ArticlePDF Available

Methylene blue treatment in experimental ischemic stroke: A mini-review

Authors:

Abstract and Figures

Stroke is among the leading causes of death and long-term disability. Methylene blue (MB), a drug grandfathered by the Food and Drug Administration with a long history of safe usage in humans for treating methemoglobinemia and cyanide poisoning, has recently been shown to be neuroprotective in neurodegenerative diseases and brain injuries. The goal of this paper is to review studies on MB in experimental stroke models.
Content may be subject to copyright.
Brain
Circulation
Volume 2 | Issue 1 | Jan-Mar 2016
ISSN 2394-8108
Brain Circulation
Publisher: Wolters Kluwer Health - Medknow
Office: B-9, Kanara Business Center, Off Link Rd, Ghatkopar (E), Mumbai-400075, India.
Email: editor@braincirculation.org
Website: http://braincirculation.org
48 © 2016 Brain Circulation | Published by Wolters Kluwer Health – Medknow
Methylene blue treatment
in experimental ischemic stroke:
A mini-review
Zhao Jiang, Timothy Q Duong1
Abstract:
Stroke is among the leading causes of death and long-term disability. Methylene blue (MB), a drug grandfathered
by the Food and Drug Administration with a long history of safe usage in humans for treating methemoglobinemia
and cyanide poisoning, has recently been shown to be neuroprotective in neurodegenerative diseases and brain
injuries. The goal of this paper is to review studies on MB in experimental stroke models.
Key words:
Ischemia, methylene blue (MB), stroke
Introduction
Stroke is the second leading cause of death
and the leading cause of long-term disability
worldwide, and the fourth leading cause of
death in the United States.[1] There are 800,000
new or recurrent strokes per year in the United
States. Of the 6 million Americans who are
stroke survivors, 71% are unable to return to
work. Over $70 billion was expended on stroke
patient care in 2013.[1] This cost is steadily rising
because the conditions that put people at the risk
of stroke (such as heart disease, hypertension,
diabetes, and obesity) are also steadily on the
rise. Recombinant tissue plasminogen activator
(rtPA), the only drug clinically approved to
treat ischemic stroke, is limited to only a small
subset of patients due to the serious risk of often
fatal hemorrhagic transformation and can only
be administered within 4.5 h of stroke onset.[2]
Recently, intraarterial therapy using primarily
stent-retriever technology to achieve mechanical
thrombectomy combined with intravenous (IV)
rtPA administration has been found to be superior
to IV rtPA alone when patients with proximal
cerebral arterial occlusions are treated within 6 h of
symptom onset.[3] Despite the tremendous efforts
taken in stroke research, our ability to minimize
infarct volume and neurological de cit remains
extremely limited. Thus, there is an urgent need
to develop new treatments for stroke to protect the
brain from the acute phase to the chronic phase.
In acute stroke, a therapeutic approach is
to buy time (i.e., protecting neurons and
glia via sustaining metabolic energy) before
recanalization.[4,5] This may allow the expansion
of the critical treatment time window. During the
reperfusion phase, it is important to minimize
reperfusion injury such as that from excessive
production of reactive oxygen species
[6,7] that
could accelerate mitochondrial damage.[8]
During the chronic phase, the brain undergoes
signi cant remodeling[9,10] and it is important to
maximize functional recovery. Thus, advanced
drug or reagent methodologies, to enhance
ischemic cells and tissues survival and assist the
effect of thrombolytic treatment, are required in
the development of effective therapies for the
management of stroke patients. Mitochondrial
targeting is one of the promising strategies that
is widely explored.[11,12]
Methylene blue (MB), a Food and Drug
Administration (FDA)-grandfathered
drug, is currently used to treat malaria,
methemoglobinemia, and cyanide poisoning
in humans.[13,14] MB has been rigorously studied
for over 120 years with 5,794 human MB studies
listed in Pubmed (searched in December 2015).
Low-dose MB (1-5 mg/kg IV) is very safe.
Its pharmacokinetics, side effect profile, and
contraindications are well-known and most
importantly minimal in humans.[15,16] There
were only a few negative reports and they were
associated with exceptionally high doses. For
example, MB has been used in parathyroid
Address for
correspondence:
Dr. Zhao Jiang, Research
Imaging Institute,
Radiology, University of
Texas Health Science
Center at San Antonio, San
Antonio, TX, United States.
E-mail: johnjiang406@
gmail.com
Submission: 26-08-2015
Revised: 13-01-2016
Accepted: 20-01-2016
Research Imaging
Institute, Radiology,
1Department of
Ophthalmology,
Radiology and
Physiology, University
of Texas Health
Science Center, San
Antonio, Texas, USA
Access this article online
Quick Response Code:
Website:
http://www.braincirculation.org
DOI:
10.4103/2394-8108.178548
Review Article
How to cite this article: Jiang Z, Duong TQ.
Methylene blue treatment in experimental ischemic
stroke: A mini-review. Brain Circ 2016;2:48-53.
This is an open access arƟ cle distributed under the terms of the
CreaƟ ve Commons AƩ ribuƟ on-NonCommercial-ShareAlike 3.0 License,
which allows others to remix, tweak, and build upon the work
non-commercially, as long as the author is credited and the new
creaƟ ons are licensed under the idenƟ cal terms.
For reprints contact: reprints@medknow.com
Jiang and Duong: A mini-review of methylene blue in ischemia
Brain Circulation - Vol 2, Issue 1, January 2016 49
surgery to aid in lymphatic mapping at doses of 3.5-10 mg/
kg. The FDA also warned physicians about possible serious
serotonin reactions in patients who received IV MB during
parathyroid surgery if taking serotonergic psychiatric drugs.
However, a subsequent report by Mayo Clinic surgeons and
pharmacologists summarized the FDA evidence and literature
and concluded “that the use of methylene blue dye at low
doses for lymphatic mapping likely carries very little risk for
serotonin neurotoxicity.”[17] There has never been any negative
report based on oral MB. Daily 4 mg/kg oral MB has been
used safely for 1 year in clinical trials.[18] MB at 1-3 mg/kg IV
is safely used as a standard treatment for metabolic poisoning
in emergency rooms.
The mechanisms of action of MB are as followed. MB
has renewable auto-oxidizing property, which acts as an
electron cycler that allows MB to redirect electrons to the
mitochondrial electron transport chain, thereby enhancing
adenosine triphosphate (ATP) production and promoting
cell survival. In bypassing complex I-III activity to generate
ATP, MB reduces reactive oxygen species production from
the mitochondrial electron transport chain. The antioxidant
property of MB is thus unique. In vitro studies have rmly
established that MB enhances cytochrome c oxidase (complex
IV) activity to produce more ATP in cells under normoxia, and
MB replaces oxygen as the oxidant to sustain ATP generation
under hypoxia while simultaneously reducing oxidative
stress.[19-22] Moreover, chronic MB treatment also modi es
mitochondrial function and induces long-lasting cellular
changes.[23] Speci cally, repeated low-dose (0.5-2.0 mg/kg) MB
has long-lasting upregulation of brain cytochrome c oxidase
activity.[20,24-26] MB readily crosses the blood–brain barrier
because of its high lipophilicity.[15]
Low-dose MB has recently been shown to reduce
neurobehavioral impairment in optic neuropathy,[19,27]
traumatic brain injury,[28] Parkinson’s disease,[23,29] Alzheimer
disease,[30-32] and ischemic stroke.[4,5,33] The goal of this article is
to review relevant MB literatures in relation to neuroprotection
in experimental stroke models.
