51
|
Mannino MH, Patel RS, Eccardt AM, Perez Magnelli RA, Robinson CLC, Janowiak BE, Warren DE, Fisher JS. Myoglobin as a versatile peroxidase: Implications for a more important role for vertebrate striated muscle in antioxidant defense. Comp Biochem Physiol B Biochem Mol Biol 2019; 234:9-17. [PMID: 31051268 DOI: 10.1016/j.cbpb.2019.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/29/2019] [Accepted: 04/23/2019] [Indexed: 12/17/2022]
Abstract
Myoglobins (Mb) are ubiquitous proteins found in striated muscle of nearly all vertebrate taxa. Although their function is most commonly associated with facilitating oxygen storage and diffusion, Mb has also been implicated in cellular antioxidant defense. The oxidized (Fe3+) form of Mb (metMB) can react with hydrogen peroxide (H2O2) to produce ferrylMb. FerrylMb can be reduced back to metMb for another round of reaction with H2O2. In the present study, we have shown that horse skeletal muscle Mb displays peroxidase activity using 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid (ABTS) and 3,3',5,5'-tetramethylbenzidine (TMB) as reducing substrates, as well as the biologically-relevant substrates NADH/NADPH, ascorbate, caffeic acid, and resveratrol. We have also shown that ferrylMb can be reduced by both ethanol and acetaldehyde, which are known to accumulate in some vertebrate tissues under anaerobic conditions, such as anoxic goldfish and crucian carp, implying a potential mechanism for ethanol detoxification in striated muscle. We found that metMb peroxidase activity is pH-dependent, increasing as pH decreases from 7.4 to 6.1, which is biologically relevant to anaerobic vertebrate muscle when incurring intracellular lactic acidosis. Finally, we found that metMb reacts with hypochlorite in a heme-dependent fashion, indicating that Mb could play a role in hypochlorite detoxification. Taken together, these data suggest that Mb peroxidase activity might be an important antioxidant mechanism in vertebrate cardiac and skeletal muscle under a variety of physiological conditions, such as those that might occur in contracting skeletal muscle or during hypoxia.
Collapse
|
52
|
Davis GR, Bellar D. Montmorency cherry supplement does not affect aerobic exercise performance in healthy men. INT J VITAM NUTR RES 2019; 90:403-410. [PMID: 30932750 DOI: 10.1024/0300-9831/a000575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Aim: To determine the effects of short-term Montmorency cherry (MC) supplementation upon exercise performance, total blood nitrate levels, muscle oxygenation, and slow-component [Formula: see text]O2 kinetics. Methods: Twelve healthy male participants ingested a MC or placebo (PL) supplement in a randomized cross-over fashion over a six day period then cycled at a power output achieved at 70% of [Formula: see text]O2 peak for a maximum of 30 minutes or until exhaustion. Near-Infrared Spectroscopy sensors were used to determine muscle oxygenation. Blood was collected one hour post-supplement consumption on day one, day six, and one hour post-exercise. Results: All results are presented as mean ± SEM. Blood nitrate (μM/L) levels were not different one hour post-ingestion (MC = 8.30 ± 2.15, PL = 8.18 ± 1.86), following six days of supplementation (MC = 9.14 ± 1.89, PL = 7.24 ± 1.75) or one hour post-exercise (MC = 9.63 ± 1.61, PL = 7.97 ± 1.92) for treatment F = 0.26, p = 0.62; for time F = 0.45, p = 0.64; or treatment by time interaction F = 2.28, p = 0.13. Muscle oxygenation was not different between treatments for the right or left vastus lateralis, F = 0.68, p = 0.81 nor was time to respiratory compensation point (minutes) (MC = 18.40 ± 1.48, PL = 17.16 ± 1.78) F = 0.52, p = 0.60. MC supplement ingestion does not alter blood nitrate levels. Conclusion: Short-term MC ingestion does not increase muscle oxygenation during cycling exercise nor does it change slow-component [Formula: see text]O2 kinetics.
Collapse
Affiliation(s)
| | - David Bellar
- University of Louisiana at Lafayette, Lafayette, LA
| |
Collapse
|
53
|
Braganza A, Quesnelle K, Bickta J, Reyes C, Wang Y, Jessup M, St Croix C, Arlotti J, Singh SV, Shiva S. Myoglobin induces mitochondrial fusion, thereby inhibiting breast cancer cell proliferation. J Biol Chem 2019; 294:7269-7282. [PMID: 30872402 DOI: 10.1074/jbc.ra118.006673] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/21/2019] [Indexed: 01/11/2023] Open
Abstract
Myoglobin is a monomeric heme protein expressed ubiquitously in skeletal and cardiac muscle and is traditionally considered to function as an oxygen reservoir for mitochondria during hypoxia. It is now well established that low concentrations of myoglobin are aberrantly expressed in a significant proportion of breast cancer tumors. Despite being expressed only at low levels in these tumors, myoglobin is associated with attenuated tumor growth and a better prognosis in breast cancer patients, but the mechanism of this myoglobin-mediated protection against further cancer growth remains unclear. Herein, we report a signaling pathway by which myoglobin regulates mitochondrial dynamics and thereby decreases cell proliferation. We demonstrate in vitro that expression of human myoglobin in MDA-MB-231, MDA-MB-468, and MCF7 breast cancer cells induces mitochondrial hyperfusion by up-regulating mitofusins 1 and 2, the predominant catalysts of mitochondrial fusion. This hyperfusion causes cell cycle arrest and subsequent inhibition of cell proliferation. Mechanistically, increased mitofusin expression was due to myoglobin-dependent free-radical production, leading to the oxidation and degradation of the E3 ubiquitin ligase parkin. We recapitulated this pathway in a murine model in which myoglobin-expressing xenografts exhibited decreased tumor volume with increased mitofusin, markers of cell cycle arrest, and decreased parkin expression. Furthermore, in human triple-negative breast tumor tissues, mitofusin and myoglobin levels were positively correlated. Collectively, these results elucidate a new function for myoglobin as a modulator of mitochondrial dynamics and reveal a novel pathway by which myoglobin decreases breast cancer cell proliferation and tumor growth by up-regulating mitofusin levels.
Collapse
Affiliation(s)
| | | | - Janelle Bickta
- the Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania 15261
| | - Christopher Reyes
- the Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania 15261
| | - Yinna Wang
- From the Vascular Medicine Institute and
| | | | | | - Julie Arlotti
- Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, and.,University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232
| | - Shivendra V Singh
- Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, and.,University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232
| | - Sruti Shiva
- From the Vascular Medicine Institute and .,Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, and.,Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
54
|
Craig JC, Colburn TD, Hirai DM, Musch TI, Poole DC. Sexual dimorphism in the control of skeletal muscle interstitial Po 2 of heart failure rats: effects of dietary nitrate supplementation. J Appl Physiol (1985) 2019; 126:1184-1192. [PMID: 30844332 DOI: 10.1152/japplphysiol.01004.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Sex differences in the mechanisms underlying cardiovascular pathophysiology of O2 transport in heart failure (HF) remain to be explored. In HF, nitric oxide (NO) bioavailability is reduced and contributes to deficits in O2 delivery-to-utilization matching. Females may rely more on NO for cardiovascular control and as such experience greater decrements in HF. We tested the hypotheses that moderate HF induced by myocardial infarction would attenuate the skeletal muscle interstitial Po2 response to contractions (Po2is; determined by O2 delivery-to-utilization matching) compared with healthy controls and females would express greater dysfunction than male counterparts. Furthermore, we hypothesized that 5 days of dietary nitrate supplementation (Nitrate; 1 mmol·kg-1·day-1) would raise Po2is in HF rats. Forty-two Sprague-Dawley rats were randomly assigned to healthy, HF, or HF + Nitrate groups (each n = 14; 7 female/7 male). Spinotrapezius Po2is was measured via phosphorescence quenching during electrically induced twitch contractions (180 s; 1 Hz). HF reduced resting Po2is for both sexes compared with healthy controls (P < 0.01), and females were lower than males (14 ± 1 vs. 17 ± 2 mmHg) (P < 0.05). In HF both sexes expressed reduced Po2is amplitudes following the onset of muscle contractions compared with healthy controls (female: -41 ± 7%, male: -26 ± 12%) (P < 0.01). In HF rats, Nitrate elevated resting Po2is to values not different from healthy rats and removed the sex difference. Female HF + Nitrate rats expressed greater resting Po2is and amplitudes compared with female HF (P < 0.05). In this model of moderate HF, O2 delivery-to-utilization matching in the interstitial space is diminished in a sex-specific manner and dietary nitrate supplementation may serve to offset this reduction in HF rats with greater effects in females. NEW & NOTEWORTHY Interstitial Po2 (Po2is; indicative of O2 delivery-to-utilization matching) determines, in part, O2 flux into skeletal muscle. We show that heart failure (HF) reduces Po2is at rest and during skeletal muscle contractions in rats and this negative effect is amplified for females. However, elevating NO bioavailability with dietary nitrate supplementation increases resting Po2is and alters the dynamic response with greater efficacy in female HF rats, particularly at rest and following the onset of muscle contractions.
Collapse
Affiliation(s)
- Jesse C Craig
- Department of Kinesiology, Kansas State University , Manhattan, Kansas
| | - Trenton D Colburn
- Department of Kinesiology, Kansas State University , Manhattan, Kansas
| | - Daniel M Hirai
- Department of Kinesiology, Kansas State University , Manhattan, Kansas
| | - Timothy I Musch
- Department of Kinesiology, Kansas State University , Manhattan, Kansas.,Department of Anatomy and Physiology, Kansas State University , Manhattan, Kansas
| | - David C Poole
- Department of Kinesiology, Kansas State University , Manhattan, Kansas.,Department of Anatomy and Physiology, Kansas State University , Manhattan, Kansas
| |
Collapse
|
55
|
Oliveira-Paula GH, Pinheiro LC, Tanus-Santos JE. Mechanisms impairing blood pressure responses to nitrite and nitrate. Nitric Oxide 2019; 85:35-43. [PMID: 30716418 DOI: 10.1016/j.niox.2019.01.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/13/2018] [Accepted: 01/29/2019] [Indexed: 02/07/2023]
Abstract
Hypertension is a multifactorial disease associated with impaired nitric oxide (NO) production and bioavailability. In this respect, restoring NO activity by using nitrite and nitrate has been considered a potential therapeutic strategy to treat hypertension. This possibility is justified by the understanding that both nitrite and nitrate may be recycled back to NO and also promote the generation of other bioactive species. This process involves a complex biological circuit known as the enterosalivary cycle of nitrate, where this anion is actively taken up by the salivary glands and converted to nitrite by nitrate-reducing bacteria in the oral cavity. Nitrite is then ingested and reduced to NO and other nitroso species under the acid conditions of the stomach, whereas reminiscent nitrite that escapes gastric reduction is absorbed systemically and can be converted into NO by nitrite-reductases in tissues. While there is no doubt that nitrite and nitrate exert antihypertensive effects, several agents can impair the blood pressure responses to these anions by disrupting the enterosalivary cycle of nitrate. These agents include dietary and smoking-derived thiocyanate, antiseptic mouthwash, proton pump inhibitors, ascorbate at high concentrations, and xanthine oxidoreductase inhibitors. In this article, we provide an overview of the physiological aspects of nitrite and nitrate bioactivation and the therapeutic potential of these anions in hypertension. We also discuss mechanisms by which agents counteracting the antihypertensive responses to nitrite and nitrate mediate their effects. These critical aspects should be taken into consideration when suggesting nitrate or nitrite-based therapies to patients.
