos, J. Y., Batinic-Haberle, I., & Schulz, R. (2005). The involvement of superoxide and iNOS-derived NO in cardiac dysfunction induced by pro-inflammatory cytokines. Journal of molecular and cellular cardiology, 39(5), 833-840.
https://doi.org/10.1016/j.yjmcc.2005.07.010
PMid:16171809

Daff, S., Sagami, I., & Shimizu, T. (1999). The 42-amino acid insert in the FMN domain of neuronal nitric-oxide synthase exerts control over Ca2+/calmodulin-dependent electron transfer. Journal of Biological Chemistry, 274(43), 30589-30595.
https://doi.org/10.1074/jbc.274.43.30589
PMid:10521442

Das, A., Xi, L., & Kukreja, R. C. (2005). Phosphodiesterase-5 inhibitor sildenafil preconditions adult cardiac myocytes against necrosis and apoptosis Essential role of nitric oxide signaling. Journal of Biological Chemistry, 280(13), 12944-12955.
https://doi.org/10.1074/jbc.M404706200
PMid:15668244

Das, S., Alagappan, V. K., Bagchi, D., Sharma, H. S., Maulik, N., & Das, D. K. (2005). Coordinated induction of iNOS–VEGF–KDR–eNOS after resveratrol consumption: A potential mechanism for resveratrol preconditioning of the heart. Vascular pharmacology, 42(5), 281-289.
https://doi.org/10.1016/j.vph.2005.02.013
PMid:15905131

Depre, C., Havaux, X., Renkin, J., Vanoverschelde, J. L. J., & Wijns, W. (1999). Expression of inducible nitric oxide synthase in human coronary atherosclerotic plaque. Cardiovascular research, 41(2), 465-472.
https://doi.org/10.1016/S0008-6363(98)00304-6

Depré, C., Wijns, W., Robert, A. M., Renkin, J. P., & Havaux, X. (1997). Pathology of unstable plaque: correlation with the clinical severity of acute coronary syndromes. Journal of the American College of Cardiology, 30(3), 694-702.
https://doi.org/10.1016/S0735-1097(97)00213-1

Dhillon, S. S., Mastropaolo, L. A., Murchie, R., Griffiths, C., Thöni, C., Elkadri, A., . . . Guo, C. (2014). Higher activity of the inducible nitric oxide synthase contributes to very early onset inflammatory bowel disease. Clinical and translational gastroenterology, 5(1), e46.
https://doi.org/10.1038/ctg.2013.17
PMid:24430113 PMCid:PMC3912315

Dias, A. S., Porawski, M., Alonso, M., Marroni, N., Collado, P. S., & González-Gallego, J. (2005). Quercetin decreases oxidative stress, NF-κB activation, and iNOS overexpression in liver of streptozotocin-induced diabetic rats. The Journal of nutrition, 135(10), 2299-2304.
https://doi.org/10.1093/jn/135.10.2299
PMid:16177186

Dwivedi, S. (2007). Terminalia arjuna Wight & Arn.—a useful drug for cardiovascular disorders. Journal of Ethnopharmacology, 114(2), 114-129.
https://doi.org/10.1016/j.jep.2007.08.003
PMid:17875376

Eckel, R. H. (1997). Obesity and heart disease. Circulation, 96(9), 3248-3250.
https://doi.org/10.1161/01.CIR.96.9.3248
PMid:9386201

Eggers, K. M., Oldgren, J., Nordenskjöld, A., & Lindahl, B. (2004). Diagnostic value of serial measurement of cardiac markers in patients with chest pain: limited value of adding myoglobin to troponin I for exclusion of myocardial infarction. American heart journal, 148(4), 574-581.
https://doi.org/10.1016/j.ahj.2004.04.030
PMid:15459585

Escames, G., Khaldy, H., León, J., González, L., & Acu-a-Castroviejo, D. (2004). Changes in iNOS activity, oxidative stress and melatonin levels in hypertensive patients treated with lacidipine. Journal of hypertension, 22(3), 629-635.
https://doi.org/10.1097/00004872-200403000-00027
PMid:15076170

Ferrari, P. F., Palmieri, D., Casazza, A. A., Aliakbarian, B., Perego, P., & Palombo, D. (2015). TNFα-induced endothelial activation is counteracted by polyphenol extract from UV-stressed cyanobacterium Arthrospira platensis. Medicinal Chemistry Research, 24(1), 275-282.
https://doi.org/10.1007/s00044-014-1126-6

Ferreiro, C. R., Chagas, A. C. P., Carvalho, M. H. C., Dantas, A. P., Jatene, M. B., de Souza, L. C. B., & da Luz, P. L. (2001). Influence of hypoxia on nitric oxide synthase activity and gene expression in children with congenital heart disease: a novel pathophysiological adaptive mechanism. Circulation, 103(18), 2272-2276.
https://doi.org/10.1161/01.CIR.103.18.2272
PMid:11342476