A Pubmed search (December 2015) resulted in 25 papers
relevant to the use of MB in stroke or related to stroke
[Table 1]. Our goal is to review pertinent findings from
most of these.
Basic Stroke-related Methylene Blue Studies
One of the earliest MB experiments was performed by Sidi
et al. in 1987.[34] Arterial pressure transiently increased followed
with MB (5 mg/kg) administration by using hemodynamic
measurements in dogs. Wu and Bohr found the contraction
produced by endothelin was augmented when the intact
aortic rings were treated with MB (105 M) in aortas from
Wistar–Kyoto rats but not in those from stroke-prone
spontaneously hypertensive rats.[37] Ishiyama et al. studied the
inhibitory action of MB against nicorandil-induced vasodilation
in dogs.[40] Kontos and Wei demonstrated that MB could
eliminate the arteriolar dilation in response to nitroprusside and
nitroglycerin after permeabilization of the cell membrane.[39]
MB has been shown to increase blood pressure and myocardial
function by inhibiting nitric oxide actions in human septic
shock disease.[41,47,50,52] These studies demonstrated that MB has
vascular effects and causes vasoconstriction transiently, thereby
improving blood pressure, which could help to defend against
hypoperfusion during stroke.
Nitric oxide generation during ischemia and reperfusion plays
a signi cant role in ischemic and reperfusion injury.[56] There is
evidence that MB decreases or inhibits nitric oxide generation
that might have the potential effect of neuroprotection in
ischemia/reperfusion injury. In order to prove that the
endocardial endothelium of Rana esculenta produces an
amount of nitric oxide that is suf cient to modulate ventricular
performance, Sys et al. measured the changes of stroke volume
(as a measure of performance in paced frog hearts) and stroke
work (as an index of systolic function) after using MB-induced
inhibition of nitric oxide synthase.[43] This reminded us that MB
could inhibit nitric oxide generation. Evgenov et al. found that
continuous infusion of MB counteracted the early myocardial
dysfunction and derangement of hemodynamics and gas
exchange by the inhibition of nitric oxide pathway in ovine
endotoxemia model.[48]
Xie et al. demonstrated that MB treatment activated 5’adenosine
monophosphate-activated protein kinase signaling but did
not inhibit mammalian target of rapamycin signaling in
serum deprivation cells and normal mouse.[57] This study
suggests that MB-induced neuroprotection is mediated, at
least in part, by macroautophagy. Additionally, MB treatment
altered the levels of microtubule-associated protein light chain
3 type II, cathepsin D, Beclin-1, and p62, suggesting that it was
a potent inducer of autophagy.[58] Thus, MB may be related to
autophagic cell death.
Ryou et al. studied the MB-induced neuroprotective
mechanism focusing on stabilization and activation of
hypoxia-inducible factor-1 in an in vitro oxygen-glucose
deprivation reoxygenation model.[55] They found that MB
activated the erythropoietin-signaling pathway with a
corresponding increase in hypoxia-inducible factor-1 and
consequently related to apoptotic cell death. Together,
these studies shed light on the molecular pathways that MB
modulates.
Methylene Blue Studies in Ischemic Stroke
While low-dose MB has recently been shown to reduce
neurobehavioral impairment in neurodegenerative diseases
(ca. Parkinson’s disease,[23,29] Alzheimer’s disease[30-32]), the
neuroprotective effects of MB on cerebral ischemia in vivo
were only recently demonstrated. In 2006, a Swedish group
found that IV MB at clinical dose was neuroprotective after
experimental cardiac arrest in piglets using histology.[59] Wen
et al. showed that MB could signi cantly reduce focal cerebral
ischemia reperfusion damage in a transient focal cerebral
ischemia rodent model in 2011 using histology.[60]
Di et al. demonstrated that MB improved neurological
function, and reduced the infarct volume and the necrosis
after acute cerebral ischemic injury by augmenting
mitophagy.[54] These improvements depended on the effect
of MB on mitochondrial structure and function. Acute
cerebral ischemia caused the disorder of and disintegration
Jiang and Duong: A mini-review of methylene blue in ischemia
50 Brain Circulation - Vol 2, Issue 1, January 2016
of mitochondrial structure while MB ameliorated the
destruction of mitochondria. They also further revealed that
the elevation of mitochondrial membrane potential by MB
under oxygen-glucose deprivation conditions mediated the
augmented mitophagy in an oxygen-glucose deprivation
model in vitro.
Shen et al. evaluated the ef cacy of MB to treat ischemic stroke
in a transient middle cerebral artery occlusion model in rats
using noninvasive multimodal magnetic resonance imaging
(MRI).[5] In a randomized double-blinded design in which
vehicle or MB was administered after reperfusion, they found
that the initial lesion volumes de ned by abnormal apparent
diffusion coef cient [61] at 30 min after ischemia were not
signi cantly different between the two groups. The nal infarct
volumes de ned by T2 changes 2 days after stroke increased
in the vehicle group but decreased in the MB group, yielding
a 30% difference in infarct volume [Figure 1]. Tracking tissue
fate on a pixel-by-pixel basis showed that MB salvaged more
initial ischemic core pixels compared to the control group,
and more mismatch pixels compared to the control group.
This study, for the rst time, evaluated the ef cacy of MB
to treat ischemic stroke in rats using longitudinal MRI and
behavioral measures.