Collapse
Affiliation(s)
- Gustavo H Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Lucas C Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
56
|
Oliveira-Paula GH, Tanus-Santos JE. Nitrite-stimulated Gastric Formation of S-nitrosothiols As An Antihypertensive Therapeutic Strategy. Curr Drug Targets 2019; 20:431-443. [DOI: 10.2174/1389450119666180816120816] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/24/2018] [Accepted: 08/07/2018] [Indexed: 12/14/2022]
Abstract
Hypertension is usually associated with deficient nitric oxide (NO) bioavailability, and therefore stimulating NO activity is an important antihypertensive strategy. Recently, many studies have shown that both nitrite and nitrate anions are not simple products of NO metabolism and indeed may be reduced back to NO. While enzymes with nitrite-reductase activity capable of generating NO from nitrite may contribute to antihypertensive effects of nitrite, another mechanism involving the generation of NO-related species in the stomach from nitrite has been validated. Under the acidic conditions of the stomach, nitrite generates NO-related species that form S-nitrosothiols. Conversely, drugs that increase gastric pH may impair the gastric formation of S-nitrosothiols, which may mediate antihypertensive effects of oral nitrite or nitrate. Therefore, it is now becoming clear that promoting gastric formation of S-nitrosothiols may result in effective antihypertensive responses, and this mechanism opens a window of opportunity in the therapy of hypertension. In this review, we discuss the recent studies supporting the gastric generation of S-nitrosothiols as a potential antihypertensive mechanism of oral nitrite. We also highlight some drugs that increase S-nitrosothiols bioavailability, which may also improve the responses to nitrite/nitrate therapy. This new approach may result in increased nitrosation of critical pharmacological receptors and enzymes involved in the pathogenesis of hypertension, which tend to respond less to their activators resulting in lower blood pressure.
Collapse
Affiliation(s)
- Gustavo H. Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jose E. Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
57
|
Lim YJ, Foo TC, Yeung AWS, Tu X, Ma Y, Hawkins CL, Witting PK, Jameson GNL, Terentis AC, Thomas SR. Human Indoleamine 2,3-Dioxygenase 1 Is an Efficient Mammalian Nitrite Reductase. Biochemistry 2019; 58:974-986. [PMID: 30585477 DOI: 10.1021/acs.biochem.8b01231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The heme enzyme indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the first reaction of l-tryptophan oxidation along the kynurenine pathway. IDO1 is a central immunoregulatory enzyme with important implications for inflammation, infectious disease, autoimmune disorders, and cancer. Here we demonstrate that IDO1 is a mammalian nitrite reductase capable of chemically reducing nitrite to nitric oxide (NO) under hypoxia. Ultraviolet-visible absorption and resonance Raman spectroscopy showed that incubation of dithionite-reduced, ferrous-IDO1 protein (FeII-IDO1) with nitrite under anaerobic conditions resulted in the time-dependent formation of an FeII-nitrosyl IDO1 species, which was inhibited by substrate l-tryptophan, dependent on the concentration of nitrite or IDO1, and independent of the concentration of the reductant, dithionite. The bimolecular rate constant for IDO1 nitrite reductase activity was determined as 5.4 M-1 s-1 (pH 7.4, 23 °C), which was comparable to that measured for myoglobin (3.6 M-1 s-1; pH 7.4, 23 °C), an efficient and biologically important mammalian heme-based nitrite reductase. IDO1 nitrite reductase activity was pH-dependent but differed with myoglobin in that it showed a reduced proton dependency at pH >7. Electron paramagnetic resonance studies measuring NO production showed that the conventional IDO1 dioxygenase reducing cofactors, ascorbate and methylene blue, enhanced IDO1's nitrite reductase activity and the time- and IDO1 concentration-dependent release of NO in a manner inhibited by l-tryptophan or the IDO inhibitor 1-methyl-l-tryptophan. These data identify IDO1 as an efficient mammalian nitrite reductase that is capable of generating NO under anaerobic conditions. IDO1's nitrite reductase activity may have important implications for the enzyme's biological actions when expressed within hypoxic tissues.
Collapse
Affiliation(s)
| | - Timothy C Foo
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | | | | | | - Clare L Hawkins
- Department of Biomedical Sciences , University of Copenhagen , Copenhagen N DK-2200 , Denmark
| | - Paul K Witting
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health , University of Sydney , Sydney , NSW 2006 , Australia
| | - Guy N L Jameson
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Parkville , VIC 3010 , Australia
| | - Andrew C Terentis
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | |
Collapse
|
58
|
Henrohn D, Björkstrand K, Lundberg JO, Granstam SO, Baron T, Ingimarsdóttir IJ, Hedenström H, Malinovschi A, Wernroth ML, Jansson M, Hedeland M, Wikström G. Effects of Oral Supplementation With Nitrate-Rich Beetroot Juice in Patients With Pulmonary Arterial Hypertension-Results From BEET-PAH, an Exploratory Randomized, Double-Blind, Placebo-Controlled, Crossover Study. J Card Fail 2018; 24:640-653. [PMID: 30244181 DOI: 10.1016/j.cardfail.2018.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND The nitrate-nitrite-nitric oxide (NO) pathway may represent a potential therapeutic target in patients with pulmonary arterial hypertension (PAH). We explored the effects of dietary nitrate supplementation, with the use of nitrate-rich beetroot juice (BRJ), in patients with PAH. METHODS AND RESULTS We prospectively studied 15 patients with PAH in an exploratory randomized, double-blind, placebo-controlled, crossover trial. The patients received nitrate-rich beetroot juice (∼16 mmol nitrate per day) and placebo in 2 treatment periods of 7 days each. The assessments included; exhaled NO and NO flow-independent parameters (alveolar NO and bronchial NO flux), plasma and salivary nitrate and nitrite, biomarkers and metabolites of the NO-system, N-terminal pro-B-type natriuretic peptide, echocardiography, ergospirometry, diffusing capacity of the lung for carbon monoxide, and the 6-minute walk test. Compared with placebo ingestion of BRJ resulted in increases in; fractional exhaled NO at all flow-rates, alveolar NO concentrations and bronchial NO flux, and plasma and salivary levels of nitrate and nitrite. Plasma ornithine levels decreased and indices of relative arginine availability increased after BRJ compared to placebo. A decrease in breathing frequency was observed during ergospirometry after BRJ. A tendency for an improvement in right ventricular function was observed after ingestion of BRJ. In addition a tendency for an increase in the peak power output to peak oxygen consumption ratio (W peak/VO2 peak) was observed, which became significant in patients reaching an increase of plasma nitrite >30% (responders). CONCLUSIONS BRJ administered for 1 week increases pulmonary NO production and the relative arginine bioavailability in patients with PAH, compared with placebo. An increase in the W peak/VO2 peak ratio was observed after BRJ ingestion in plasma nitrite responders. These findings indicate that supplementation with inorganic nitrate increase NO synthase-independent NO production from the nitrate-nitrite-NO pathway.
Collapse
Affiliation(s)
- Dan Henrohn
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden.
| | - Kristoffer Björkstrand
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Olof Granstam
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| | - Tomasz Baron
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden; Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden; Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Inga J Ingimarsdóttir
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| | - Hans Hedenström
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| | - Andrei Malinovschi
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| | - Mona-Lisa Wernroth
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Martin Jansson
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| | - Mikael Hedeland
- Department of Chemistry, Environment and Feed Hygiene, National Veterinary Institute, (SVA), Uppsala, Sweden; Division of Analytical Pharmaceutical Chemistry, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Gerhard Wikström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
59
|
Tropea T, Wareing M, Greenwood SL, Feelisch M, Sibley CP, Cottrell EC. Nitrite mediated vasorelaxation in human chorionic plate vessels is enhanced by hypoxia and dependent on the NO-sGC-cGMP pathway. Nitric Oxide 2018; 80:82-88. [PMID: 30179715 PMCID: PMC6199414 DOI: 10.1016/j.niox.2018.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/05/2018] [Accepted: 08/30/2018] [Indexed: 11/25/2022]
Abstract
Adequate perfusion of the placental vasculature is essential to meet the metabolic demands of fetal growth and development. Lacking neural control, local tissue metabolites, circulating and physical factors contribute significantly to blood flow regulation. Nitric oxide (NO) is a key regulator of fetoplacental vascular tone. Nitrite, previously considered an inert end-product of NO oxidation, has been shown to provide an important source of NO. Reduction of nitrite to NO may be particularly relevant in tissue when the oxygen-dependent NO synthase (NOS) activity is compromised, e.g. in hypoxia. The contribution of this pathway in the placenta is currently unknown. We hypothesised that nitrite vasodilates human placental blood vessels, with enhanced efficacy under hypoxia. Placentas were collected from uncomplicated pregnancies and the vasorelaxant effect of nitrite (10-6-5x10-3 M) was assessed using wire myography on isolated pre-constricted chorionic plate arteries (CPAs) and veins (CPVs) under normoxic (pO2 ∼5%) and hypoxic (pO2 ∼1%) conditions. The dependency on the NO-sGC-cGMP pathway and known nitrite reductase (NiR) activities was also investigated. Nitrite caused concentration-dependent vasorelaxation in both arteries and veins, and this effect was enhanced by hypoxia, significantly in CPVs (P < 0.01) and with a trend in CPAs (P = 0.054). Pre-incubation with NO scavengers (cPTIO and oxyhemoglobin) attenuated (P < 0.01 and P < 0.0001, respectively), and the sGC inhibitor ODQ completely abolished nitrite-mediated vasorelaxation, confirming the involvement of NO and sGC. Inhibition of potential NiR enzymes xanthine oxidoreductase, mitochondrial aldehyde dehydrogenase and mitochondrial bc1 complex did not attenuate vasorelaxation. This data suggests that nitrite may provide an important reservoir of NO bioactivity within the placenta to enhance blood flow when fetoplacental oxygenation is impaired, as occurring in pregnancy diseases such as pre-eclampsia and fetal growth restriction.