Fukumura, D., Gohongi, T., Kadambi, A., Izumi, Y., Ang, J., Yun, C.-O., . . . Jain, R. K. (2001). Predominant role of endothelial nitric oxide synthase in vascular endothelial growth factor-induced angiogenesis and vascular permeability. Proceedings of the National Academy of Sciences, 98(5), 2604-2609.
https://doi.org/10.1073/pnas.041359198
PMid:11226286 PMCid:PMC30185

Funakoshi, H., Kubota, T., Kawamura, N., Machida, Y., Feldman, A. M., Tsutsui, H., . . . Takeshita, A. (2002). Disruption of inducible nitric oxide synthase improves β-adrenergic inotropic responsiveness but not the survival of mice with cytokine-induced cardiomyopathy. Circulation research, 90(9), 959-965.
https://doi.org/10.1161/01.RES.0000017632.83720.68
PMid:12016261

Funakoshi, H., Kubota, T., Machida, Y., Kawamura, N., Feldman, A. M., Tsutsui, H., . . . Takeshita, A. (2002). Involvement of inducible nitric oxide synthase in cardiac dysfunction with tumor necrosis factor-α. American Journal of Physiology-Heart and Circulatory Physiology, 282(6), H2159-H2166.
https://doi.org/10.1152/ajpheart.00872.2001
PMid:12003824

 

Gupta, A., Rhodes, G. J., Berg, D. T., Gerlitz, B., Molitoris, B. A., & Grinnell, B. W. (2007). Activated protein C ameliorates LPS-induced acute kidney injury and downregulates renal INOS and angiotensin 2. American Journal of Physiology-Renal Physiology, 293(1), F245-F254.
https://doi.org/10.1152/ajprenal.00477.2006
PMid:17409278

Ham, Y.-M., Ko, Y.-J., Song, S.-M., Kim, J., Kim, K.-N., Yun, J.-H., . . . Yoon, W.-J. (2015). Anti-inflammatory effect of litsenolide B2 isolated from Litsea japonica fruit via suppressing NF-κB and MAPK pathways in LPS-induced RAW264. 7 cells. Journal of functional foods, 13, 80-88.
https://doi.org/10.1016/j.jff.2014.12.031

Haywood, G. A., Tsao, P. S., Heiko, E., Mann, M. J., Keeling, P. J., Trindade, P. T., . . . Bishopric, N. H. (1996). Expression of inducible nitric oxide synthase in human heart failure. Circulation, 93(6), 1087-1094.
https://doi.org/10.1161/01.CIR.93.6.1087
PMid:8653828

Heger, J., Gödecke, A., Flögel, U., Merx, M. W., Molojavyi, A., Kühn-Velten, W. N., & Schrader, J. (2002). Cardiac-specific overexpression of inducible nitric oxide synthase does not result in severe cardiac dysfunction. Circulation research, 90(1), 93-99.
https://doi.org/10.1161/hh0102.102757
PMid:11786524

Heron, M., & Anderson, R. N. (2016). Changes in the leading cause of death: recent patterns in heart disease and cancer mortality. Cancer, 400(500,000), 600,000.

Heymes, C., Bendall, J. K., Ratajczak, P., Cave, A. C., Samuel, J.-L., Hasenfuss, G., & Shah, A. M. (2003). Increased myocardial NADPH oxidase activity in human heart failure. Journal of the American College of Cardiology, 41(12), 2164-2171.
https://doi.org/10.1016/S0735-1097(03)00471-6

Ishibashi, M., Hiasa, K.-i., Zhao, Q., Inoue, S., Ohtani, K., Kitamoto, S., . . . Kura, S. (2004). Critical role of monocyte chemoattractant protein-1 receptor CCR2 on monocytes in hypertension-induced vascular inflammation and remodeling. Circulation research, 94(9), 1203-1210.
https://doi.org/10.1161/01.RES.0000126924.23467.A3
PMid:15059935

Islam, M. N., Choi, R. J., Jin, S. E., Kim, Y. S., Ahn, B. R., Zhao, D., . . . Choi, J. S. (2012). Mechanism of anti-inflammatory activity of umbelliferone 6-carboxylic acid isolated from Angelica decursiva. Journal of ethnopharmacology, 144(1), 175-181.
https://doi.org/10.1016/j.jep.2012.08.048
PMid:22981803

Islam, M. N., Ishita, I. J., Jin, S. E., Choi, R. J., Lee, C. M., Kim, Y. S., . . . Choi, J. S. (2013). Anti-inflammatory activity of edible brown alga Saccharina japonica and its constituents pheophorbide a and pheophytin a in LPS-stimulated RAW 264.7 macrophage cells. Food and Chemical Toxicology, 55, 541-548.
https://doi.org/10.1016/j.fct.2013.01.054
PMid:23402855

Janakiram, N. B., & Rao, C. V. (2012). iNOS-selective inhibitors for cancer prevention: promise and progress. Future medicinal chemistry, 4(17), 2193-2204.
https://doi.org/10.4155/fmc.12.168
PMid:23190107 PMCid:PMC3588580