Table 1: Published papers about MB related to stroke (searched in Pubmed in December 2015)
Year Cell/animal Dose Function
1987 Dog 1-5 mg/kg Increased arterial pressure transiently[34]
1988 Dog 10-5 M Relaxation of middle cerebral arterial strips was attenuated[35]
1990 Human 10-5 M Inhibited the relaxations induced by thrombin or bradykinin in human
basilar arteries[36]
1990 Rat 10-5 M Augmented the contraction produced by endothelin in intact aortic rings[37]
1991 Feline 10-5 M Inhibited the magnesium de ciency-related dilations on the tone of middle
cerebral arteries[38]
1993 Cat 5 mM Eliminated the arteriolar dilation after permeabilization of the cell
membrane[39]
1994 Dog 10-5 M Inhibitory action of methylene blue against nicorandil-induced vasodilation
in pial vessels[40]
1995 Human 2 mg/kg Transiently and reproduciblely increased arterial pressure associated with
an improvement in cardiac function[41]
1996 Dog 5 mg/kg Increased arterial pressure, pulmonary arterial pressure, and systemic and
pulmonary vascular resistances but decreased cardiac index and regional
blood ow[42]
1997 Frog 10-6 M Inhibition of nitric oxide synthase[43]
1999 Rat 10 MAttenuated endothelium-dependent relaxation in the mesenteric artery[44]
1999 Human 4 mg/kg Increases systemic vascular resistance and may improve myocardial
function[45]
2001 Fish 10-6 M Inhibited nitric oxide synthase[46]
2001 Human 2 mg/kg and 2 mg/kg/h for 1 h Counteracted myocardial depression; maintained oxygen transport and
reduced concurrent adrenergic support[47]
2001 Sheep 10 mg/kg and 2.5 mg/kg/h for 5 h Counteracted the early myocardial dysfunction and derangement of
hemodynamics and gas exchange by inhibiting the nitric oxide pathway[48]
2002 Rat 10-4 and 10-5 M Attenuated endothelium-dependent relaxation in aorta[49]
2002 Human 3 mg/kg Acute vasoconstrictive and positive inotropic effects during septic shock[50]
2005 Human 2 mg/kg Preoperative methylene blue administration reduced the incidence and
severity of vasoplegic syndrome[51]
2010 Human 1 mg/kg, 3 mg/kg, and 7 mg/kg High dose of MB enhanced splanchnic perfusion[52]
2012 HT22 cells 5 MAttenuated superoxide production and antioxidant[53]
2013 Rat 0.5 mg/kg and 1 mg/kg MB treatment minimized ischemic brain injury and improved functional
outcomes.[5]
2014 Rat 1 mg/kg and 3 mg/kg MB delayed the growth rate of the perfusion-diffusion mismatch into
infarction in permanent stroke models[4]
2015 Rat and PC12 cell 1 mg/kg, 5 mg/kg, or 10 mg/kg for
rat and 0.5 M for cell
MB promoted mitophagy by maintaining the MMP§ at a relatively high
level, which contributed to a decrease in necrosis and an improvement
in neurological function, thereby protecting against acute cerebral
ischemic injury[54]
2015 HT22 cells 1 M and 10 MMB protects the hippocampus-derived neuronal cells against OGD-
reoxygenation injury by enhancing energy metabolism and increasing
HIF-1 protein content accompanied by an activation of the EPO
signaling pathway.[55]
2015 Rat 1 mg/kg MB induced neuroprotection by enhancing autophagy and reducing
apoptosis in the perfusion-diffusion mismatch tissue following
ischemic stroke[33]
MB: Methylene blue, §MMP: Matrix metalloproteinase, OGD: Oxygen-glucose deprivation, EPO: Erythropoietin receptor
Jiang and Duong: A mini-review of methylene blue in ischemia
Brain Circulation - Vol 2, Issue 1, January 2016 51
Rodriguez et al. applied a similar multimodal MRI to test
the hypothesis that MB treatment delays progression of
at-risk tissue (ca. perfusion-diffusion mismatch) to infarct in
permanent middle cerebral artery occlusion in rats at two MB
treatment doses.[4] MB signi cantly prolonged the perfusion-
diffusion mismatch, and mildly increased the cerebral blood
flow in the hypoperfused tissue. MRI is now a routine
neuroimaging tool in the clinic. MRI plays an important role
in diagnosing, evaluating, and monitoring the cerebral tissue
undergoing stroke and thereby, providing a noninvasive means
to longitudinally evaluate treatment ef cacy.
To further probe the underlying molecular mechanisms of
neuroprotection of MB following transient ischemic stroke in
rats, Jiang et al. employed noninvasive MRI to guide extraction
of the different ischemic tissue types for western blot analysis
of apoptotic and autophaphic cascades.[33] Multimodal MRI
during the acute phase and at 24 h were used to de ne three
regions of interest (ROIs):
1. The perfusion-diffusion mismatch salvaged by reperfusion,
2. The perfusion-diffusion mismatch not salvaged by
reperfusion, and
3. The ischemic core. The tissues from these ROIs were
extracted for western blot analyses of autophagic and
apoptotic markers.
The major ndings were:
1. MB improved cerebral blood ow to the perfusion-diffusion
mismatch tissue after reperfusion and minimized harmful
hyperperfusion 24 h after stroke,
2. MB reduces infarct volume and behavioral deficits
following transient ischemic stroke in rats,
3. MB improves cerebral blood ow (CBF) to at-risk tissue
after reperfusion and minimizes harmful hyperperfusion
24 h after MCAO,
4. MB inhibits apoptosis and enhances autophagy in the
at-risk tissue but not within the ischemic core,
5. MB modulates the p53-Bax-Bcl2-caspase3 cascade,
inhibiting apoptotic signaling pathways,
6.MB modulates p53-AMPK-TSC2-mTOR cascades,
enhancing autophagic signaling pathways [Figure 2].
Conclusion
Low-dose MB has a long history of safe usage in humans for
treating methemoglobinemia and cyanide poisoning. MB also
has energy-enhancing and antioxidant properties. There are
substantial evidences that MB is neuroprotective for ischemic
stroke. A number of studies have now investigated the
mechanisms of action in ischemic stroke. Noninvasive MRI
offers a means to identify neural correlates of neuroprotection,
target speci c tissue types for further investigation of molecular
Figure 1: Initial lesion (30 mins ADC) and nal infarct (24 hrs T2) volumes of vehicle- and MB-treated rats subjected to 60 min middle cerebral
artery occlusion. Initial lesion and nal infarct volumes of individual animal were connected using dot lines. Mean initial lesion and nal infarct
were connected using solid lines. Adapted from the reference paper5]
Jiang and Duong: A mini-review of methylene blue in ischemia
52 Brain Circulation - Vol 2, Issue 1, January 2016
mechanisms of action, and longitudinally evaluate treatment
ef cacy. The excellent safety pro le of low-dose MB in humans,
together with noninvasive MRI, could expedite MB stroke
clinical trials. MB treatments could offer novel therapeutic
regimens in combination or alone to improve patient care
following a stroke.
Financial support and sponsorship
This work was supported by NIH/NINDS R01 NS45879.
Con icts of interest
There are no con icts of interest.
References
1. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ,
et al.; American Heart Association Statistics Committee and Stroke
Statistics Subcommittee. Heart disease and stroke statistics-2014
update: A report from the American Heart Association.
Circulation 2014;129:e28-292.
2. Davis SM, Donnan GA. 4.5 hours: The new time window for tissue
plasminogen activator in stroke. Stroke 2009;40: 2266-7.
3. Berkhemer OA, Fransen PS, Beumer D, van den Berg LA,
Lingsma HF, Yoo AJ, et al.; MR CLEAN Investigators.
A randomized trial of intraarterial treatment for acute ischemic
stroke. N Engl J Med 2015;372:11-20.
4. Rodriguez P, Jiang Z, Huang S, Shen Q, Duong TQ. Methylene
blue treatment delays progression of perfusion-diffusion
mismatch to infarct in permanent ischemic stroke. Brain Res
2014;1588:144-9.
5. Shen Q, Du F, Huang S, Rodriguez P, Watts LT, Duong TQ.
Neuroprotective ef cacy of methylene blue in ischemic stroke:
An MRI study. PLoS One 2013;8:e79833.
6. Liu Y, Fiskum G, Schubert D. Generation of reactive oxygen
species by the mitochondrial electron transport chain.
J Neurochem 2002;80:780-7.
7. Chen Q, Vazquez EJ, Moghaddas S, Hoppel CL, Lesnefsky EJ.
Production of reactive oxygen species by mitochondria: Central
role of complex III. J Biol Chem 2003;278:36027-31.
8. Crack PJ, Taylor JM. Reactive oxygen species and the modulation
of stroke. Free Radic Biol Med 2005;38:1433-44.
9. Murphy TH, Corbett D. Plasticity during stroke recovery: From
synapse to behaviour. Nat Rev Neurosci 2009;10:861-72.
10. Chopp M, Zhang ZG, Jiang Q. Neurogenesis, angiogenesis,
and MRI indices of functional recovery from stroke. Stroke
2007;38(Suppl):827-31.