Collapse
Affiliation(s)
- Teresa Tropea
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom.
| | - Mark Wareing
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom
| | - Susan L Greenwood
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital and Institute for Life Sciences, University of Southampton, United Kingdom
| | - Colin P Sibley
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom
| | - Elizabeth C Cottrell
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom
| |
Collapse
|
60
|
Craig JC, Broxterman RM, Smith JR, Allen JD, Barstow TJ. Effect of dietary nitrate supplementation on conduit artery blood flow, muscle oxygenation, and metabolic rate during handgrip exercise. J Appl Physiol (1985) 2018; 125:254-262. [DOI: 10.1152/japplphysiol.00772.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Dietary nitrate supplementation has positive effects on mitochondrial and muscle contractile efficiency during large muscle mass exercise in humans and on skeletal muscle blood flow (Q̇) in rats. However, concurrent measurement of these effects has not been performed in humans. Therefore, we assessed the influence of nitrate supplementation on Q̇ and muscle oxygenation characteristics during moderate- (40 %peak) and severe-intensity(85% peak) handgrip exercise in a randomized, double-blind, crossover design. Nine healthy men (age: 25 ± 2 yr) completed four constant-power exercise tests (2/intensity) randomly assigned to condition [nitrate-rich (nitrate) or nitrate-poor (placebo) beetroot supplementation] and intensity (40 or 85% peak). Resting mean arterial pressure was lower after nitrate compared with placebo (84 ± 4 vs. 89 ± 4 mmHg, P < 0.01). All subjects were able to sustain 10 min of exercise at 40% peak in both conditions. Nitrate had no effect on exercise tolerance during 85% peak (nitrate: 358 ± 29; placebo: 341 ± 34 s; P = 0.3). Brachial artery Q̇ was not different after nitrate at rest or any time during exercise. Deoxygenated [hemoglobin + myoglobin] was not different for 40% peak ( P > 0.05) but was elevated throughout 85% peak ( P < 0.05) after nitrate. The metabolic cost (V̇o2) was not different at the end of exercise; however, the V̇o2 primary amplitude at the onset of exercise was elevated after nitrate for the 85% peak work rate (96 ± 20 vs. 72 ± 12 ml/min, P < 0.05) and had a faster response. These findings suggest that an acute dose of nitrate reduces resting blood pressure and speeds V̇o2 kinetics in young adults but does not augment Q̇ or reduce steady-state V̇o2 during small muscle mass handgrip exercise. NEW & NOTEWORTHY We show that acute dietary nitrate supplementation via beetroot juice increases the amplitude and speed of local muscle V̇o2 on kinetics parameters during severe- but not moderate-intensity handgrip exercise. These changes were found in the absence of an increased blood flow response, suggesting that the increased V̇o2 was attained via improvements in fractional O2 extraction and/or spatial distribution of blood flow within the exercising muscle.
Collapse
Affiliation(s)
- Jesse C. Craig
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
| | - Ryan M. Broxterman
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas
| | - Joshua R. Smith
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
| | - Jason D. Allen
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
- Department of Kinesiology, University of Virginia, Charlottesville, Virginia
| | - Thomas J. Barstow
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
| |
Collapse
|
61
|
Van Acker ZP, Luyckx E, Dewilde S. Neuroglobin Expression in the Brain: a Story of Tissue Homeostasis Preservation. Mol Neurobiol 2018; 56:2101-2122. [DOI: 10.1007/s12035-018-1212-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 06/26/2018] [Indexed: 12/19/2022]
|
62
|
Kashfi K. The dichotomous role of H 2S in cancer cell biology? Déjà vu all over again. Biochem Pharmacol 2018; 149:205-223. [PMID: 29397935 PMCID: PMC5866221 DOI: 10.1016/j.bcp.2018.01.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/17/2018] [Indexed: 02/09/2023]
Abstract
Nitric oxide (NO) a gaseous free radical is one of the ten smallest molecules found in nature, while hydrogen sulfide (H2S) is a gas that bears the pungent smell of rotten eggs. Both are toxic yet they are gasotransmitters of physiological relevance. There appears to be an uncanny resemblance between the general actions of these two gasotransmitters in health and disease. The role of NO and H2S in cancer has been quite perplexing, as both tumor promotion and inflammatory activities as well as anti-tumor and antiinflammatory properties have been described. These paradoxes have been explained for both gasotransmitters in terms of each having a dual or biphasic effect that is dependent on the local flux of each gas. In this review/commentary, I have discussed the major roles of NO and H2S in carcinogenesis, evaluating their dual nature, focusing on the enzymes that contribute to this paradox and evaluate the pros and cons of inhibiting or inducing each of these enzymes.
Collapse
Affiliation(s)
- Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA.
| |
Collapse
|
63
|
Liu T, Zhang M, Terry MH, Schroeder H, Wilson SM, Power GG, Li Q, Tipple TE, Borchardt D, Blood AB. Nitrite potentiates the vasodilatory signaling of S-nitrosothiols. Nitric Oxide 2018; 75:60-69. [PMID: 29428841 DOI: 10.1016/j.niox.2018.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 12/27/2022]
Abstract
Nitrite and S-nitrosothiols (SNOs) are both byproducts of nitric oxide (NO) metabolism and are proposed to cause vasodilation via activation of soluble guanylate cyclase (sGC). We have previously reported that while SNOs are potent vasodilators at physiological concentrations, nitrite itself only produces vasodilation at supraphysiological concentrations. Here, we tested the hypothesis that sub-vasoactive concentrations of nitrite potentiate the vasodilatory effects of SNOs. Multiple exposures of isolated sheep arteries to S-nitroso-glutathione (GSNO) resulted in a tachyphylactic decreased vasodilatory response to GSNO but not to NO, suggesting attenuation of signaling steps upstream from sGC. Exposure of arteries to 1 μM nitrite potentiated the vasodilatory effects of GSNO in naive arteries and abrogated the tachyphylactic response to GSNO in pre-exposed arteries, suggesting that nitrite facilitates GSNO-mediated activation of sGC. In intact anesthetized sheep and rats, inhibition of NO synthases to decrease plasma nitrite levels attenuated vasodilatory responses to exogenous infusions of GSNO, an effect that was reversed by exogenous infusion of nitrite at sub-vasodilating levels. This study suggests nitrite potentiates SNO-mediated vasodilation via a mechanism that lies upstream from activation of sGC.
Collapse
Affiliation(s)
- Taiming Liu
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Meijuan Zhang
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Michael H Terry
- Department of Respiratory Care, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Hobe Schroeder
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Sean M Wilson
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Gordon G Power
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Qian Li
- Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Trent E Tipple
- Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Dan Borchardt
- Department of Chemistry, University of California, Riverside, CA 92521, United States
| | - Arlin B Blood
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States; Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States.
| |
Collapse
|
64
|
Forgan LG, Sofele M, McNeill BA, Cameron MS, Donald JA. Vasoactivity of nitrite in the iliac artery of the toad Rhinella marina. Am J Physiol Regul Integr Comp Physiol 2018; 314:R242-R251. [PMID: 29046317 DOI: 10.1152/ajpregu.00315.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nitrite ([Formula: see text]) causes vasodilation in mammals due to the formation of (nitric oxide) NO by endogenous [Formula: see text] reduction in the vascular wall. In this study, we determined if a similar mechanism operates in amphibians. Dual-wire myography of the iliac artery from Rhinella marina showed that applied [Formula: see text] caused a concentration-dependent vasodilation in normoxia (21% O2; EC50: 438 µM). Hypoxia (0.63% O2) significantly increased the maximal dilation to [Formula: see text] by 5% ( P = 0.0398). The addition of oxyhemoglobin significantly increased the EC50 ( P = 0.0144; EC50: 2,236 µM) but did not affect the maximal vasodilation. In contrast, partially deoxygenated hemoglobin (90% desaturation) did not affect the EC50 ( P = 0.1189) but significantly ( P = 0.0012) increased the maximal dilation to [Formula: see text] by 11%. The soluble guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) completely abolished the response to [Formula: see text] ( P < 0.0001), and of the nitric oxide synthase inhibitors, only N5-(1-imino-3-butenyl)-l-ornithine (vinyl-l-NIO; P = 0.0028) significantly reduced the [Formula: see text] vasodilation. The xanthine oxidoreductase inhibitor allopurinol ( P = 0.927), the nitric oxide-scavenger 2-(4-carboxyphenyl)-4,5-dihydro-4,4,5,5-tetramethyl-1H-imidazolyl-1-oxy-3-oxide (C-PTIO; P = 0.478), and disruption of the endothelium ( P = 0.094) did not affect the [Formula: see text] vasodilation. Incubation of iliac arteries with 1 mM [Formula: see text] did not a cause a change in the cGMP concentration (P = 0.407). Plasma [Formula: see text] was found to be 0.86 ± 0.20 µmol/l, while nitrate ([Formula: see text]) was 19.55 ± 2.55 µmol/l. Both cygb and ngb mRNAs were expressed in the iliac artery, and it is possible that these globins facilitate [Formula: see text] reduction in hypoxia. In addition, [Formula: see text] intracellular disproportionation processes could be important in the generation of NO from [Formula: see text].
Collapse
Affiliation(s)
- Leonard G Forgan
- School of Life and Environmental Sciences, Deakin University , Geelong , Australia
| | - Melenaite Sofele
- School of Life and Environmental Sciences, Deakin University , Geelong , Australia
| | | | - Melissa S Cameron
- School of Life and Environmental Sciences, Deakin University , Geelong , Australia
| | - John A Donald
- School of Life and Environmental Sciences, Deakin University , Geelong , Australia
| |
Collapse
|
65
|
Totzeck M, Mincu RI, Mrotzek S, Schadendorf D, Rassaf T. Cardiovascular diseases in patients receiving small molecules with anti-vascular endothelial growth factor activity: A meta-analysis of approximately 29,000 cancer patients. Eur J Prev Cardiol 2018; 25:482-494. [DOI: 10.1177/2047487318755193] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Targeted therapy with tyrosine kinase inhibitors with anti-vascular endothelial growth factor activity improves survival of cancer patients. Cardiovascular complications are critical and it is unknown whether these require specific treatment strategies. We aimed to clarify the associated risk of cardiovascular adverse events in patients treated with tyrosine kinase inhibitors. Design The design of this study was a meta-analysis of randomised controlled trials. Methods We searched PubMed, Cochrane, EMBASE and Web of Science databases for randomised controlled trials published until January 2017 that assessed patients with different types of cancer treated with or without tyrosine kinase inhibitors in addition to standard chemotherapy. Results A total of 29,252 patients from 71 randomised controlled trials were included. Tyrosine kinase inhibitor treatment was associated with a higher cardiac ischaemia relative risk (relative risk = 1.69; 95% confidence interval: 1.12–2.57; p = 0.01), with the highest risks observed for sorafenib and patients with renal cancer. Risk of thrombocytopaenia (relative risk = 2.2; 95% confidence interval: 1.73–2.79; p < 0.001) was highest for regorafenib and patients with breast cancer. Left ventricular systolic dysfunction was increased after tyrosine kinase inhibitor therapy (relative risk = 2.53; 95% confidence interval:1.79 – 3.57; p < 0.001), with the highest risks reported for sunitinib and hepatocellular cancer. QT corrected interval prolongation (relative risk = 6.25; 95% confidence interval: 3.44–11.38; p < 0.001) and arterial hypertension (relative risk = 3.78; 95% confidence interval: 3.15-4.54; p < 0.001) were reported. The relative risks of arterial adverse events, cerebral ischaemia, venous adverse events and pulmonary embolism were similar across groups. Conclusion Tyrosine kinase inhibitors increase the risk of severe cardiovascular and particularly thrombotic adverse events. Specific treatment regimens when prescribing tyrosine kinase inhibitor therapies appear desirable.