Janssen-Heininger, Y. M., Macara, I., & Mossman, B. T. (1999). Cooperativity between Oxidants and Tumor Necrosis Factor in the Activation of Nuclear Factor (NF)-κ B: Requirement of Ras/Mitogen-Activated Protein Kinases in the Activation of NF-κ B by Oxidants. American journal of respiratory cell and molecular biology, 20(5), 942-952.
https://doi.org/10.1165/ajrcmb.20.5.3452
PMid:10226064

Jiang, Q., Elson-Schwab, I., Courtemanche, C., & Ames, B. N. (2000). γ-Tocopherol and its major metabolite, in contrast to α-tocopherol, inhibit cyclooxygenase activity in macrophages and epithelial cells. Proceedings of the National Academy of Sciences, 97(21), 11494-11499.
https://doi.org/10.1073/pnas.200357097
PMid:11005841 PMCid:PMC17228

Jin, M., Suh, S.-J., Yang, J. H., Lu, Y., Kim, S. J., Kwon, S., . . . Ahn, G. W. (2010). Anti-inflammatory activity of bark of Dioscorea batatas DECNE through the inhibition of iNOS and COX-2 expressions in RAW264. 7 cells via NF-κB and ERK1/2 inactivation. Food and chemical toxicology, 48(11), 3073-3079.
https://doi.org/10.1016/j.fct.2010.07.048
PMid:20691245

Jung, H. A., Jin, S. E., Ahn, B. R., Lee, C. M., & Choi, J. S. (2013). Anti-inflammatory activity of edible brown alga Eisenia bicyclis and its constituents fucosterol and phlorotannins in LPS-stimulated RAW264. 7 macrophages. Food and Chemical Toxicology, 59, 199-206.
https://doi.org/10.1016/j.fct.2013.05.061
PMid:23774261

Kapoor, D., Vijayvergiya, R., & Dhawan, V. (2014). Terminalia arjuna in coronary artery disease: ethnopharmacology, pre-clinical, clinical & safety evaluation. Journal of ethnopharmacology, 155(2), 1029-1045.
https://doi.org/10.1016/j.jep.2014.06.056
PMid:25014508

Kapur, S., Marcotte, B., & Marette, A. (1999). Mechanism of adipose tissue iNOS induction in endotoxemia. American Journal of Physiology-Endocrinology and Metabolism, 276(4), E635-E641.
https://doi.org/10.1152/ajpendo.1999.276.4.E635

Kengatharan, K. M., De Kimpe, S., Robson, C., Foster, S. J., & Thiemermann, C. (1998). Mechanism of gram-positive shock: identification of peptidoglycan and lipoteichoic acid moieties essential in the induction of nitric oxide synthase, shock, and multiple organ failure. Journal of Experimental Medicine, 188(2), 305-315.
https://doi.org/10.1084/jem.188.2.305
PMid:9670043

Keyes, K. T., Ye, Y., Lin, Y., Zhang, C., Perez-Polo, J. R., Gjorstrup, P., & Birnbaum, Y. (2010). Resolvin E1 protects the rat heart against reperfusion injury. American Journal of Physiology-Heart and Circulatory Physiology, 299(1), H153-H164.
https://doi.org/10.1152/ajpheart.01057.2009
PMid:20435846

Khan, M. I., Ostadhadi, S., Mumtaz, F., Momeny, M., Moghaddaskho, F., Hassanipour, M., . . . Dehpour, A. R. (2017). Thalidomide attenuates the development and expression of antinociceptive tolerance to μ-opioid agonist morphine through l-arginine-iNOS and nitric oxide pathway. Biomedicine & Pharmacotherapy, 85, 493-502.
https://doi.org/10.1016/j.biopha.2016.11.056
PMid:27899254

Khan, S., & Jena, G. (2014). Sodium butyrate, a HDAC inhibitor ameliorates eNOS, iNOS and TGF-β1-induced fibrogenesis, apoptosis and DNA damage in the kidney of juvenile diabetic rats. Food and chemical toxicology, 73, 127-139.
https://doi.org/10.1016/j.fct.2014.08.010
PMid:25158305

Khanduja, K. L., Sohi, K. K., Pathak, C. M., & Kaushik, G. (2006). Nimesulide inhibits lipopolysaccharide-induced production of superoxide anions and nitric oxide and iNOS expression in alveolar macrophages. Life sciences, 78(15), 1662-1669.
https://doi.org/10.1016/j.lfs.2005.07.033
PMid:16243362

Kibbe, M. R., Li, J., Nie, S., Watkins, S. C., Lizonova, A., Kovesdi, I., . . . Tzeng, E. (2000). Inducible nitric oxide synthase (iNOS) expression upregulates p21 and inhibits vascular smooth muscle cell proliferation through p42/44 mitogen-activated protein kinase activation and independent of p53 and cyclic guanosine monophosphate. Journal of vascular surgery, 31(6), 1214-1228.
https://doi.org/10.1067/mva.2000.105006
PMid:10842159