11. Chaturvedi RK, Beal MF. Mitochondrial approaches for
neuroprotection. Ann N Y Acad Sci 2008;1147:395-412.
12. Galluzzi L, Blomgren K, Kroemer G. Mitochondrial membrane
permeabilization in neuronal injury. Nat Rev Neurosci
2009;10:481-94.
13. Scheindlin S. Something old... something blue. Mol Interv
2008;8:268-73.
14. Clifton J 2nd, Leikin JB. Methylene blue. Am J Ther 2003;10:289-91.
15. Peter C, Hongwan D, Küpfer A, Lauterburg BH. Pharmacokinetics
and organ distribution of intravenous and oral methylene blue.
Eur J Clin Pharmacol 2000;56:247-50.
16. Walter-Sack I, Rengelshausen J, Oberwittler H, Burhenne J,
Mueller O, Meissner P, et al. High absolute bioavailability of
methylene blue given as an aqueous oral formulation. Eur J Clin
Pharmacol 2009;65:179-89.
17. Shah-Khan MG, Lovely J, Degnim AC. Safety of methylene blue
dye for lymphatic mapping in patients taking selective serotonin
reuptake inhibitors. Am J Surg 2012;204:798-9.
18. Naylor GJ, Martin B, Hopwood SE, Watson Y. A two-year
double-blind crossover trial of the prophylactic effect of methylene
blue in manic-depressive psychosis. Biol Psychiatry 1986;21:915-20.
19. Zhang X, Rojas JC, Gonzalez-Lima F. Methylene blue prevents
neurodegeneration caused by rotenone in the retina. Neurotox
Res 2006;9:47-57.
20. Riha PD, Bruchey AK, Echevarria DJ, Gonzalez-Lima F.
Memory facilitation by methylene blue: Dose-dependent effect
on behavior and brain oxygen consumption. Eur J Pharmacol
2005;511:151-8.
21. Scott A, Hunter FE Jr. Support of thyroxine-induced swelling of
liver mitochondria by generation of high energy intermediates
at any one of three sites in electron transport. J Biol Chem
1966;241:1060-6.
22. Lindahl PE, Oberg KE. The effect of rotenone on respiration and
its point of attack. Exp Cell Res 1961;23:228-37.
23. Rojas JC, Bruchey AK, Gonzalez-Lima F. Meurometabolic
mechanisms for memory enhancement and neuroprotection of
methylene blue. Prog Neurobiol 2012;96:32-45.
24. Gonzalez-Lima F, Bruchey AK. Extinction memory improvement
by the metabolic enhancer methylene blue. Learn Mem
2004;11:633-40.
25. Wrubel KM, Riha PD, Maldonado MA, McCollum D,
Gonzalez-Lima F. The brain metabolic enhancer methylene blue
improves discrimination learning in rats. Pharmacol Biochem
Behav 2007;86:712-7.
26. Vasquez B, Bieber AL. Direct visualization of IMP — GMP:
Pyrophosphate phosphoribosyltransferase in polyacrylamide
gels. Anal Biochem 1978;84:504-11.
27. Rojas JC, John JM, Lee J, Gonzalez-Lima F. Methylene blue
provides behavioral and metabolic neuroprotection against optic
neuropathy. Neurotox Res 2009;15:260-73.
28. Talley Watts L, Long JA, Chemello J, Van Koughnet S, Fernandez A,
Huang S, et al. Methylene blue is neuroprotective against mild
traumatic brain injury. J Neurotrauma 2014;13: 1063-71.
29. Ishiwata A, Sakayori O, Minoshima S, Mizumura S, Kitamura S,
Katayama Y. Preclinical evidence of Alzheimer changes in
progressive mild cognitive impairment: A qualitative and
quantitative SPECT study. Acta Neurol Scand 2006;114:91-6.
30. Oz M, Lorke DE, Petroianu GA. Methylene blue and Alzheimer’s
disease. Biochem Pharmacol 2009;78:927-32.
31. O’Leary JC 3rd, Li Q, Marinec P, Blair LJ, Congdon EE, Johnson AG,
et al. Phenothiazine-mediated rescue of cognition in tau transgenic
Figure 2: MB induces neuroprotection in neurons following cerebral
ischemia through both the apoptotic p53-Bcl-2-Bax signaling
pathway and autophagic p53-AMPK-TSC2-mTOR signaling
pathway. The arrows show the effect of MB on these pathways.
Adapted from the reference paper[33]
Jiang and Duong: A mini-review of methylene blue in ischemia
Brain Circulation - Vol 2, Issue 1, January 2016 53
mice requires neuroprotection and reduced soluble tau burden.
Mol Neurodegener 2010;5:45.
32. Medina DX, Caccamo A, Oddo S. Methylene blue reduces a
levels and rescues early cognitive de cit by increasing proteasome
activity. Brain Pathol 2011;21:140-9.
33. Jiang Z, Watts LT, Huang S, Shen Q, Rodriguez P, Chen C, et al.
The effects of methylene blue on autophagy and apoptosis in
mri-de ned normal tissue, ischemic penumbra and ischemic core.
PLoS One 2015;10:e0131929.
34. Sidi A, Paulus DA, Rush W, Gravenstein N, Davis RF. Methylene
blue and indocyanine green artifactually lower pulse oximetry
readings of oxygen saturation. Studies in dogs. J Clin Monit
1987;3:249-56.
35. Onoue H, Nakamura N, Toda N. Endothelium-dependent and
-independent responses to vasodilators of isolated dog cerebral
arteries. Stroke 1988;19:1388-94.
36. Hatake K, Kakishita E, Wakabayashi I, Sakiyama N, Hishida S.
Effect of aging on endothelium-dependent vascular relaxation of
isolated human basilar artery to thrombin and bradykinin. Stroke
1990;21:1039-43.
37. Wu CC, Bohr DF. Role of endothelium in the response to
endothelin in hypertension. Hypertension 1990;16:677-81.
38. Szabó C, Faragó M, Dóra E, Horváth I, Kovách AG. Endothelium-
dependent in uence of small changes in extracellular magnesium
concentration on the tone of feline middle cerebral arteries. Stroke
1991;22:785-9.
39. Kontos HA, Wei EP. Hydroxyl radical-dependent inactivation of
guanylate cyclase in cerebral arterioles by methylene blue and by
LY83583. Stroke 1993;24:427-34.
40. Ishiyama T, Dohi S, Iida H, Akamatsu S, Ohta S, Shimonaka H.
Mechanisms of vasodilation of cerebral vessels induced by
the potassium channel opener nicorandil in canine in vivo
experiments. Stroke 1994;25:1644-50.
41. Preiser JC, Lejeune P, Roman A, Carlier E, De Backer D,
Leeman M, et al. Methylene blue administration in septic shock:
A clinical trial. Crit Care Med 1995;23:259-64.
42. Zhang H, Rogiers P, Friedman G, Preiser JC, Spapen H,
Buurman WA, et al. Effects of nitric oxide donor SIN-1 on oxygen
availability and regional blood ow during endotoxic shock.
Arch Surg 1996;131:767-74.
43. Sys SU, Pellegrino D, Mazza R, Gattuso A, Andries LJ, Tota L.
Endocardial endothelium in the avascular heart of the frog:
Morphology and role of nitric oxide. J Exp Biol 1997;200:3109-18.