Collapse
Affiliation(s)
- Matthias Totzeck
- Department of Cardiology and Vascular Medicine, University Hospital Essen, Germany
| | - Raluca-Ileana Mincu
- Department of Cardiology and Vascular Medicine, University Hospital Essen, Germany
| | - Simone Mrotzek
- Department of Cardiology and Vascular Medicine, University Hospital Essen, Germany
| | | | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, University Hospital Essen, Germany
| |
Collapse
|
66
|
Angiotensin-(1-7)-induced Mas receptor activation attenuates atherosclerosis through a nitric oxide-dependent mechanism in apolipoproteinE-KO mice. Pflugers Arch 2018; 470:661-667. [PMID: 29352340 DOI: 10.1007/s00424-018-2108-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 01/15/2023]
Abstract
Angiotensin (Ang)-(1-7) ameliorates vascular injury by increasing nitric oxide (NO) bioavailability. Evidence that Ang-(1-7) attenuates the development of atherosclerosis through a NO-dependent mechanism is still missing. Moreover, it has been postulated that Ang-(1-7) may mediate its effects by other mechanisms than Mas receptor activation. To investigate Ang-(1-7)-dependent Mas receptor function, we treated apoE-KO and apoE/Mas-KO mice chronically with Ang-(1-7) (82 μg/kg per hour) or saline for 6 weeks. Flow-mediated dilation (FMD), a measure for NO-dependent vasodilation and the most accepted prognostic marker for the development of atherosclerosis, was measured in vivo. Chronic Ang-(1-7) treatment improved FMD and attenuated the development of atherosclerosis in apolipoproteinE (apoE)-KO but not in apoE/Mas-KO mice. These effects were accompanied by increased aortic nitrite and cGMP levels. To test whether Ang-(1-7) modulates atherosclerosis through a NO-dependent mechanism, apoE-KO mice were treated with the NO synthase inhibitor L-NAME (20 mg/kg/day) in the presence or absence of Ang-(1-7). L-NAME treatment reduced aortic nitrite content and increased blood pressure and exaggerated atherosclerosis compared to untreated apoE-KO mice. In L-NAME-treated apoE-KO mice, chronic Ang-(1-7) treatment did not increase aortic nitrite content and consequently showed no effect on blood pressure and the development of atherosclerosis. The present study proves that Ang-(1-7) mediates its protective vascular effects through Mas receptor activation. Moreover, Ang-(1-7)-mediated NO generation is essential for improving vascular function and prevents atherosclerosis in apoE-KO mice.
Collapse
|
67
|
Abstract
Nitric oxide (NO) is a cellular signalling molecule widely conserved among organisms, including microorganisms such as bacteria, yeasts, and fungi, and higher eukaryotes such as plants and mammals. NO is mainly produced by the activities of NO synthase (NOS) or nitrite reductase (NIR). There are several NO detoxification systems, including NO dioxygenase (NOD) and S-nitrosoglutathione reductase (GSNOR). NO homeostasis, based on the balance between NO synthesis and degradation, is important for regulating its physiological functions, since an excess of NO causes nitrosative stress due to the high reactivity of NO and NO-derived compounds. In yeast, NO may be involved in stress responses, but the role of NO and the mechanism underlying NO signalling are poorly understood due to the lack of mammalian NOS orthologs in the yeast genome. NOS and NIR activities have been observed in yeast cells, but the gene-encoding NOS and the mechanism by which NO production is catalysed by NIR remain unclear. On the other hand, yeast cells employ NOD and GSNOR to maintain intracellular redox balance following endogenous NO production, treatment with exogenous NO, or exposure to environmental stresses. This article reviews NO metabolism (synthesis, degradation) and its regulation in yeast. The physiological roles of NO in yeast, including the oxidative stress response, are also discussed. Such investigations into NO signalling are essential for understanding how NO modulates the genetics and physiology of yeast. In addition to being responsible for the pathology and pharmacology of various degenerative diseases, NO signalling may be a potential target for the construction and engineering of industrial yeast strains.
Collapse
|
68
|
Mouse cardiac mitochondria do not separate in subsarcolemmal and interfibrillar subpopulations. Mitochondrion 2018; 38:1-5. [DOI: 10.1016/j.mito.2017.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 06/30/2017] [Accepted: 07/13/2017] [Indexed: 11/19/2022]
|
69
|
Buerk DG, Liu Y, Zaccheo KA, Barbee KA, Jaron D. Nitrite-Mediated Hypoxic Vasodilation Predicted from Mathematical Modeling and Quantified from in Vivo Studies in Rat Mesentery. Front Physiol 2017; 8:1053. [PMID: 29321744 PMCID: PMC5733546 DOI: 10.3389/fphys.2017.01053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/01/2017] [Indexed: 12/26/2022] Open
Abstract
Nitric oxide (NO) generated from nitrite through nitrite reductase activity in red blood cells has been proposed to play a major role in hypoxic vasodilation. However, we have previously predicted from mathematical modeling that much more NO can be derived from tissue nitrite reductase activity than from red blood cell nitrite reductase activity. Evidence in the literature suggests that tissue nitrite reductase activity is associated with xanthine oxidoreductase (XOR) and/or aldehyde oxidoreductase (AOR). We investigated the role of XOR and AOR in nitrite-mediated vasodilation from computer simulations and from in vivo exteriorized rat mesentery experiments. Vasodilation responses to nitrite in the superfusion medium bathing the mesentery equilibrated with 5% O2 (normoxia) or zero O2 (hypoxia) at either normal or acidic pH were quantified. Experiments were also conducted following intraperitoneal (IP) injection of nitrite before and after inhibiting XOR with allopurinol or inhibiting AOR with raloxifene. Computer simulations for NO and O2 transport using reaction parameters reported in the literature were also conducted to predict nitrite-dependent NO production from XOR and AOR activity as a function of nitrite concentration, PO2 and pH. Experimentally, the largest arteriolar responses were found with nitrite >10 mM in the superfusate, but no statistically significant differences were found with hypoxic and acidic conditions in the superfusate. Nitrite-mediated vasodilation with IP nitrite injections was reduced or abolished after inhibiting XOR with allopurinol (p < 0.001). Responses to IP nitrite before and after inhibiting AOR with raloxifene were not as consistent. Our mathematical model predicts that under certain conditions, XOR and AOR nitrite reductase activity in tissue can significantly elevate smooth muscle cell NO and can serve as a compensatory pathway when endothelial NO production is limited by hypoxic conditions. Our theoretical and experimental results provide further evidence for a role of tissue nitrite reductases to contribute additional NO to compensate for reduced NO production by endothelial nitric oxide synthase during hypoxia. Our mathematical model demonstrates that under extreme hypoxic conditions with acidic pH, endogenous nitrite levels alone can be sufficient for a functionally significant increase in NO bioavailability. However, these conditions are difficult to achieve experimentally.
Collapse
Affiliation(s)
- Donald G Buerk
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Yien Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Kelly A Zaccheo
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Dov Jaron
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
70
|
Abstract
BACKGROUND Nitrite has been shown to reduce right ventricle (RV) remodeling in experimental pulmonary hypertension. However, whether this effect is due to a reduction in RV afterload (ie, reduction in pulmonary artery pressure) or a direct effect on the RV itself remains unanswered. We hypothesize that nitrite has direct effects on RV remodeling and studied its effects in mice with pulmonary artery banding (PAB). METHODS AND RESULTS PAB decreased exercise tolerance and reduced RV systolic and diastolic function. Nitrite treatment attenuated the decrease in RV systolic function and improved the RV diastolic function. Nitrite-treated mice with PAB had similar exercise tolerance compared with a control group. PAB induced RV hypertrophy and fibrosis which were associated with increased expression of phospho-Akt. Interestingly, nitrite treatment attenuated PAB-induced RV hypertrophy and reduced the expression of phospho-Akt in RV tissue from mice with PAB. In neonatal rat cardiac fibroblast, nitrite also attenuated hypoxia-induced increase in expression of phospho-Akt. CONCLUSION Our study indicates that nitrite treatment has direct beneficial effects on RV and improves function and attenuates remodeling in RV exposed to chronic pressure overload. These beneficial effects, at least in part, could be due to the inhibition of the phospho-Akt (p-Akt) pathway activation.
Collapse
|
71
|
Cortese-Krott MM, Koning A, Kuhnle GG, Nagy P, Bianco CL, Pasch A, Wink DA, Fukuto JM, Jackson AA, van Goor H, Olson KR, Feelisch M. The Reactive Species Interactome: Evolutionary Emergence, Biological Significance, and Opportunities for Redox Metabolomics and Personalized Medicine. Antioxid Redox Signal 2017; 27:684-712. [PMID: 28398072 PMCID: PMC5576088 DOI: 10.1089/ars.2017.7083] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 04/10/2017] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Oxidative stress is thought to account for aberrant redox homeostasis and contribute to aging and disease. However, more often than not, administration of antioxidants is ineffective, suggesting that our current understanding of the underlying regulatory processes is incomplete. Recent Advances: Similar to reactive oxygen species and reactive nitrogen species, reactive sulfur species are now emerging as important signaling molecules, targeting regulatory cysteine redox switches in proteins, affecting gene regulation, ion transport, intermediary metabolism, and mitochondrial function. To rationalize the complexity of chemical interactions of reactive species with themselves and their targets and help define their role in systemic metabolic control, we here introduce a novel integrative concept defined as the reactive species interactome (RSI). The RSI is a primeval multilevel redox regulatory system whose architecture, together with the physicochemical characteristics of its constituents, allows efficient sensing and rapid adaptation to environmental changes and various other stressors to enhance fitness and resilience at the local and whole-organism level. CRITICAL ISSUES To better characterize the RSI-related processes that determine fluxes through specific pathways and enable integration, it is necessary to disentangle the chemical biology and activity of reactive species (including precursors and reaction products), their targets, communication systems, and effects on cellular, organ, and whole-organism bioenergetics using system-level/network analyses. FUTURE DIRECTIONS Understanding the mechanisms through which the RSI operates will enable a better appreciation of the possibilities to modulate the entire biological system; moreover, unveiling molecular signatures that characterize specific environmental challenges or other forms of stress will provide new prevention/intervention opportunities for personalized medicine. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Miriam M. Cortese-Krott
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Anne Koning
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gunter G.C. Kuhnle
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Peter Nagy
- Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary
| | | | - Andreas Pasch
- Department of Clinical Chemistry, University of Bern and Calciscon AG, Bern, Switzerland
| | - David A. Wink
- Cancer and Inflammation Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Jon M. Fukuto
- Department of Chemistry, Sonoma State University, Rohnert Park, California
| | - Alan A. Jackson
- NIHR Southampton Biomedical Research Center, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Kenneth R. Olson
- Indiana University School of Medicine-South Bend, South Bend, Indiana
| | - Martin Feelisch
- NIHR Southampton Biomedical Research Center, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital and Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
72
|
Impact of mitochondrial nitrite reductase on hemodynamics and myocardial contractility. Sci Rep 2017; 7:12092. [PMID: 28935964 PMCID: PMC5608763 DOI: 10.1038/s41598-017-11531-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/22/2017] [Indexed: 01/11/2023] Open
Abstract
Inorganic nitrite (NO2−) can be reduced back to nitric oxide (NO) by several heme proteins called nitrite reductases (NR) which affect both the vascular tonus and hemodynamics. The objective of this study was to clarify the impact of several NRs on the regulation of hemodynamics, for which hemodynamic parameters such as heart rate, blood pressure, arterial stiffness, peripheral resistance and myocardial contractility were characterized by pulse wave analysis. We have demonstrated that NO2− reduced to NO in RBCs predominantly influences the heart rate, while myoglobin (Mb) and mitochondria-derived NO regulates arterial stiffness, peripheral resistance and myocardial contractility. Using ex vivo on-line NO-detection, we showed that Mb is the strongest NR occurring in heart, which operates sufficiently only at very low oxygen levels. In contrast, mitochondrial NR operates under both hypoxia and normoxia. Additional experiments with cardiomyocytes suggested that only mitochondria-derived generation of NO regulates cGMP levels mediating the contractility of cardiomyocytes. Our data suggest that a network of NRs is involved in NO2− mediated regulation of hemodynamics. Oxygen tension and hematocrit define the activity of specific NRs.