Korkmaz, B., Buharalioglu, K., Sahan-Firat, S., Cuez, T., Demiryurek, A. T., & Tunctan, B. (2011). Activation of MEK1/ERK1/2/iNOS/sGC/PKG pathway associated with peroxynitrite formation contributes to hypotension and vascular hyporeactivity in endotoxemic rats. Nitric Oxide, 24(3), 160-172.
https://doi.org/10.1016/j.niox.2011.02.004
PMid:21354320

Lechner, M., Lirk, P., & Rieder, J. (2005). Inducible nitric oxide synthase (iNOS) in tumor biology: the two sides of the same coin. Paper presented at the Seminars in cancer biology.
https://doi.org/10.1016/j.semcancer.2005.04.004
PMid:15914026

Lee, D.-H., Cho, H. J., Kim, J.-T., Choi, J. S., & Joo, C.-K. (2001). Expression of vascular endothelial growth factor and inducible nitric oxide synthase in pterygia. Cornea, 20(7), 738-742.
https://doi.org/10.1097/00003226-200110000-00013
PMid:11588427

Lee, H.-H., Paudel, K. R., & Kim, D.-W. (2015). Terminalia chebula fructus inhibits migration and proliferation of vascular smooth muscle cells and production of inflammatory mediators in RAW 264.7. Evidence-Based Complementary and Alternative Medicine, 2015.

Lee, J. K., Sayers, B. C., Chun, K.-S., Lao, H.-C., Shipley-Phillips, J. K., Bonner, J. C., & Langenbach, R. (2012). Multi-walled carbon nanotubes induce COX-2 and iNOS expression via MAP kinase-dependent and-independent mechanisms in mouse RAW264. 7 macrophages. Particle and fibre toxicology, 9(1), 14.
https://doi.org/10.1186/1743-8977-9-14
PMid:22571318 PMCid:PMC3485091

Li, X., Ye, X., Li, X., Sun, X., Liang, Q., Tao, L., . . . Chen, J. (2011). Salidroside protects against MPP+-induced apoptosis in PC12 cells by inhibiting the NO pathway. Brain research, 1382, 9-18.
https://doi.org/10.1016/j.brainres.2011.01.015
PMid:21241673

Li, Y., Shen, L., & Luo, H. (2016). Luteolin ameliorates dextran sulfate sodium-induced colitis in mice possibly through activation of the Nrf2 signaling pathway. Int Immunopharmacol, 40, 24-31.
https://doi.org/10.1016/j.intimp.2016.08.020
https://doi.org/10.1016/j.intimp.2016.01.027

Lim, H.-W., Kumar, H., Kim, B.-W., More, S. V., Kim, I.-W., Park, J.-I., . . . Choi, D.-K. b-Asarone (cis-2, 4, 5-trimethoxy-1-allyl phenyl), attenuates pro-inflammatory mediators by inhibiting NF-jB signaling and the JNK pathway in LPS activated BV-2 microglia cells.

Liu, F.-q., Zhang, X.-l., Gong, L., Wang, X.-p., Wang, J., Hou, X.-g., . . . Zhang, Y. (2011). Glucagon-like peptide 1 protects microvascular endothelial cells by inactivating the PARP-1/iNOS/NO pathway. Molecular and cellular endocrinology, 339(1-2), 25-33.
https://doi.org/10.1016/j.mce.2011.03.007
PMid:21458523

Liu, X.-m., Peyton, K. J., Wang, X., & Durante, W. (2012). Sildenafil stimulates the expression of gaseous monoxide-generating enzymes in vascular smooth muscle cells via distinct signaling pathways. Biochemical pharmacology, 84(8), 1045-1054.
https://doi.org/10.1016/j.bcp.2012.07.023
PMid:22864061 PMCid:PMC3487387

Loh, S. Y., Giribabu, N., & Salleh, N. (2017). Changes in plasma aldosterone and electrolytes levels, kidney epithelial sodium channel (ENaC) and blood pressure in normotensive WKY and hypertensive SHR rats following gonadectomy and chronic testosterone treatment. Steroids.
https://doi.org/10.1016/j.steroids.2017.09.008
PMid:28954214

Lowenstein, C. J., & Padalko, E. (2004). iNOS (NOS2) at a glance. Journal of cell science, 117(14), 2865-2867.
https://doi.org/10.1242/jcs.01166
PMid:15197240

Luoma, J. S., Strålin, P., Marklund, S. L., Hiltunen, T. P., Särkioja, T., & Ylä-Herttuala, S. (1998). Expression of extracellular SOD and iNOS in macrophages and smooth muscle cells in human and rabbit atherosclerotic lesions: colocalization with epitopes characteristic of oxidized LDL and peroxynitrite-modified proteins. Arteriosclerosis, thrombosis, and vascular biology, 18(2), 157-167.
https://doi.org/10.1161/01.ATV.18.2.157

MacMicking, J., Xie, Q.-w., & Nathan, C. (1997). Nitric oxide and macrophage function. Annual review of immunology, 15(1), 323-350.
https://doi.org/10.1146/annurev.immunol.15.1.323
PMid:9143691