44. Sunano S, Watanabe H, Tanaka S, Sekiguchi F, Shimamura K.
Endothelium-derived relaxing, contracting and hyperpolarizing
factors of mesenteric arteries of hypertensive and normotensive
rats. Br J Pharmacol 1999;126:709-16.
45. Weingartner R, Oliveira E, Oliveira ES, Sant’Anna UL,
Oliveira RP, Azambuja LA, et al. Blockade of the action of
nitric oxide in human septic shock increases systemic vascular
resistance and has detrimental effects on pulmonary function
after a short infusion of methylene blue. Braz J Med Biol Res
1999;32:1505-13.
46. Imbrogno S, De Iuri L, Mazza R, Tota B. Nitric oxide modulates
cardiac performance in the heart of Anguilla anguilla. J Exp Biol
2001;204:1719-27.
47. Kirov MY, Evgenov OV, Evgenov NV, Egorina EM,
Sovershaev MA, Sveinbjørnsson B, et al. Infusion of methylene
blue in human septic shock: A pilot, randomized, controlled
study. Crit Care Med 2001;29:1860-7.
48. Evgenov OV, Sveinbjørnsson B, Bjertnaes LJ. Continuously
infused methylene blue modulates the early cardiopulmonary
response to endotoxin in awake sheep. Acta Anaesthesiol Scand
2001;45:1246-54.
49. Sekiguchi F, Miyake Y, Kashimoto T, Sunano S. Unaltered
caffeine-induced relaxation in the aorta of stroke-prone
spontaneously hypertensive rats (SHRSP). J Smooth Muscle Res
2002 38:11-22.
50. Donati A, Conti G, Loggi S, Münch C, Coltrinari R, Pelaia P, et al.
Does methylene blue administration to septic shock patients
affect vascular permeability and blood volume? Crit Care Med
2002;30:2271-7.
51. Ozal E, Kuralay E, Yildirim V, Kilic S, Bolcal C, Kücükarslan N,
et al. Preoperative methylene blue administration in patients at
high risk for vasoplegic syndrome during cardiac surgery. Ann
Thorac Surg 2005;79:1615-9.
52. Juffermans NP, Vervloet MG, Daemen-Gubbels CR, Binnekade JM,
de Jong M, Groeneveld AB. A dose- nding study of methylene
blue to inhibit nitric oxide actions in the hemodynamics of human
septic shock. Nitric Oxide 2010;22:275-80.
53. Poteet E, Winters A, Yan LJ, Shufelt K, Green KN, Simpkins JW,
et al. Neuroprotective actions of methylene blue and its
derivatives. PLoS One 2012;7:e48279.
54. Di Y, He YL, Zhao T, Huang X, Wu KW, Liu SH, et al. Methylene
blue reduces acute cerebral ischemic injury via the induction of
mitophagy. Mol Med 2015;21:420-9.
55. Ryou MG, Choudhury GR, Li W, Winters A, Yuan F, Liu R, et al.
Methylene blue-induced neuronal protective mechanism against
hypoxia-reoxygenation stress. Neuroscience 2015;301:193-203.
56. Gürsoy-Ozdemir Y, Bolay H, Saribaş O, Dalkara T. Role of
endothelial nitric oxide generation and peroxynitrite formation
in reperfusion injury after focal cerebral ischemia. Stroke
2000;31:1974-81.
57. Xie L, Li W, Winters A, Yuan F, Jin K, Yang S. Methylene blue
induces macroautophagy through 5’ adenosine monophosphate-
activated protein kinase pathway to protect neurons from serum
deprivation. Front Cell Neurosci 2013;7:56.
58. Congdon EE, Wu JW, Myeku N, Figueroa YH, Herman M,
Marinec PS, et al. Methylthioninium chloride (methylene blue)
induces autophagy and attenuates tauopathy in vitro and in vivo.
Autophagy 2012;8:609-22.
59. Sharma HS, Miclescu A, Wiklund L. Cardiac arrest-induced
regional blood-brain barrier breakdown, edema formation and
brain pathology: A light and electron microscopic study on a new
model for neurodegeneration and neuroprotection in porcine
brain. J Neural Transm (Vienna) 2011;118:87-114.
60. Wen Y, Li W, Poteet EC, Xie L, Tan C, Yan LJ, et al. Alternative
mitochondrial electron transfer as a novel strategy for
neuroprotection. J Biol Chem 2011;286:16504-15.
61. Moseley ME, Cohen Y, Mintorovitch J, Chileuitt L, Shimizu H,
Kucharczyk J, et al. Early detection of regional cerebral ischemia
in cats: Comparison of diffusion- and T2-weighted MRI and
spectroscopy. Magn Reson Med 1990;14:330-46.
... 31 MB has been proven to have a neuroprotective effect, mostly in animal studies. 32 MB could reduce the infarct size and neurological deficit, especially in transient cerebral ischemia, in rats. 33 The recently known mechanisms were reducing apoptosis and increasing autophagy through the suppression of p53 expression. ...
... 33 The recently known mechanisms were reducing apoptosis and increasing autophagy through the suppression of p53 expression. 32 However, in this study, MB does not show a significant protection to the cerebrovascular accident in VS patients. Though, all of the analyzed studies showed a similar result which was less CVA incidence found in patients administered with MB compared to the control group. ...
Article
Full-text available
Background: To evaluate the benefit of methylene blue as an adjunct treatment by assessing hemodynamic, morbidity rate, intensive care unit length of stay, and mortality rate outcomes in adult patients with vasoplegic syndrome. Methods: A systematic search through electronic databases including Pubmed, Embase, Scopus, and Medline for studies assessing the use of methylene blue in patients with vasoplegic syndrome compared to control treatments. The Newcastle-Ottawa Scale tool was used for observational studies, and Jadad Scale was used for controlled trials to assess the risk of bias. Results: This systematic review included six studies for qualitative synthesis and five studies for quantitative synthesis. Pooled analysis revealed that mean arterial pressure, systemic vascular resistance, heart rate, and hospital stay were not statistically significant in methylene blue administration compared to control. However, administration of methylene blue in vasoplegic syndrome patients significantly reduces renal failure (OR = 0.25; 95% CI = 0.08-0.75), development of multiple organ failure (OR = 0.09; 95% CI = 0.02-0.51), and mortality rate (OR = 0.12; 95% CI = 0.03-0.46). Conclusion: Adjunct administration of methylene blue for vasoplegic syndrome patients significantly reduces renal failure, multiple organ failure, and mortality.
... Azure A (AZA) and Methylene Blue (MB) are phenothiazine dyes with the same basic phenothiazine skeleton, however, differing in the CH 3 groups present at position 3 ( Fig. 1) [11]. These dyes have the capacity to inactivate several kinds of pathogens, and have been applied in the treatment of many diseases, including neurodegenerative diseases [12], cerebral ischemia [13], malaria [14], and cancer [15], owing to their high redox potential and their ability to effectively interact with visible light [16]. However, to the best of our knowledge, the kinetics and thermodynamics of the interactions between phenothiazine dyes and bovine lactoferrin have not been investigated, which makes this study even more relevant. ...
... (12) can be rewritten as Eq. (13). ...