Collapse
|
73
|
Jourd'heuil FL, Xu H, Reilly T, McKellar K, El Alaoui C, Steppich J, Liu YF, Zhao W, Ginnan R, Conti D, Lopez-Soler R, Asif A, Keller RK, Schwarz JJ, Thanh Thuy LT, Kawada N, Long X, Singer HA, Jourd'heuil D. The Hemoglobin Homolog Cytoglobin in Smooth Muscle Inhibits Apoptosis and Regulates Vascular Remodeling. Arterioscler Thromb Vasc Biol 2017; 37:1944-1955. [PMID: 28798140 DOI: 10.1161/atvbaha.117.309410] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/26/2017] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The role of hemoglobin and myoglobin in the cardiovascular system is well established, yet other globins in this context are poorly characterized. Here, we examined the expression and function of cytoglobin (CYGB) during vascular injury. APPROACH AND RESULTS We characterized CYGB content in intact vessels and primary vascular smooth muscle (VSM) cells and used 2 different vascular injury models to examine the functional significance of CYGB in vivo. We found that CYGB was strongly expressed in medial arterial VSM and human veins. In vitro and in vivo studies indicated that CYGB was lost after VSM cell dedifferentiation. In the rat balloon angioplasty model, site-targeted delivery of adenovirus encoding shRNA specific for CYGB prevented its reexpression and decreased neointima formation. Similarly, 4 weeks after complete ligation of the left common carotid, Cygb knockout mice displayed little to no evidence of neointimal hyperplasia in contrast to their wild-type littermates. Mechanistic studies in the rat indicated that this was primarily associated with increased medial cell loss, terminal uridine nick-end labeling staining, and caspase-3 activation, all indicative of prolonged apoptosis. In vitro, CYGB could be reexpressed after VSM stimulation with cytokines and hypoxia and loss of CYGB sensitized human and rat aortic VSM cells to apoptosis. This was reversed after antioxidant treatment or NOS2 (nitric oxide synthase 2) inhibition. CONCLUSIONS These results indicate that CYGB is expressed in vessels primarily in differentiated medial VSM cells where it regulates neointima formation and inhibits apoptosis after injury.
Collapse
Affiliation(s)
- Frances L Jourd'heuil
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Haiyan Xu
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Timothy Reilly
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Keneta McKellar
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Chaymae El Alaoui
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Julia Steppich
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Yong Feng Liu
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Wen Zhao
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Roman Ginnan
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - David Conti
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Reynold Lopez-Soler
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Arif Asif
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Rebecca K Keller
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - John J Schwarz
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Le Thi Thanh Thuy
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Norifumi Kawada
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Xiaochun Long
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - Harold A Singer
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.)
| | - David Jourd'heuil
- From the Department of Molecular and Cellular Physiology (F.L.J., H.X., T.R., K.M., C.E.A., J.S., Y.F.L., W.Z., R.G., R.K.K., J.J.S., X.L., H.A.S., D.J.) and Surgery Transplantation (D.C., R.L.-S.), Albany Medical Center, NY; Seton Hall-Hackensack Meridian School of Medicine, Jersey Shore University Medical Center, Hackensack-Meridian Health, Neptune, NJ (A.A.); and Department of Hepatology, Graduate School of Medicine, Osaka City University, Japan (L.T.T.T., N.K.).
| |
Collapse
|
74
|
Totzeck M, Mincu RI, Rassaf T. Cardiovascular Adverse Events in Patients With Cancer Treated With Bevacizumab: A Meta-Analysis of More Than 20 000 Patients. J Am Heart Assoc 2017; 6:JAHA.117.006278. [PMID: 28862931 PMCID: PMC5586462 DOI: 10.1161/jaha.117.006278] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background The monoclonal antibody bevacizumab effectively inhibits angiogenesis in several types of cancers by blocking vascular endothelial growth factor. However, life‐threatening cardiovascular adverse effects could limit its use and may warrant specific follow‐up strategies. Methods and Results We systematically searched MEDLINE, Cochrane, EMBASE, and Web of Science for randomized controlled trials published until November 2016 that assessed patients with cancer treated with or without bevacizumab in addition to standard chemotherapy. A total of 20 050 patients with a broad range of cancer types from 22 studies were included in this analysis (10 394 in the bevacizumab group and 9656 in the control group). The risks of arterial and venous adverse events were higher in the bevacizumab groups (relative risk [RR], 1.37; 95% CI, 1.10–1.70 [P=0.004] and RR, 1.29; 95% CI, 1.12–1.47 [P<0.001], respectively), and more arterial adverse events occurred in patients taking high‐dose bevacizumab regimens. Bevacizumab treatment was associated with the highest risk of cardiac and cerebral ischemia in the high‐dose bevacizumab groups (RR, 4.4; 95% CI, 1.59–12.70 [P=0.004] and RR, 6.67; 95% CI, 2.17–20.66 [P=0.001], respectively). In addition, the risk of bleeding and arterial hypertension were higher in the bevacizumab groups (RR, 2.74; 95% CI, 2.38–3.15 [P<0.001] and RR, 4.73; 95% CI, 4.15–5.39 [P<0.00001], respectively), with higher values for patiens taking high‐dose regimens. Conclusions Treatment with bevacizumab increases the risk of arterial adverse events, particularly cardiac and cerebral ischemia, venous adverse events, bleeding, and arterial hypertension. This risk is additionally increased with high doses of bevacizumab. Further studies should determine the appropriate options for cardio‐oncology management. Clinical Trial Registration URL: https://www.crd.york.ac.uk. Unique identifier: PROSPERO(CRD42016054305).
Collapse
Affiliation(s)
- Matthias Totzeck
- Department of Cardiology and Vascular Medicine, Medical Faculty, West German Heart and Vascular Center, University Hospital Essen, Essen, Germany
| | - Raluca Ileana Mincu
- Department of Cardiology and Vascular Medicine, Medical Faculty, West German Heart and Vascular Center, University Hospital Essen, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, Medical Faculty, West German Heart and Vascular Center, University Hospital Essen, Essen, Germany
| |
Collapse
|
75
|
Sakhaei Z, Kundu S, Donnelly JM, Bertke JA, Kim WY, Warren TH. Nitric oxide release via oxygen atom transfer from nitrite at copper(ii). Chem Commun (Camb) 2017; 53:549-552. [PMID: 27973637 DOI: 10.1039/c6cc08745k] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nitric oxide is a vital signaling molecule that controls blood flow and oxygenation and nitrite serves as an important reservoir for nitric oxide in biology. While copper containing enzymes are known to reduce nitrite to nitric oxide, herein we report a new pathway to release nitric oxide via oxygen atom transfer from nitrite at a copper(ii) site.
Collapse
Affiliation(s)
- Zeinab Sakhaei
- Department of Chemistry, Georgetown University, Washington, DC, USA.
| | - Subrata Kundu
- Department of Chemistry, Georgetown University, Washington, DC, USA.
| | - Jane M Donnelly
- Department of Chemistry, Georgetown University, Washington, DC, USA.
| | - Jeffery A Bertke
- Department of Chemistry, Georgetown University, Washington, DC, USA.
| | - William Y Kim
- Department of Chemistry, Georgetown University, Washington, DC, USA.
| | - Timothy H Warren
- Department of Chemistry, Georgetown University, Washington, DC, USA.
| |
Collapse
|
76
|
Totzeck M, Korste S, Miinalainen I, Hendgen-Cotta UB, Rassaf T. S -nitrosation of calpains is associated with cardioprotection in myocardial I/R injury. Nitric Oxide 2017; 67:68-74. [DOI: 10.1016/j.niox.2017.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/03/2017] [Accepted: 04/05/2017] [Indexed: 01/23/2023]
|
77
|
Erkens R, Suvorava T, Kramer CM, Diederich LD, Kelm M, Cortese-Krott MM. Modulation of Local and Systemic Heterocellular Communication by Mechanical Forces: A Role of Endothelial Nitric Oxide Synthase. Antioxid Redox Signal 2017; 26:917-935. [PMID: 27927026 PMCID: PMC5455615 DOI: 10.1089/ars.2016.6904] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this review, we discuss the role of nitric oxide (NO) as a key physiological mechanotransducer modulating both local and systemic heterocellular communication and contributing to the integrated (patho)physiology of the cardiovascular system. A deeper understanding of mechanotransduction-mediated local and systemic nodes controlling heterocellular communication between the endothelium, blood cells, and other cell types (e.g., cardiomyocytes) may suggest novel therapeutic strategies for endothelial dysfunction and cardiovascular disease. Recent Advances: Mechanical forces acting on mechanoreceptors on endothelial cells activate the endothelial NO synthase (eNOS) to produce NO. NO participates in (i) abluminal heterocellular communication, inducing vasorelaxation, and thereby regulating vascular tone and blood pressure; (ii) luminal heterocellular communication, inhibiting platelet aggregation, and controlling hemostasis; and (iii) systemic heterocellular communication, contributing to adaptive physiological processes in response to exercise and remote ischemic preconditioning. Interestingly, shear-induced eNOS-dependent activation of vascular heterocellular communication constitutes the molecular basis of all methods applied in the clinical routine for evaluation of endothelial function. Critical Issues and Future Directions: The integrated physiology of heterocellular communication is still not fully understood. Dedicated experimental models are needed to analyze messengers and mechanisms underpinning heterocellular communication in response to physical forces in the cardiovascular system (and elsewhere). Antioxid. Redox Signal. 26, 917-935.
Collapse
Affiliation(s)
- Ralf Erkens
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Tatsiana Suvorava
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Lukas D Diederich
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
78
|
Nitrite-Nitric Oxide Signaling and Cardioprotection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:335-346. [DOI: 10.1007/978-3-319-55330-6_18] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
79
|
Fernandez-Moure JS, Evangelopoulos M, Colvill K, Van Eps JL, Tasciotti E. Nanoantibiotics: a new paradigm for the treatment of surgical infection. Nanomedicine (Lond) 2017; 12:1319-1334. [PMID: 28520517 DOI: 10.2217/nnm-2017-0401] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Infections following orthopedic device implantations often impose a substantial health burden and result in high medical costs. Currently, preventative methods are often employed following an orthopedic implant to reduce risk of infection; however, contamination of the surgical site can still occur. Although antibiotics have demonstrated a substantial reduction in bacterial growth and maintenance, biofilm formation around the implant can often minimize efficacy of the antibiotic. Recently, nanotechnology has garnered significant interest, resulting in the development of several antibiotic delivery strategies that exhibit extended release and increased efficacy. In this review, treatment methods of orthopedic-device-related infections will be discussed and an overview of antimicrobial-based nanotechnologies will be provided. Specifically, nonmetal-, metal- and oxide-based nanotechnologies, incorporating antibacterial strategies, will be discussed.