MacMicking, J. D., North, R. J., LaCourse, R., Mudgett, J. S., Shah, S. K., & Nathan, C. F. (1997). Identification of nitric oxide synthase as a protective locus against tuberculosis. Proceedings of the National Academy of Sciences, 94(10), 5243-5248.
https://doi.org/10.1073/pnas.94.10.5243

Marumo, T., Nakaki, T., Hishikawa, K., Hirahashi, J., Suzuki, H., Kato, R., & Saruta, T. (1995). Natriuretic peptide-augmented induction of nitric oxide synthase through cyclic guanosine 3', 5'-monophosphate elevation in vascular smooth muscle cells. Endocrinology, 136(5), 2135-2142.
https://doi.org/10.1210/endo.136.5.7536663
PMid:7536663

Mayer, B., & Hemmens, B. (1997). Biosynthesis and action of nitric oxide in mammalian cells. Trends in biochemical sciences, 22(12), 477-481.
https://doi.org/10.1016/S0968-0004(97)01147-X

Metwaly, H., Ebrahem, M. A.-M., & Saku, T. (2014). Vascular endothelial growth factor (VEGF) and inducible nitric oxide synthase (iNOS) in oral lichen planus: An immunohistochemical study for the correlation between vascular and inflammatory reactions. Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology, 26(3), 390-396.
https://doi.org/10.1016/j.ajoms.2013.09.009

Mogensen, J., Murphy, R. T., Shaw, T., Bahl, A., Redwood, C., Watkins, H., . . . McKenna, W. J. (2004). Severe disease expression of cardiac troponin C and T mutations in patients with idiopathic dilated cardiomyopathy. Journal of the American College of Cardiology, 44(10), 2033-2040.
https://doi.org/10.1016/j.jacc.2004.08.027
PMid:15542288

Mohib, M. M., Afnan, K., Paran, T. Z., Khan, S., Sarker, J., Hasan, N., . . . Sagor, M. A. T. (2017). Beneficial Role of Citrus Fruit Polyphenols Against Hepatic Dysfunctions: A Review. Journal of dietary supplements, 1-29.

Mohib, M. M., Hasan, I., Chowdhury, W. K., Chowdhury, N. U., Mohiuddin, S., Sagor, M. A. T., . . . Alam, M. A. (2016). Role of angiotensin ii in hepatic inflammation through MAPK pathway: A review. Hepatitis.

Mohib, M. M., Rabby, S. F., Paran, T. Z., Hasan, M. M., Ahmed, I., Hasan, N., . . . Mohiuddin, S. (2016). Protective role of green tea on diabetic nephropathy—A review. Cogent Biology, 2(1), 1248166.

Mohib, M. M., Rabby, S. M. F., Paran, T. Z., Hasan, M. M., Ahmed, I., Hasan, N., . . . Hsu, T.-C. (2016). Protective role of green tea on diabetic nephropathy -A review. Cogent Biology, 1248166. doi:10.1080/23312025.2016.1248166
https://doi.org/10.1080/23312025.2016.1248166

Müller-Werdan, U., Schumann, H., Fuchs, R., Reithmann, C., Loppnow, H., Koch, S., . . . Jungblut, P. (1997). Tumor Necrosis Factorα (TNFα) is Cardiodepressant in Pathophysiologically Relevant Concentrations Without Inducing Inducible Nitric Oxide-(NO)-Synthase (iNOS) or Triggering Serious Cytotoxicity. Journal of molecular and cellular cardiology, 29(11), 2915-2923.
https://doi.org/10.1006/jmcc.1997.0526
PMid:9405166

Mungrue, I. N., Gros, R., You, X., Pirani, A., Azad, A., Csont, T., . . . Husain, M. (2002a). Cardiomyocyte overexpression of iNOS in mice results in peroxynitrite generation, heart block, and sudden death. The Journal of clinical investigation, 109(6), 735-743.
https://doi.org/10.1172/JCI0213265
PMid:11901182 PMCid:PMC150906

 

Mungrue, I. N., Gros, R., You, X., Pirani, A., Azad, A., Csont, T., . . . Husain, M. (2002b). Cardiomyocyte overexpression of iNOS in mice results in peroxynitrite generation, heart block, and sudden death. The Journal of clinical investigation, 109(6), 735.
https://doi.org/10.1172/JCI0213265
PMid:11901182 PMCid:PMC150906

Nathan, C. (1992). Nitric oxide as a secretory product of mammalian cells. The FASEB journal, 6(12), 3051-3064.
https://doi.org/10.1096/fasebj.6.12.1381691
PMid:1381691

Padureanu, V., Enescu, A. S., Silosi, I., Fortofoiu, M., Enescu, A., Bogdan, M., . . . Mita, A. (2017). The association between chronic pancreatitis and the iNOS-2087A> G polymorphism. Romanian Journal of Internal Medicine, 55(2), 89-95.
https://doi.org/10.1515/rjim-2017-0009
PMid:28125406

Palmer, J., Guillard, F., Laurijssens, B., Wentz, A., Dixon, R., & Williams, P. (2009). A randomised, single-blind, placebo-controlled, adaptive clinical trial of GW274150, a selective iNOS inhibitor, in the treatment of acute migraine. Cephalalgia, 29(1), 124.