Article
Life manifestation is mainly based on biopolymer-ligand molecular recognition; therefore, the elucidation of energy and speed associated with protein-ligand binding is strategic in understanding and modulating biological systems. In this study, the interactions between methylene blue (MB) or azure A (AZA) dyes and bovine lactoferrin (BLF) were investigated by surface plasmon resonance, fluorescence spectroscopy, and isothermal titration microcalorimetry. Despite the molecular similarities between the dyes, the BLF-AZA binding thermodynamic parameters (ΔGAZAo = −30.50 and ΔHAZAo = 10.8 (kJ·mol−1)) were higher in magnitude than those of the BLF-MB systems (ΔGMBo = −27.3 and ΔHMBo = 5.72 (kJ·mol−1)). To increase the systems' entropy (TΔSAZAo = 41.3 and TΔSMBo = 33.0 (kJ·mol−1)), the hydrophobic interactions must outweigh the electrostatic repulsion, thereby promoting BLF-dye binding. The activation complex formation (Eac, aMB = 33, Eac, aAZA = 32, ∆Ha, MB‡ = 31, ∆Ha, AZA‡ = 30, ∆Ga, MB‡ = 51.84, ∆Ga, AZA‡ = 50.7, T∆Sa, MB‡ = −21, T∆Sa, AZA‡ = −21 (kJ·mol−1)), owing to free BLF and MB (or AZA) associations, was not affected by the dye chemical structure, while for the thermodynamically stable BLF-dye complex dissociation, the same energetic parameters (Eac, dMB = 16, Eac, dAZA = 6.4, ∆Hd, MB‡ = 14, ∆Hd, AZA‡ = 3.9, ∆Gd, MB‡ = 81.4, ∆Gd, AZA‡ = 74.93, T∆Sd, MB‡ = −68, T∆Sd, AZA‡ = −71.0 (kJ·mol−1)) were considerably affected by the number of methyl groups. Our results may be very useful to determine binding processes controlled by kinetic parameters, as well as to optimize the application of these photosensitive dyes in biological systems.
... Azure A (AZA) and Methylene Blue (MB) are phenothiazine dyes with the same basic phenothiazine skeleton, however, differing in the CH 3 groups present at position 3 ( Fig. 1) [11]. These dyes have the capacity to inactivate several kinds of pathogens, and have been applied in the treatment of many diseases, including neurodegenerative diseases [12], cerebral ischemia [13], malaria [14], and cancer [15], owing to their high redox potential and their ability to effectively interact with visible light [16]. However, to the best of our knowledge, the kinetics and thermodynamics of the interactions between phenothiazine dyes and bovine lactoferrin have not been investigated, which makes this study even more relevant. ...
... (12) can be rewritten as Eq. (13). ...
Article
The complexation between protein and polyphenol affects their biological functions. A complete understanding of such interactions requires comprehensive thermodynamic and kinetic characterizations. Surface plasmon resonance (SPR)and fluorescence spectroscopy (FS)described similarly the thermodynamic of interaction between bovine lactoferrin (bLF)and epigallocatechin-3-gallate (EGCG). The formation of the bLF-EGCG complex is spontaneous (ΔG SPRo ≈ -29.00 kJ mol ⁻¹ , ΔG FSo ≈ -26.00 kJ mol ⁻¹ )and entropically driven (ΔH SPRo = 14.26, ΔH FSo = 10.20 kJ mol ⁻¹ and TΔS SPRo ≈ 43.00, TΔS FSo ≈ 36.00 kJ mol ⁻¹ ). The kinetic parameters obtained by SPR showed that the reaction occurs through an activated complex, whose energetic formation parameters from the association of free molecules (E act(a) = 49.5 kJ mol ⁻¹ , ΔH a‡ = 47.0 kJ mol ⁻¹ , and TΔS a‡ = −2.10 kJ mol ⁻¹ )were higher than those in the opposite direction (namely the dissociation of the stable complex, E act(d) = 17.4 kJ mol ⁻¹ , ΔH d‡ = 32.8 kJ mol ⁻¹ , and TΔS d‡ = −45.10 kJ mol ⁻¹ ), except for ΔG ‡ (ΔG a‡ = 49.1 kJ mol ⁻¹ and ΔG d‡ = 77.9 kJ mol ⁻¹ ). This study provides useful information for optimizing the use of bLF–EGCGcomplex as a bioactive compound in different systems, such as medical, food, cosmetic, and pharmaceutical formulations.
... Methylene Blue is a drug that has been used safely in humans for treating methemoglobinemia and cyanide poisoning [6][7][8]. Several studies have been focused on methylene blue and its neuroprotective effects in neurodegenerative diseases and brain injuries [9,10]. In recent year, many methods have been developed for degrading MB and decreasing its environmental risks [11], such as advanced fenton oxidation [12], chemical degradation [13], biological degradation [14], and electrochemical degradation [15]. ...
Article
Full-text available
In this work, Zinc oxide (ZnO) thin films were synthesised by the successive ionic layer adsorption and reaction (SILAR) technique at various number of cycles (10-50 cycles). The effect of film thickness on the structural properties, surface morphology, optical and electrical properties, and sunlight assisted photocatalytic activities through photocatalytic degradation of Methylene Blue (MB) dye of ZnO thin films were studied. The energy dispersive X-rays (EDX) analysis confirmed the presence Zn and O elements. The X-rays diffraction (XRD) pattern showed the polycrystalline nature of ZnO thin films and the crystallite size increases with film thickness. The SEM images showed that a greater film thickness resulted in the growth of hexagonal nanorods arrays. Atomic force microscopy (AFM) images revealed that the surface roughness increases with film thickness yielding in an enhanced specific surface area. The UV-visible transmission spectra showed that increasing film thickness results in band gap expansion from 3.15 eV to 3.31 eV together with a reduction in optical transmittance. The estimated sheet resistance and resistivity were found to be in the range of 1.34-7.1 Ω/sq and 0.09-2.12 x 10-1 Ω.cm. The photocatalytic studies reveal that increasing film thickness leads to an improved photocatalytic efficiency of ZnO films. The enhanced photocatalytic activity of ZnO films is due to the increased surface area and low recombination rate of carriers charges (e-/h+), resulting from band gap expansion.
... Studies showed that low doses of MB could improve both spatial [25] and non-spatial [26] memories. Different studies in animal models of Alzheimer's disease [21], Parkinson's disease [22], hypoxia [23], and stroke [24] also indicate that the MB has neuroprotective effects. Improvements in brain function and memory tasks were also observed by MB in humans [25,26]. ...
Article
Full-text available
Low-level laser therapy (LLLT) and methylene blue (MB) were proved to have neuroprotective effects. In this study, we evaluated the preventive effects of LLLT and MB alone and in combination to examine their efficacy against sleep deprivation (SD)–induced cognitive impairment. Sixty Balb/c male mice were randomly divided into five groups as follows: wide platform (WP), SD, LLLT, MB, LMB (treatment with both LLLT and MB). Daily MB (0.5 mg/kg) was injected for ten consecutive days. An 810-nm, 10-Hz pulsed laser was used in LLLT every other day. We used the T-maze test, social interaction test (SIT), and shuttle box to assess learning and memory and PSD-95, GAP-43, and synaptophysin (SYN) markers to examine synaptic proteins levels in the hippocampus. Our results showed that SD decreased alternation rate in the T-maze test, sociability and social novelty in SIT, and memory index in the shuttle box. Single treatments were not able to reverse these in most of the behavioral parameters. However, behavioral tests showed a significant difference between combined therapy and the SD group. The levels of synaptic plasticity markers were also significantly reduced after SD. There was a significant difference between the MB group and SD animals in GAP-43 and SYN biomarkers. Combination treatment with LLLT and MB also increased GAP-43, PSD-95, and SYN compared to the SD group. We found that the combined use of LLLT and MB pretreatment is more effective in protecting SD-induced cognitive impairment, which may be imparted via modulation of synaptic proteins.