Collapse
Affiliation(s)
- Joseph S Fernandez-Moure
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA.,Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | | | - Kayla Colvill
- University of Texas McGovern Medical School, Houston, TX, USA
| | - Jeffrey L Van Eps
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, USA.,Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | - Ennio Tasciotti
- Department of Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
80
|
Liu Y, Buerk DG, Barbee KA, Jaron D. Nitric oxide release by deoxymyoglobin nitrite reduction during cardiac ischemia: A mathematical model. Microvasc Res 2017; 112:79-86. [PMID: 28363495 DOI: 10.1016/j.mvr.2017.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/16/2017] [Accepted: 03/19/2017] [Indexed: 12/19/2022]
Abstract
Interactions between cardiac myoglobin (Mb), nitrite, and nitric oxide (NO) are vital in regulating O2 storage, transport, and NO homeostasis. Production of NO through the reduction of endogenous myocardial nitrite by deoxygenated myoglobin has been shown to significantly reduce myocardial infarction damage and ischemic injury. We developed a mathematical model for a cardiac arteriole and surrounding myocardium to examine the hypothesis that myoglobin switches functions from being a strong NO scavenger to an NO producer via the deoxymyoglobin nitrite reductase pathway. Our results predict that under ischemic conditions of flow, blood oxygen level, and tissue pH, deoxyMb nitrite reduction significantly elevates tissue and smooth muscle cell NO. The size of the effect is consistent at different flow rates, increases with decreasing blood oxygen and tissue pH and, in extreme pathophysiological conditions, NO can even be elevated above the normoxic levels. Our simulations suggest that cardiac deoxyMb nitrite reduction is a plausible mechanism for preserving or enhancing NO levels using endogenous nitrite despite the rate-limiting O2 levels for endothelial NO production. This NO could then be responsible for mitigating deleterious effects under ischemic conditions.
Collapse
Affiliation(s)
- Yien Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA.
| |
Collapse
|
81
|
Koch CD, Gladwin MT, Freeman BA, Lundberg JO, Weitzberg E, Morris A. Enterosalivary nitrate metabolism and the microbiome: Intersection of microbial metabolism, nitric oxide and diet in cardiac and pulmonary vascular health. Free Radic Biol Med 2017; 105:48-67. [PMID: 27989792 PMCID: PMC5401802 DOI: 10.1016/j.freeradbiomed.2016.12.015] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/18/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Recent insights into the bioactivation and signaling actions of inorganic, dietary nitrate and nitrite now suggest a critical role for the microbiome in the development of cardiac and pulmonary vascular diseases. Once thought to be the inert, end-products of endothelial-derived nitric oxide (NO) heme-oxidation, nitrate and nitrite are now considered major sources of exogenous NO that exhibit enhanced vasoactive signaling activity under conditions of hypoxia and stress. The bioavailability of nitrate and nitrite depend on the enzymatic reduction of nitrate to nitrite by a unique set of bacterial nitrate reductase enzymes possessed by specific bacterial populations in the mammalian mouth and gut. The pathogenesis of pulmonary hypertension (PH), obesity, hypertension and CVD are linked to defects in NO signaling, suggesting a role for commensal oral bacteria to shape the development of PH through the formation of nitrite, NO and other bioactive nitrogen oxides. Oral supplementation with inorganic nitrate or nitrate-containing foods exert pleiotropic, beneficial vascular effects in the setting of inflammation, endothelial dysfunction, ischemia-reperfusion injury and in pre-clinical models of PH, while traditional high-nitrate dietary patterns are associated with beneficial outcomes in hypertension, obesity and CVD. These observations highlight the potential of the microbiome in the development of novel nitrate- and nitrite-based therapeutics for PH, CVD and their risk factors.
Collapse
Affiliation(s)
- Carl D Koch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA.
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA
| | - Bruce A Freeman
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| |
Collapse
|
82
|
Cytosolic BNIP3 Dimer Interacts with Mitochondrial BAX Forming Heterodimers in the Mitochondrial Outer Membrane under Basal Conditions. Int J Mol Sci 2017; 18:ijms18040687. [PMID: 28333095 PMCID: PMC5412273 DOI: 10.3390/ijms18040687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 02/03/2023] Open
Abstract
The primary function of mitochondria is energy production, a task of particular importance especially for cells with a high energy demand like cardiomyocytes. The B-cell lymphoma (BCL-2) family member BCL-2 adenovirus E1B 19 kDa-interacting protein 3 (BNIP3) is linked to mitochondrial targeting after homodimerization, where it functions in inner membrane depolarization and permeabilization of the mitochondrial outer membrane (MOM) mediating cell death. We investigated the basal distribution of cardiac BNIP3 in vivo and its physical interaction with the pro-death protein BCL2 associated X, apoptosis regulator (BAX) and with mitochondria using immunoblot analysis, co-immunoprecipitation, and continuous wave and pulsed electron paramagnetic resonance spectroscopy techniques. We found that BNIP3 is present as a dimer in the cytosol and in the outer membrane of cardiac mitochondria under basal conditions. It forms disulfide-bridged, but mainly non-covalent dimers in the cytosol. Heterodimers with BAX are formed exclusively in the MOM. Furthermore, our results suggest that BNIP3 interacts with the MOM directly via mitochondrial BAX. However, the physical interactions with BAX and the MOM did not affect the membrane potential and cell viability. These findings suggest that another stimulus other than the mere existence of the BNIP3/BAX dimer in the MOM is required to promote BNIP3 cell-death activity; this could be a potential disturbance of the BNIP3 distribution homeostasis, namely in the direction of the mitochondria.
Collapse
|
83
|
Helms CC, Liu X, Kim-Shapiro DB. Recent insights into nitrite signaling processes in blood. Biol Chem 2017; 398:319-329. [PMID: 27611767 DOI: 10.1515/hsz-2016-0263] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 08/30/2016] [Indexed: 11/15/2022]
Abstract
Nitrite was once thought to be inert in human physiology. However, research over the past few decades has established a link between nitrite and the production of nitric oxide (NO) that is potentiated under hypoxic and acidic conditions. Under this new role nitrite acts as a storage pool for bioavailable NO. The NO so produced is likely to play important roles in decreasing platelet activation, contributing to hypoxic vasodilation and minimizing blood-cell adhesion to endothelial cells. Researchers have proposed multiple mechanisms for nitrite reduction in the blood. However, NO production in blood must somehow overcome rapid scavenging by hemoglobin in order to be effective. Here we review the role of red blood cell hemoglobin in the reduction of nitrite and present recent research into mechanisms that may allow nitric oxide and other reactive nitrogen signaling species to escape the red blood cell.
Collapse
|
84
|
Hendgen-Cotta UB, Esfeld S, Coman C, Ahrends R, Klein-Hitpass L, Flögel U, Rassaf T, Totzeck M. A novel physiological role for cardiac myoglobin in lipid metabolism. Sci Rep 2017; 7:43219. [PMID: 28230173 PMCID: PMC5322402 DOI: 10.1038/srep43219] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/20/2017] [Indexed: 02/06/2023] Open
Abstract
Continuous contractile activity of the heart is essential and the required energy is mostly provided by fatty acid (FA) oxidation. Myocardial lipid accumulation can lead to pathological responses, however the underlying mechanisms remain elusive. The role of myoglobin in dioxygen binding in cardiomyocytes and oxidative skeletal muscle has widely been appreciated. Our recent work established myoglobin as a protector of cardiac function in hypoxia and disease states. We here unravel a novel role of cardiac myoglobin in governing FA metabolism to ensure the physiological energy production through β-oxidation, preventing myocardial lipid accumulation and preserving cardiac functions. In vivo1H magnetic resonance spectroscopy unveils a 3-fold higher deposition of lipids in mouse hearts lacking myoglobin, which was associated with depressed cardiac function compared to wild-type hearts as assessed by echocardiography. Mass spectrometry reveals a marked increase in tissue triglycerides with preferential incorporation of palmitic and oleic acids. Phospholipid levels as well as the metabolome, transcriptome and proteome related to FA metabolism tend to be unaffected by myoglobin ablation. Our results reveal a physiological role of myoglobin in FA metabolism with the lipid accumulation-suppressing effects of myoglobin preventing cardiac lipotoxicity.
Collapse
Affiliation(s)
- Ulrike B Hendgen-Cotta
- University Hospital Essen, Medical Faculty, West German Heart and Vascular Center, Department of Cardiology and Department of Angiology, Hufelandstr. 55, 45147 Essen, Germany
| | - Sonja Esfeld
- University Hospital Essen, Medical Faculty, West German Heart and Vascular Center, Department of Cardiology and Department of Angiology, Hufelandstr. 55, 45147 Essen, Germany
| | - Cristina Coman
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V. Otto-Hahn-Str. 6b, 44227 Dortmund, Germany
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V. Otto-Hahn-Str. 6b, 44227 Dortmund, Germany
| | - Ludger Klein-Hitpass
- University Hospital Essen, Institute of Cell Biology, Medical Faculty, Virchowstr. 173, 45122 Essen, Germany
| | - Ulrich Flögel
- University Hospital Düsseldorf, Department of Molecular Cardiology, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Tienush Rassaf
- University Hospital Essen, Medical Faculty, West German Heart and Vascular Center, Department of Cardiology and Department of Angiology, Hufelandstr. 55, 45147 Essen, Germany
| | - Matthias Totzeck
- University Hospital Essen, Medical Faculty, West German Heart and Vascular Center, Department of Cardiology and Department of Angiology, Hufelandstr. 55, 45147 Essen, Germany
| |
Collapse
|
85
|
Jones DA, Rathod KS, Ahluwalia A. Update on Nitrite Reduction in Ischemic Disease: Mechanisms and Clinical Translation. NITRIC OXIDE 2017:195-211. [DOI: 10.1016/b978-0-12-804273-1.00015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
86
|
Schicho A, Hellerbrand C, Krüger K, Beyer LP, Wohlgemuth W, Niessen C, Hohenstein E, Stroszczynski C, Pereira PL, Wiggermann P. Impact of Different Embolic Agents for Transarterial Chemoembolization (TACE) Procedures on Systemic Vascular Endothelial Growth Factor (VEGF) Levels. J Clin Transl Hepatol 2016; 4:288-292. [PMID: 28097096 PMCID: PMC5225147 DOI: 10.14218/jcth.2016.00058] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/05/2016] [Accepted: 12/22/2016] [Indexed: 02/07/2023] Open
Abstract
Background and Aims: Intermediate stage hepatocellular carcinoma (HCC) can be treated by transarterial chemoembolization (TACE). However, there appear to be side effects, such as induction of proangiogenic factors, e.g. vascular endothelial growth factor (VEGF), which have been shown to be associated with a poor prognosis. This prospective study was designed to compare serum VEGF level response after TACE with different embolic agents in patients with HCC. Methods: Patients were assigned to one of three different TACE regimens: degradable starch microspheres (DSM) TACE, drug-eluting bead (DEBDOX) TACE or Lipiodol TACE (cTACE). All patients received 50 mg doxorubicin/m2 body surface area (BSA) during TACE. Serum VEGF levels were assessed before TACE treatment, 24 h post-treatment and 4 weeks later. Results: Twenty-two patients with 30 TACE treatments were enrolled. Compared to baseline VEGF levels, a marked increase was observed for 24 h post-TACE (164% of baseline level) and during the 4-week follow-up (170% of baseline level) only for the cTACE arm (p < 0.05). In contrast, the increase of serum VEGF levels were only 114% and 123% for DEBDOX and 121% and 124% for DSM, respectively. Conclusions: Conventional TACE using Lipiodol shows marked increase in blood levels of the proangiogenic factor VEGF, while DEBDOX and DSM TACE induce only a moderate VEGF response.