Papapetropoulos, A., García-Carde-a, G., Madri, J. A., & Sessa, W. C. (1997). Nitric oxide production contributes to the angiogenic properties of vascular endothelial growth factor in human endothelial cells. The Journal of clinical investigation, 100(12), 3131-3139.
https://doi.org/10.1172/JCI119868
PMid:9399960 PMCid:PMC508526

Piplani, H., Vaish, V., & Sanyal, S. N. (2012). Dolastatin 15, a mollusk linear peptide, and Celecoxib, a selective cyclooxygenase-2 inhibitor, prevent preneoplastic colonic lesions and induce apoptosis through inhibition of the regulatory transcription factor NF-κB and an inflammatory protein, iNOS. European Journal of Cancer Prevention, 21(6), 511-522.
https://doi.org/10.1097/CEJ.0b013e328351c69d
PMid:22441428

Ramlagan, P., Rondeau, P., Planesse, C., Neergheen-Bhujun, V. S., Fawdar, S., Bourdon, E., & Bahorun, T. (2017). Punica granatum L. mesocarp suppresses advanced glycation end products (AGEs)-and H 2 O 2-induced oxidative stress and pro-inflammatory biomarkers. Journal of Functional Foods, 29, 115-126.
https://doi.org/10.1016/j.jff.2016.12.007

Rasoanaivo, P., Wright, C. W., Willcox, M. L., & Gilbert, B. (2011). Whole plant extracts versus single compounds for the treatment of malaria: synergy and positive interactions. Malaria Journal, 10(1), S4.
https://doi.org/10.1186/1475-2875-10-S1-S4
PMid:21411015 PMCid:PMC3059462

Rodrigues, J. P. F., Caldas, I. S., Gonçalves, R. V., Almeida, L. A., Souza, R. L. M., & Novaes, R. D. (2017). S. mansoni-T. cruzi co-infection modulates arginase-1/iNOS expression, liver and heart disease in mice. Nitric Oxide, 66, 43-52.
https://doi.org/10.1016/j.niox.2017.02.013
PMid:28268114

Romano, B., Pagano, E., Orlando, P., Capasso, R., Cascio, M. G., Pertwee, R., . . . Borrelli, F. (2016). Pure Δ 9-tetrahydrocannabivarin and a Cannabis sativa extract with high content in Δ 9-tetrahydrocannabivarin inhibit nitrite production in murine peritoneal macrophages. Pharmacological Research, 113, 199-208.
https://doi.org/10.1016/j.phrs.2016.07.045
PMid:27498155

Ryu, S., Shin, J.-S., Cho, Y.-W., Kim, H. K., Paik, S. H., Lee, J. H., . . . Lee, K.-T. (2013). Fimasartan, anti-hypertension drug, suppressed inducible nitric oxide synthase expressions via nuclear factor-kappa B and activator protein-1 inactivation. Biological and Pharmaceutical Bulletin, 36(3), 467-474.
https://doi.org/10.1248/bpb.b12-00859
PMid:23449332

Sagor, M. A. T., Mohib, M., Tabassum, N., Ahmed, I., & Reza, H. (2016). Fresh seed supplementation of Syzygium cumini attenuated oxidative stress, inflammation, fibrosis, iron overload, hepatic dysfunction and renal injury in acetaminophen induced rats. Journal of Drug Metabolism & Toxicology, 7(2).

Sagor, M. A. T., Tabassum, N., Potol, M. A., & Alam, M. A. (2015). Xanthine oxidase inhibitor, allopurinol, prevented oxidative stress, fibrosis, and myocardial damage in isoproterenol induced aged rats. Oxidative medicine and cellular longevity, 2015.
https://doi.org/10.1155/2015/478039
PMid:26137187 PMCid:PMC4475550

Sagor, M. A. T., Tabassum, N., Potol, M. A., & Alam, M. A. (2015). Xanthine Oxidase Inhibitor, Allopurinol, Prevented Oxidative Stress, Fibrosis, and Myocardial Damage in Isoproterenol Induced Aged Rats. Oxidative Medicine and Cellular Longevity, 2015, 9. doi:10.1155/2015/478039
https://doi.org/10.1155/2015/478039

Salerno, J. C., Harris, D. E., Irizarry, K., Patel, B., Morales, A. J., Smith, S. M., . . . Jones, C. L. (1997). An autoinhibitory control element defines calcium-regulated isoforms of nitric oxide synthase. Journal of Biological Chemistry, 272(47), 29769-29777.
https://doi.org/10.1074/jbc.272.47.29769
PMid:9368047

Salvemini, D., Nucci, G., Sneddon, J. M., & Vane, J. R. (1989). Superoxide anions enhance platelet adhesion and aggregation. British journal of pharmacology, 97(4), 1145-1150.
https://doi.org/10.1111/j.1476-5381.1989.tb12572.x
PMid:2551440 PMCid:PMC1854596