... It was also found that MB modulated the p53-5' adenosine monophosphate -activated protein kinase-Tuberous Sclerosis Complex 2-mammalian target of rapamycin cascade, enhancing autophagic signaling pathways. [44] The manipulation of p53-induced pathways with treatment shows positive results, and the studies should be continued to find new ways to manipulate p53 pathways, producing better stroke outcomes. Stem cells also need to present neural markers, [45] and p53 may provide a way to regulate these. ...
Article
Full-text available
The potential use of stem cells as a therapeutic treatment for many neurological disorders, such as stroke, has spiked an interest in their properties. Due to limitations of the present-day treatments, regenerative and protective therapies could prove very beneficial if a safe and effective treatment is identified. Using human amniotic fluid stem (hAFS) cells could theoretically provide both neuroprotective and regenerative properties to patients, and knowledge of p53's activity and function could be a key component in understanding the behavior and characteristics of these stem cells to harness their full potential. Many recent studies on p53 have provided new and valuable information that could give rise to new ideas for treatment options. More specifically, p53's activity inside hAFS cells lead them closer to becoming a potential therapeutic stem cell. Other neuroprotective treatments, such as hyperoxia and hypoxia sessions, are showing positive results. In combination, these data are helping to get closer to an effective treatment for neurological disorders.
... [98,99] Large interest is arising for methylene blue, currently approved by the FDA for the treatment of Alzheimer's and Parkinson's diseases, for stroke patients. [100] It has been shown that methylene blue can modulate the electrons flow through the ETC. In particular, being a carrier of electrons between NADH and cytochrome c, it can allow electrons to bypass Complex I and III resulting in decrease of electron leakage and improvement in ATP production along with a consequent reduction of ROS and oxidative stress. ...
Article
Full-text available
Stroke is a main cause of mortality and morbidity worldwide. Despite the increasing development of innovative treatments for stroke, most are unsuccessful in clinical trials. In recent years, an encouraging strategy for stroke therapy has been identified in stem cells transplantation. In particular, grafting cells and their secretion products are leading with functional recovery in stroke patients by promoting the growth and function of the neurovascular unit – a communication framework between neurons, their supply microvessels along with glial cells – underlying stroke pathology and recovery. Mitochondrial dysfunction has been recently recognized as a hallmark in ischemia/reperfusion neural damage. Emerging evidence of mitochondria transfer from stem cells to ischemic-injured cells points to transfer of healthy mitochondria as a viable novel therapeutic strategy for ischemic diseases. Hence, a more in-depth understanding of the cellular and molecular mechanisms involved in mitochondrial impairment may lead to new tools for stroke treatment. In this review, we focus on the current evidence of mitochondrial dysfunction in stroke, investigating favorable approaches of healthy mitochondria transfer in ischemic neurons, and exploring the potential of mitochondria-based cellular therapy for clinical applications. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed.
Article
Full-text available
Cells have evolved extensive signaling mechanisms to maintain redox homeostasis. While basal levels of oxidants are critical for normal signaling, a tipping point is reached when the level of oxidant species exceed cellular antioxidant capabilities. Myriad pathological conditions are characterized by elevated oxidative stress, which can cause alterations in cellular operations and damage to cellular components including nucleic acids. Maintenance of nuclear chromatin are critically important for host survival and eukaryotic organisms possess an elaborately orchestrated response to initiate repair of such DNA damage. Recent evidence indicates links between the cellular antioxidant response, the DNA damage response (DDR), and the epigenetic status of the cell under conditions of elevated oxidative stress. In this emerging model, the cellular response to excessive oxidants may include redox sensors that regulate both the DDR and an orchestrated change to the epigenome in a tightly controlled program that both protects and regulates the nuclear genome. Herein we use sepsis as a model of an inflammatory pathophysiological condition that results in elevated oxidative stress, upregulation of the DDR, and epigenetic reprogramming of hematopoietic stem cells (HSCs) to discuss new evidence for interplay between the antioxidant response, the DNA damage response, and epigenetic status.
Article
Full-text available
Epilepsy is a chronic neurodegenerative disease, and accumulating evidence suggests its pathological progression is closely associated with peroxynitrite (ONOO−). However, understanding the function remains challenging due to a lack of in vivo imaging probes for ONOO− determination in epileptic brains. Here, the first near‐infrared imaging probe (named ONP) is presented for tracking endogenous ONOO− in brains of kainate‐induced epileptic seizures with high sensitivity and selectivity. Using this probe, the dynamic changes of endogenous ONOO− fluxes in epileptic brains are effectively monitored with excellent temporal and spatial resolution. In vivo visualization and in situ imaging of hippocampal regions clearly reveal that a higher concentration of ONOO− in the epileptic brains associates with severe neuronal damage and epileptogenesis; curcumin administration can eliminate excessively increased ONOO−, further effectively protecting neuronal cells. Moreover, by combining high‐content analysis and ONP, a high‐throughput screening method for antiepileptic inhibitors is constructed, which provides a rapid imaging/screening approach for understanding epilepsy pathology and accelerating antiseizure therapeutic discovery. The first near‐infrared imaging probe for tracking endogenous peroxynitrite (ONOO−) in brains of kainate‐induced epileptic seizures is developed, which is named ONP. A higher concentration of ONOO− in the epileptic brains associated with severe neuronal damage and epileptogenesis is observed by imaging of hippocampal regions with ONP. By combining high‐content analysis and ONP, a high‐throughput screening method for antiepileptic inhibitors is constructed.
Article
Full-text available
Methylene blue (MB) USP, which has energy-enhancing and antioxidant properties, is currently used to treat methemoglobinemia and cyanide poisoning in humans. We recently showed that MB administration reduces infarct volume and behavioral deficits in rat models of ischemic stroke and traumatic brain injury. This study reports the underlying molecular mechanisms of MB neuroprotection following transient ischemic stroke in rats. Rats were subjected to transient (60-mins) ischemic stroke. Multimodal MRI during the acute phase and at 24hrs were used to define three regions of interest (ROIs): i) the perfusion-diffusion mismatch salvaged by reperfusion, ii) the perfusion-diffusion mismatch not salvaged by reperfusion, and iii) the ischemic core. The tissues from these ROIs were extracted for western blot analyses of autophagic and apoptotic markers. The major findings were: 1) MB treatment reduced infarct volume and behavioral deficits, 2) MB improved cerebral blood flow to the perfusion-diffusion mismatch tissue after reperfusion and minimized harmful hyperperfusion 24hrs after stroke, 3) MB inhibited apoptosis and enhanced autophagy in the perfusion-diffusion mismatch, 4) MB inhibited apoptotic signaling cascades (p53-Bax-Bcl2-Caspase3), and 5) MB enhanced autophagic signaling cascades (p53-AMPK-TSC2-mTOR). MB induced neuroprotection, at least in part, by enhancing autophagy and reducing apoptosis in the perfusion-diffusion mismatch tissue following ischemic stroke.