Collapse
Affiliation(s)
- Andreas Schicho
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
- *Correspondence to: Andreas Schicho, Department of Radiology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg D-93053, Germany. Tel: +49-941-944-7412, Fax: +49-941-944-7469, E-mail:
| | - Claus Hellerbrand
- Friedrich-Alexander University Erlangen-Nürnberg, Institute of Biochemistry (Emil-Fischer Zentrum), Erlangen, Germany
| | - Kristina Krüger
- Clinic for Radiology, Minimally-invasive Therapies and Nuclear Medicine, SLK Kliniken Heilbronn GmbH, Heilbronn, Germany
| | - Lukas P. Beyer
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Walter Wohlgemuth
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Christoph Niessen
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Ernst Hohenstein
- Clinic for Radiology, Minimally-invasive Therapies and Nuclear Medicine, SLK Kliniken Heilbronn GmbH, Heilbronn, Germany
| | | | - Philippe L. Pereira
- Clinic for Radiology, Minimally-invasive Therapies and Nuclear Medicine, SLK Kliniken Heilbronn GmbH, Heilbronn, Germany
| | - Philipp Wiggermann
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
87
|
Kuleva NV, Krasovskaya IE. A new role for myoglobin in cardiac and skeletal muscle function. Biophysics (Nagoya-shi) 2016. [DOI: 10.1134/s000635091605016x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
88
|
Totzeck M, Hendgen-Cotta UB, French BA, Rassaf T. A practical approach to remote ischemic preconditioning and ischemic preconditioning against myocardial ischemia/reperfusion injury. J Biol Methods 2016; 3. [PMID: 28066791 PMCID: PMC5218813 DOI: 10.14440/jbm.2016.149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although urgently needed in clinical practice, a cardioprotective therapeutic approach against myocardial ischemia/ reperfusion injury remains to be established. Remote ischemic preconditioning (rIPC) and ischemic preconditioning (IPC) represent promising tools comprising three entities: the generation of a protective signal, the transfer of the signal to the target organ, and the response to the transferred signal resulting in cardioprotection. However, in light of recent scientific advances, many controversies arise regarding the efficacy of the underlying signaling. We here show methods for the generation of the signaling cascade by rIPC as well as IPC in a mouse model for in vivo myocardial ischemia/ reperfusion injury using highly reproducible approaches. This is accomplished by taking advantage of easily applicable preconditioning strategies compatible with the clinical setting. We describe methods for using laser Doppler perfusion imaging to monitor the cessation and recovery of perfusion in real time. The effects of preconditioning on cardiac function can also be assessed using ultrasound or magnetic resonance imaging approaches. On a cellular level, we confirm how tissue injury can be monitored using histological assessment of infarct size in conjunction with immunohistochemistry to assess both aspects in a single specimen. Finally, we outline, how the rIPC-associated signaling can be transferred to the target cell via conservation of the signal in the humoral (blood) compartment. This compilation of experimental protocols including a conditioning regimen comparable to the clinical setting should proof useful to both beginners and experts in the field of myocardial infarction, supplying information for the detailed procedures as well as troubleshooting guides.
Collapse
Affiliation(s)
- Matthias Totzeck
- Department of Cardiology and Department of Angiology, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen 45147, Germany
| | - Ulrike B Hendgen-Cotta
- Department of Cardiology and Department of Angiology, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen 45147, Germany
| | - Brent A French
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| | - Tienush Rassaf
- Department of Cardiology and Department of Angiology, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen 45147, Germany
| |
Collapse
|
89
|
Nitric oxide signaling in yeast. Appl Microbiol Biotechnol 2016; 100:9483-9497. [DOI: 10.1007/s00253-016-7827-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/17/2016] [Accepted: 08/22/2016] [Indexed: 12/11/2022]
|
90
|
Chirinos JA, Zamani P. The Nitrate-Nitrite-NO Pathway and Its Implications for Heart Failure and Preserved Ejection Fraction. Curr Heart Fail Rep 2016; 13:47-59. [PMID: 26792295 DOI: 10.1007/s11897-016-0277-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathogenesis of exercise intolerance in patients with heart failure and preserved ejection fraction (HFpEF) is likely multifactorial. In addition to cardiac abnormalities (diastolic dysfunction, abnormal contractile reserve, chronotropic incompetence), several peripheral abnormalities are likely to be involved. These include abnormal pulsatile hemodynamics, abnormal arterial vasodilatory responses to exercise, and abnormal peripheral O2 delivery, extraction, and utilization. The nitrate-nitrite-NO pathway is emerging as a potential target to modify key physiologic abnormalities, including late systolic left ventricular (LV) load from arterial wave reflections (which has deleterious short- and long-term consequences for the LV), arterial vasodilatory reserve, muscle O2 delivery, and skeletal muscle mitochondrial function. In a recently completed randomized trial, the administration of a single dose of exogenous inorganic nitrate has been shown to exert various salutary arterial hemodynamic effects, ultimately leading to enhanced aerobic capacity in patients with HFpEF. These effects have the potential for both immediate improvements in exercise tolerance and for long-term "disease-modifying" effects. In this review, we provide an overview of key mechanistic contributors to exercise intolerance in HFpEF, and of the potential therapeutic role of drugs that target the nitrate-nitrite-NO pathway.
Collapse
Affiliation(s)
- Julio A Chirinos
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA. .,Hospital of the University of Pennsylvania, Philadelphia, PA, USA. .,Ghent University, Ghent, Belgium.
| | - Payman Zamani
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
91
|
Differential vascular reactivity responses acutely following ingestion of a nitrate rich red spinach extract. Eur J Appl Physiol 2016; 116:2267-2279. [DOI: 10.1007/s00421-016-3478-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/20/2016] [Indexed: 10/20/2022]
|
92
|
Grau M, Lauten A, Hoeppener S, Goebel B, Brenig J, Jung C, Bloch W, Suhr F. Regulation of red blood cell deformability is independent of red blood cell-nitric oxide synthase under hypoxia. Clin Hemorheol Microcirc 2016; 63:199-215. [DOI: 10.3233/ch-162044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Marijke Grau
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Germany
- The German Research Center of Elite Sport (momentum), German Sport University Cologne, Germany
| | - Alexander Lauten
- Department of Internal Medicine I (Cardiology, Angiology, Pneumology), Friedrich-Schiller University, Jena, Germany
| | - Steffen Hoeppener
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Germany
| | - Bjoern Goebel
- Department of Internal Medicine I (Cardiology, Angiology, Pneumology), Friedrich-Schiller University, Jena, Germany
| | - Julian Brenig
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Germany
| | - Christian Jung
- Department of Internal Medicine I (Cardiology, Angiology, Pneumology), Friedrich-Schiller University, Jena, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Germany
- The German Research Center of Elite Sport (momentum), German Sport University Cologne, Germany
| | - Frank Suhr
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Germany
- The German Research Center of Elite Sport (momentum), German Sport University Cologne, Germany
| |
Collapse
|
93
|
Shepherd AI, Wilkerson DP, Fulford J, Winyard PG, Benjamin N, Shore AC, Gilchrist M. Effect of nitrate supplementation on hepatic blood flow and glucose homeostasis: a double-blind, placebo-controlled, randomized control trial. Am J Physiol Gastrointest Liver Physiol 2016; 311:G356-64. [PMID: 27418682 PMCID: PMC5076007 DOI: 10.1152/ajpgi.00203.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/11/2016] [Indexed: 01/31/2023]
Abstract
Nitric oxide alters gastric blood flow, improves vascular function, and mediates glucose uptake within the intestines and skeletal muscle. Dietary nitrate, acting as a source of nitric oxide, appears to be a potential low-cost therapy that may help maintain glucose homeostasis. In a randomized, double-blind, placebo-controlled crossover study, 31 young and older adult participants had a standardized breakfast, supplemented with either nitrate-rich beetroot juice (11.91 mmol nitrate) or nitrate-depleted beetroot juice as placebo (0.01 mmol nitrate). MRI was used to assess apparent diffusion coefficient (ADC), portal vein flux, and velocity. Plasma glucose, incretin, and C-peptide concentrations and blood pressure were assessed. Outcome variables were measured at baseline and hourly for 3 h. Compared with a placebo, beetroot juice resulted in a significant elevation in plasma nitrate and plasma nitrite concentration. No differences were seen for the young or older adult cohorts between placebo and beetroot juice for ADC, or portal vein flux. There was an interaction effect in the young adults between visits for portal vein velocity. Nitrate supplementation did not reduce plasma glucose, active GLP-1, total GLP-1, or plasma C-peptide concentrations for the young or older adult cohorts. Despite a significant elevation in plasma nitrite concentration following an acute dose of (11.91 mmol) nitrate, there was no effect on hepatic blood flow, plasma glucose, C-peptide, or incretin concentration in healthy adults.
Collapse
Affiliation(s)
- Anthony I. Shepherd
- 1College of Life and Environmental Sciences, Sport and Health Sciences, University of Exeter, Devon, United Kingdom; ,2University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital, Exeter, Devon, United Kingdom; ,4Department of Sport and Exercise Science, Portsmouth, United Kingdom
| | - Daryl P. Wilkerson
- 1College of Life and Environmental Sciences, Sport and Health Sciences, University of Exeter, Devon, United Kingdom;
| | - Jon Fulford
- 2University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital, Exeter, Devon, United Kingdom;
| | - Paul G. Winyard
- 2University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital, Exeter, Devon, United Kingdom;
| | - Nigel Benjamin
- 2University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital, Exeter, Devon, United Kingdom; ,3Torbay Hospital, Heart and Lung Unit, Torquay, Devon, United Kingdom; and
| | - Angela C. Shore
- 2University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital, Exeter, Devon, United Kingdom;
| | - Mark Gilchrist
- 2University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital, Exeter, Devon, United Kingdom;
| |
Collapse
|
94
|
Globin X is a six-coordinate globin that reduces nitrite to nitric oxide in fish red blood cells. Proc Natl Acad Sci U S A 2016; 113:8538-43. [PMID: 27407144 DOI: 10.1073/pnas.1522670113] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The discovery of novel globins in diverse organisms has stimulated intense interest in their evolved function, beyond oxygen binding. Globin X (GbX) is a protein found in fish, amphibians, and reptiles that diverged from a common ancestor of mammalian hemoglobins and myoglobins. Like mammalian neuroglobin, GbX was first designated as a neuronal globin in fish and exhibits six-coordinate heme geometry, suggesting a role in intracellular electron transfer reactions rather than oxygen binding. Here, we report that GbX to our knowledge is the first six-coordinate globin and the first globin protein apart from hemoglobin, found in vertebrate RBCs. GbX is present in fish erythrocytes and exhibits a nitrite reduction rate up to 200-fold faster than human hemoglobin and up to 50-fold higher than neuroglobin or cytoglobin. Deoxygenated GbX reduces nitrite to form nitric oxide (NO) and potently inhibits platelet activation in vitro, to a greater extent than hemoglobin. Fish RBCs also reduce nitrite to NO and inhibit platelet activation to a greater extent than human RBCs, whereas GbX knockdown inhibits this nitrite-dependent NO signaling. The description of a novel, six-coordinate globin in RBCs with dominant electron transfer and nitrite reduction functionality provides new insights into the evolved signaling properties of ancestral heme-globins.