Sangartit, W., Pakdeechote, P., Kukongviriyapan, V., Donpunha, W., Shibahara, S., & Kukongviriyapan, U. (2016). Tetrahydrocurcumin in combination with deferiprone attenuates hypertension, vascular dysfunction, baroreflex dysfunction, and oxidative stress in iron-overloaded mice. Vascular Pharmacology, 87, 199-208.
https://doi.org/10.1016/j.vph.2016.10.001
PMid:27713040

Sarady, J. K., Zuckerbraun, B. S., Bilban, M., Wagner, O., Usheva, A., Liu, F., . . . Otterbein, L. E. (2004). Carbon monoxide protection against endotoxic shock involves reciprocal effects on iNOS in the lung and liver. The FASEB journal, 18(7), 854-856.
https://doi.org/10.1096/fj.03-0643fje
PMid:15001560

Satoh, M., Nakamura, M., Tamura, G., Makita, S., Segawa, I., Tashiro, A., . . . Hiramori, K. (1997). Inducible nitric oxide synthase and tumor necrosis factor-alpha in myocardium in human dilated cardiomyopathy. Journal of the American College of Cardiology, 29(4), 716-724.
https://doi.org/10.1016/S0735-1097(96)00567-0

Seccombe, J. F., Pearson, P. J., & Schaff, H. V. (1994). Oxygen radical—mediated vascular injury selectively inhibits receptor-dependent release of nitric oxide from canine coronary arteries. The Journal of thoracic and cardiovascular surgery, 107(2), 505-509.
PMid:8302070

Seo, S., Shin, J.-S., Lee, W.-S., Rhee, Y. K., Cho, C.-W., Hong, H.-D., & Lee, K.-T. (2017). Anti-colitis effect of Lactobacillus sakei K040706 via suppression of inflammatory responses in the dextran sulfate sodium-induced colitis mice model. Journal of Functional Foods, 29, 256-268.
https://doi.org/10.1016/j.jff.2016.12.045

Shabeeh, H., Khan, S., Jiang, B., Brett, S., Melikian, N., Casadei, B., . . . Shah, A. M. (2017). Blood Pressure in Healthy Humans Is Regulated by Neuronal NO SynthaseNovelty and Significance. Hypertension, 69(5), 970-976.
https://doi.org/10.1161/HYPERTENSIONAHA.116.08792
PMid:28264923 PMCid:PMC5389591

Shimabukuro, M., Ohneda, M., Lee, Y., & Unger, R. H. (1997). Role of nitric oxide in obesity-induced beta cell disease. Journal of Clinical Investigation, 100(2), 290.
https://doi.org/10.1172/JCI119534
PMid:9218505 PMCid:PMC508191

Simsekyilmaz, S., Cabrera-Fuentes, H. A., Meiler, S., Kostin, S., Baumer, Y., Liehn, E. A., . . . Zernecke, A. (2013). The role of extracellular RNA in atherosclerotic plaque formation in mice. Circulation, CIRCULATIONAHA. 113.002562.

Sun, Y., Carretero, O. A., Xu, J., Rhaleb, N.-E., Wang, F., Lin, C., . . . Yang, X.-P. (2005). Lack of inducible NO synthase reduces oxidative stress and enhances cardiac response to isoproterenol in mice with deoxycorticosterone acetate–salt hypertension. Hypertension, 46(6), 1355-1361.
https://doi.org/10.1161/01.HYP.0000192651.06674.3f
PMid:16286571 PMCid:PMC4601605

Taylor, B. S., & Geller, D. A. (2000). Molecular regulation of the human inducible nitric oxide synthase (iNOS) gene. Shock (Augusta, Ga.), 13(6), 413-424.
https://doi.org/10.1097/00024382-200006000-00001

Tikellis, C., Thomas, M. C., Harcourt, B. E., Coughlan, M. T., Pete, J., Bialkowski, K., . . . Forbes, J. M. (2008). Cardiac inflammation associated with a Western diet is mediated via activation of RAGE by AGEs. American Journal of Physiology-Endocrinology and Metabolism, 295(2), E323-E330.
https://doi.org/10.1152/ajpendo.00024.2008
PMid:18477705 PMCid:PMC2652498

Vannini, F., Kashfi, K., & Nath, N. (2015). The dual role of iNOS in cancer. Redox biology, 6, 334-343.
https://doi.org/10.1016/j.redox.2015.08.009
PMid:26335399 PMCid:PMC4565017

Vejlstrup, N. G., Bouloumie, A., Boesgaard, S., Andersen, C. B., Nielsen-Kudsk, J. E., Mortensen, S. A., . . . Aldershvile, J. (1998). Inducible nitric oxide synthase (iNOS) in the human heart: expression and localization in congestive heart failure. Journal of molecular and cellular cardiology, 30(6), 1215-1223.
https://doi.org/10.1006/jmcc.1998.0686
PMid:9689595

Wang, W. Z., Fang, X. H., Stepheson, L. L., Khiabani, K. T., & Zamboni, W. A. (2004). NOS upregulation attenuates vascular endothelial dysfunction in the late phase of ischemic preconditioning in skeletal muscle. Journal of orthopaedic research, 22(3), 578-585.
https://doi.org/10.1016/j.orthres.2003.10.004
PMid:15099638

Westermann, D., Lindner, D., Kasner, M., Zietsch, C., Savvatis, K., Escher, F., . . . Riad, A. (2010). Cardiac inflammation contributes to changes in the extracellular matrix in patients with heart failure and normal ejection fraction. Circulation: Heart Failure, CIRCHEARTFAILURE. 109.931451.