Collapse
|
95
|
Loudon BL, Noordali H, Gollop ND, Frenneaux MP, Madhani M. Present and future pharmacotherapeutic agents in heart failure: an evolving paradigm. Br J Pharmacol 2016; 173:1911-24. [PMID: 26993743 PMCID: PMC4882493 DOI: 10.1111/bph.13480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/28/2016] [Accepted: 02/26/2016] [Indexed: 02/06/2023] Open
Abstract
Many conditions culminate in heart failure (HF), a multi‐organ systemic syndrome with an intrinsically poor prognosis. Pharmacotherapeutic agents that correct neurohormonal dysregulation and haemodynamic instability have occupied the forefront of developments within the treatment of HF in the past. Indeed, multiple trials aimed to validate these agents in the 1980s and early 1990s, resulting in a large and robust evidence‐base supporting their use clinically. An established treatment paradigm now exists for the treatment of HF with reduced ejection fraction (HFrEF), but there have been very few notable developments in recent years. HF remains a significant health concern with an increasing incidence as the population ages. We may indeed be entering the surgical era for HF treatment, but these therapies remain expensive and inaccessible to many. Newer pharmacotherapeutic agents are slowly emerging, many targeting alternative therapeutic pathways, but with mixed results. Metabolic modulation and manipulation of the nitrate/nitrite/nitric oxide pathway have shown promise and could provide the answers to fill the therapeutic gap between medical interventions and surgery, but further definitive trials are warranted. We review the significant evidence base behind the current medical treatments for HFrEF, the physiology of metabolic impairment in HF, and discuss two promising novel agents, perhexiline and nitrite.
Collapse
Affiliation(s)
- Brodie L Loudon
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Hannah Noordali
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Nicholas D Gollop
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Michael P Frenneaux
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
96
|
Van Hoose PM, Kelm NQ, Piell KM, Cole MP. Conjugated linoleic acid and nitrite attenuate mitochondrial dysfunction during myocardial ischemia. J Nutr Biochem 2016; 34:8-16. [PMID: 27156147 DOI: 10.1016/j.jnutbio.2016.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/01/2016] [Accepted: 04/11/2016] [Indexed: 12/18/2022]
Abstract
Cardiovascular health is influenced by dietary composition and the western diet is composed of varying types/amounts of fat. Conjugated linoleic acid (cLA) is an abundant dietary unsaturated fatty acid associated with health benefits but its biological signaling is not well understood. Nitrite is enriched in vegetables within the diet and can impact signaling of unsaturated fatty acids; however, its role on cLA signaling is not well understood. Elucidating how nitrite may impact the biological signaling of cLA is important due to the dietary consumption of both cLA and nitrite in the western diet. Since co-administration of cLA and nitrite results in cardioprotection during myocardial infarction (MI), it was hypothesized that cLA and nitrite may affect cardiac mitochondrial respiratory function and complex activity in MI. C57BL/6J mice were treated with cLA and nitrite for either 10 or 13days, where MI was induced on day 3. Following treatment, respiration and complex activity were measured. Among the major findings of this study, cLA treatment (10days) decreases state 3 respiration in vivo. Following MI, nitrite alone and in combination with cLA attenuates increased state 3 respiration and decreases hydrogen peroxide levels. Further, nitrite and cLA co-treatment attenuates increased complex III activity after MI. These results suggest that cLA, nitrite and the combination significantly alter cardiac mitochondrial respiratory and electron transport chain activity in vivo and following MI. Overall, the daily consumption of cLA and nitrite in the diet can have diverse cardiovascular implications, some of which occur at the mitochondrial level.
Collapse
Affiliation(s)
- Patrick M Van Hoose
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Natia Qipshidze Kelm
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Kellianne M Piell
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Marsha P Cole
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
97
|
Wajih N, Liu X, Shetty P, Basu S, Wu H, Hogg N, Patel RP, Furdui CM, Kim-Shapiro DB. The role of red blood cell S-nitrosation in nitrite bioactivation and its modulation by leucine and glucose. Redox Biol 2016; 8:415-21. [PMID: 27156251 PMCID: PMC4864376 DOI: 10.1016/j.redox.2016.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 04/27/2016] [Accepted: 04/29/2016] [Indexed: 02/07/2023] Open
Abstract
Previous work has shown that red blood cells (RBCs) reduce nitrite to NO under conditions of low oxygen. Strong support for the ability of red blood cells to promote nitrite bioactivation comes from using platelet activation as a NO-sensitive process. Whereas addition of nitrite to platelet rich plasma in the absence of RBCs has no effect on inhibition of platelet activation, when RBCs are present platelet activation is inhibited by an NO-dependent mechanism that is potentiated under hypoxia. In this paper, we demonstrate that nitrite bioactivation by RBCs is blunted by physiologically-relevant concentrations of nutrients including glucose and the important signaling amino acid leucine. Our mechanistic investigations demonstrate that RBC mediated nitrite bioactivation is largely dependent on nitrosation of RBC surface proteins. These data suggest a new expanded paradigm where RBC mediated nitrite bioactivation not only directs blood flow to areas of low oxygen but also to areas of low nutrients. Our findings could have profound implications for normal physiology as well as pathophysiology in a variety of diseases including diabetes, sickle cell disease, and arteriosclerosis.
Collapse
Affiliation(s)
- Nadeem Wajih
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Xiaohua Liu
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Pragna Shetty
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Swati Basu
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA; Translational Science Center, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Hanzhi Wu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Neil Hogg
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Rakesh P Patel
- Department of Pathology, University of Alabama at Birmingham, 901 19th St. South, BMRII 532, Birmingham, AL 35294, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Daniel B Kim-Shapiro
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA; Translational Science Center, Wake Forest University, Winston-Salem, NC 27109, USA.
| |
Collapse
|
98
|
Sethumadhavan S, Whitsett J, Bennett B, Ionova IA, Pieper GM, Vasquez-Vivar J. Increasing tetrahydrobiopterin in cardiomyocytes adversely affects cardiac redox state and mitochondrial function independently of changes in NO production. Free Radic Biol Med 2016; 93:1-11. [PMID: 26826575 PMCID: PMC5498285 DOI: 10.1016/j.freeradbiomed.2016.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 01/25/2016] [Indexed: 02/07/2023]
Abstract
Tetrahydrobiopterin (BH4) represents a potential strategy for the treatment of cardiac remodeling, fibrosis and/or diastolic dysfunction. The effects of oral treatment with BH4 (Sapropterin™ or Kuvan™) are however dose-limiting with high dose negating functional improvements. Cardiomyocyte-specific overexpression of GTP cyclohydrolase I (mGCH) increases BH4 several-fold in the heart. Using this model, we aimed to establish the cardiomyocyte-specific responses to high levels of BH4. Quantification of BH4 and BH2 in mGCH transgenic hearts showed age-based variations in BH4:BH2 ratios. Hearts of mice (<6 months) have lower BH4:BH2 ratios than hearts of older mice while both GTPCH activity and tissue ascorbate levels were higher in hearts of young than older mice. No evident changes in nitric oxide (NO) production assessed by nitrite and endogenous iron-nitrosyl complexes were detected in any of the age groups. Increased BH4 production in cardiomyocytes resulted in a significant loss of mitochondrial function. Diminished oxygen consumption and reserve capacity was verified in mitochondria isolated from hearts of 12-month old compared to 3-month old mice, even though at 12 months an improved BH4:BH2 ratio is established. Accumulation of 4-hydroxynonenal (4-HNE) and decreased glutathione levels were found in the mGCH hearts and isolated mitochondria. Taken together, our results indicate that the ratio of BH4:BH2 does not predict changes in neither NO levels nor cellular redox state in the heart. The BH4 oxidation essentially limits the capacity of cardiomyocytes to reduce oxidant stress. Cardiomyocyte with chronically high levels of BH4 show a significant decline in redox state and mitochondrial function.
Collapse
Affiliation(s)
- Savitha Sethumadhavan
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jennifer Whitsett
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Brian Bennett
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Department of Physics, Marquette University, Milwaukee, 1250 W Wisconsin Ave, Milwaukee, WI 53233, USA
| | - Irina A Ionova
- Department of Surgery Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Galen M Pieper
- Department of Surgery Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jeannette Vasquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
99
|
Combined iron sucrose and protoporphyrin treatment protects against ischemic and toxin-mediated acute renal failure. Kidney Int 2016; 90:67-76. [PMID: 27165818 DOI: 10.1016/j.kint.2016.01.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/08/2016] [Accepted: 01/14/2016] [Indexed: 01/07/2023]
Abstract
Tissue preconditioning, whereby various short-term stressors initiate organ resistance to subsequent injury, is well recognized. However, clinical preconditioning of the kidney for protection against acute kidney injury (AKI) has not been established. Here we tested whether a pro-oxidant agent, iron sucrose, combined with a protoporphyrin (Sn protoporphyrin), can induce preconditioning and protect against acute renal failure. Mice were pretreated with iron sucrose, protoporphyrin, cyanocobalamin, iron sucrose and protoporphyrin, or iron sucrose and cyanocobalamin. Eighteen hours later, ischemic, maleate, or glycerol models of AKI were induced, and its severity was assessed the following day (blood urea nitrogen, plasma creatinine concentrations; post-ischemic histology). Agent impact on cytoprotective gene expression (heme oxygenase 1, hepcidin, haptoglobin, hemopexin, α1-antitrypsin, α1-microglobulin, IL-10) was assessed as renal mRNA and protein levels. AKI-associated myocardial injury was gauged by plasma troponin I levels. Combination agent administration upregulated multiple cytoprotective genes and, unlike single agent administration, conferred marked protection against each tested model of acute renal failure. Heme oxygenase was shown to be a marked contributor to this cytoprotective effect. Preconditioning also blunted AKI-induced cardiac troponin release. Thus, iron sucrose and protoporphyrin administration can upregulate diverse cytoprotective genes and protect against acute renal failure. Associated cardiac protection implies potential relevance to both AKI and its associated adverse downstream effects.
Collapse
|
100
|
Hess DC, Khan MB, Hoda N, Morgan JC. Remote ischemic conditioning: a treatment for vascular cognitive impairment. Brain Circ 2015; 1:133-139. [PMID: 30221201 DOI: 10.4103/2394-8108.172885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
There is a strong link between hypoperfusion and white matter (WM) damage in patients with leukoaraiosis and vascular cognitive impairment (VCI). Other than management of vascular risk factors, there is no treatment for WM damage and VCI that delays progression of the disease process to dementia. Observational studies suggest that exercise may prevent or slow down the progression of Alzheimer's disease (AD) and VCI. However, getting patients to exercise is challenging especially with advancing age and disability. Remote ischemic conditioning, an "exercise equivalent", allows exercise to be given with a "device" in the home for long periods of time. Since RIC increases CBF in pre-clinical studies and in humans, RIC may be an ideal therapy to treat VCI and WM disease and perhaps even sporadic AD. By using MRI imaging of WM progression, a sample size in the range of about 100 subjects per group could determine if RIC has activity in WM disease and VCI.
Collapse
Affiliation(s)
- David C Hess
- Department of Neurology Medical College of Georgia, Georgia Regent's University, Augusta, GA USA
| | - Mohammad B Khan
- Department of Neurology Medical College of Georgia, Georgia Regent's University, Augusta, GA USA
| | - Nasrul Hoda
- Department of Medical Laboratory, Imaging, and Radiologic Sciences, College of Allied Health Sciences, Georgia Regent's University, Augusta, GA USA
| | - John C Morgan
- Department of Neurology Medical College of Georgia, Georgia Regent's University, Augusta, GA USA
| |
Collapse
|