Wever, R. M., Lüscher, T. F., Cosentino, F., & Rabelink, T. J. (1998). Atherosclerosis and the two faces of endothelial nitric oxide synthase. Circulation, 97(1), 108-112.
https://doi.org/10.1161/01.CIR.97.1.108
PMid:9443438

Wong, M.-L., Rettori, V., Amer, A.-S., Bongiorno, P. B., Canteros, G., McCann, S. M., . . . Licinio, J. (1996). Inducible nitric oxide synthase gene expression in the brain during systemic inflammation. Nature medicine, 2(5), 581-584.
https://doi.org/10.1038/nm0596-581
PMid:8616720

Xia, M., Ling, W. H., Ma, J., Kitts, D. D., & Zawistowski, J. (2003). Supplementation of diets with the black rice pigment fraction attenuates atherosclerotic plaque formation in apolipoprotein e deficient mice. The Journal of nutrition, 133(3), 744-751.
https://doi.org/10.1093/jn/133.3.744
PMid:12612147

Xia, X., Ling, W., Ma, J., Xia, M., Hou, M., Wang, Q., . . . Tang, Z. (2006). An anthocyanin-rich extract from black rice enhances atherosclerotic plaque stabilization in apolipoprotein E–deficient mice. The Journal of nutrition, 136(8), 2220-2225.
https://doi.org/10.1093/jn/136.8.2220
PMid:16857844

Xu, W., Charles, I. G., Moncada, S., Gorman, P., Sheer, D., Liu, L., & Emson, P. (1994). Mapping of the genes encoding human inducible and endothelial nitric oxide synthase (NOS2 and NOS3) to the pericentric region of chromosome 17 and to chromosome 7, respectively. Genomics, 21(2), 419-422.
https://doi.org/10.1006/geno.1994.1286
PMid:7522210

Xuan, Y.-T., Guo, Y., Han, H., Zhu, Y., & Bolli, R. (2001). An essential role of the JAK-STAT pathway in ischemic preconditioning. Proceedings of the National Academy of Sciences, 98(16), 9050-9055.
https://doi.org/10.1073/pnas.161283798
PMid:11481471 PMCid:PMC55371

Yang, B., Larson, D. F., & Watson, R. R. (2004). Modulation of iNOS activity in age-related cardiac dysfunction. Life sciences, 75(6), 655-667.
https://doi.org/10.1016/j.lfs.2003.09.076
PMid:15172175

Ying, L., & Hofseth, L. J. (2007). An emerging role for endothelial nitric oxide synthase in chronic inflammation and cancer. Cancer Research, 67(4), 1407-1410.
https://doi.org/10.1158/0008-5472.CAN-06-2149
PMid:17308075

Yoda, Y., Amagase, K., Kato, S., Tokioka, S., Murano, M., Kakimoto, K., . . . Higuchi, K. (2010). Prevention by lansoprazole, a proton pump inhibitor, of indomethacin-induced small intestinal ulceration in rats through induction of heme oxygenase-1. Journal of Physiology and Pharmacology, 61(3), 287.
PMid:20610858

Yu, G., Cheng, M., Wang, W., Zhao, R., & Liu, Z. (2017). Involvement of WNK1-mediated potassium channels in the sexual dimorphism of blood pressure. Biochemical and Biophysical Research Communications, 485(2), 255-260.
https://doi.org/10.1016/j.bbrc.2017.02.098
PMid:28237360

Yu, X. W., Chen, Q., Kennedy, R. H., & Liu, S. J. (2005). Inhibition of sarcoplasmic reticular function by chronic interleukin-6 exposure via iNOS in adult ventricular myocytes. The Journal of physiology, 566(2), 327-340.
https://doi.org/10.1113/jphysiol.2005.086686
https://doi.org/10.1113/jphysiol.2002.034223
PMid:15845578

Zhang, J., Peng, B., & Chen, X. (2005). Expressions of nuclear factor κB, inducible nitric oxide synthase, and vascular endothelial growth factor in adenoid cystic carcinoma of salivary glands: correlations with the angiogenesis and clinical outcome. Clinical Cancer Research, 11(20), 7334-
https://doi.org/10.1158/1078-0432.CCR-05-0241
PMid:16243805

Committee on Publication Ethics

PDF
Abstract
Export Citation

View Dimensions


View Plumx


View Altmetric




Save
0
Citation
1054
View

Share