Article
Full-text available
Background: In patients with acute ischemic stroke caused by a proximal intracranial arterial occlusion, intraarterial treatment is highly effective for emergency revascularization. However, proof of a beneficial effect on functional outcome is lacking. Methods: We randomly assigned eligible patients to either intraarterial treatment plus usual care or usual care alone. Eligible patients had a proximal arterial occlusion in the anterior cerebral circulation that was confirmed on vessel imaging and that could be treated intraarterially within 6 hours after symptom onset. The primary outcome was the modified Rankin scale score at 90 days; this categorical scale measures functional outcome, with scores ranging from 0 (no symptoms) to 6 (death). The treatment effect was estimated with ordinal logistic regression as a common odds ratio, adjusted for prespecified prognostic factors. The adjusted common odds ratio measured the likelihood that intraarterial treatment would lead to lower modified Rankin scores, as compared with usual care alone (shift analysis). Results: We enrolled 500 patients at 16 medical centers in The Netherlands (233 assigned to intraarterial treatment and 267 to usual care alone). The mean age was 65 years (range, 23 to 96), and 445 patients (89.0%) were treated with intravenous alteplase before randomization. Retrievable stents were used in 190 of the 233 patients (81.5%) assigned to intraarterial treatment. The adjusted common odds ratio was 1.67 (95% confidence interval [CI], 1.21 to 2.30). There was an absolute difference of 13.5 percentage points (95% CI, 5.9 to 21.2) in the rate of functional independence (modified Rankin score, 0 to 2) in favor of the intervention (32.6% vs. 19.1%). There were no significant differences in mortality or the occurrence of symptomatic intracerebral hemorrhage. Conclusions: In patients with acute ischemic stroke caused by a proximal intracranial occlusion of the anterior circulation, intraarterial treatment administered within 6 hours after stroke onset was effective and safe. (Funded by the Dutch Heart Foundation and others; MR CLEAN Netherlands Trial Registry number, NTR1804, and Current Controlled Trials number, ISRCTN10888758.).
Article
Full-text available
In endotoxemia and septic shock, enhanced generation of endogenous nitric oxide (NO) contributes to myocardial depression, hypotension, and derangement of gas exchange. We hypothesized that continuous infusion of methylene blue (MB), an inhibitor of the NO pathway, would counteract these effects in endotoxemic sheep. Twenty-one sheep were anesthetized and instrumented for a chronic study with vascular catheters. On the day of the experiment, 18 conscious animals randomly received either an intravenous injection of MB 10 mg x kg(-1) or isotonic saline. Thirty minutes later, sheep received a 20-min intravenous infusion of Escherichia coli endotoxin 1 microg x kg(-1) and either an intravenous infusion of MB 2.5 mg x kg(-1) x h(-1) or isotonic saline, respectively, for 5 h. In addition, 3 animals were exposed to the same dose of MB alone. MB reduced the early endotoxin-induced declines in stroke volume, left ventricular stroke work and cardiac indices, and prevented mean arterial pressure from falling. Moreover, MB ameliorated the increases in pulmonary arterial pressure and pulmonary vascular resistance index. In addition, MB reduced the increments in venous admixture and AaPO2, decreased the falls in PaO2, SaO2, and oxygen delivery, and maintained oxygen consumption. MB also prevented the rises in body temperature and plasma nitrites and nitrates, and delayed the elevation of plasma lactate. When given alone to healthy sheep, MB transiently reduced plasma lactate and PaO2, and increased AaPO2. In ovine endotoxemia, continuously infused MB counteracts the early myocardial dysfunction and derangement of hemodynamics and gas exchange.
Article
Full-text available
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Methylene blue (MB) has known energy-enhancing and antioxidant properties. This study tested the hypothesis that MB treatment reduces lesion volume and behavioral deficits in a rat model of mild TBI. In a randomized double-blinded design, animals received either MB (n=5) or vehicle (n=6) after TBI. Studies were performed on 0, 1, 2, 7 and 14 days following an impact to the primary forelimb somatosensory cortex. MRI lesion was not apparent 1 hour after TBI, became apparent 3 hours after TBI, and peaked at 2 days for both groups. The MB-treated animals showed significantly smaller MRI lesion volume than the vehicle-treated animals at all time points studied. The MB-treated animals exhibited significantly improved scores on forelimb placement asymmetry and foot fault tests compared to the vehicle-treated animals at all time points studied. Smaller numbers of dark-stained Nissl cells and Fluro Jade positive cells were observed in the MB-treated group compared to vehicle-treated animals 14 days post TBI. In conclusion, MB treatment minimized lesion volume, behavioral deficits, and neuronal degeneration following mild TBI. MB is already FDA-approved to treat a number of indications, likely expediting future clinical trials in TBI.
Article
Brain ischemia and reperfusion (I/R) injury occurs in various pathological conditions, but there is no effective treatment currently available in clinical practice. Methylene blue (MB) is a century old drug with a newly discovered protective function in the ischemic stroke model. In the current investigation we studied the MB-induced neuroprotective mechanism focusing on stabilization and activation of hypoxia inducible factor-1α (HIF-1α) in an in vitro oxygen and glucose deprivation (OGD)-reoxygenation model. HT22 cells were exposed to OGD (0.1% O2, 6h) and reoxygenation (21% O2, 24h). Cell viability was determined with the calcein AM assay. The dynamic change of intracellular O2 concentration was monitored by fluorescence lifetime imaging microscopy (FLTIM). Glucose uptake was quantified using the 2-[N-(7-Nitrobenz-2-Oxa- 1,3-Diazol-4-yl)Amino]- 2-Deoxy-D-Glucose (2-NBDG) assay. ATP concentration and glycolytic enzyme activity were examined by spectrophotometry. Protein content changes were measured by immunoblot: HIF-1α, prolyl hydroxylase 2(PHD2), erythropoietin (EPO), Akt, mTOR, and PIP5K. The contribution of HIF-1α activation in the MB-induced neuroprotective mechanism was confirmed by blocking HIF-1α activation with 2-methoxyestradiol-2 (2-MeOE2) and by transiently transfecting constitutively active HIF-1α. MB increases cell viability by about 50% vs. OGD control. Compared to the corresponding control, MB increases intracellular O2 concentration and glucose uptake as well as the activities of hexokinase and G-6-PDH, and ATP concentration. MB activates the EPO signaling pathway with a corresponding increase in HIF-1α. Phosphorylation of Akt was significantly increased with MB treatment followed by activation of the mTOR pathway. Importantly, we observed, MB increased nuclear translocation of HIF-1α vs. control (about 3 folds), which was shown by a ratio of nuclear:cytoplasmic HIF-1α protein content. We conclude that MB protects the hippocampus derived neuronal cells against OGD-reoxygenation injury by enhancing energy metabolism and increasing HIF-1α protein content accompanied by an activation of the EPO signaling pathway. Copyright © 2015. Published by Elsevier Ltd.
Article
The treatment of stroke is limited by a short therapeutic window and a lack of effective clinical drugs. Methylene blue (MB) has been used in laboratories and clinics since the 1890s. Few studies have reported the neuroprotective role of MB in cerebral ischemia-reperfusion injury. However, whether and how MB protects against acute cerebral ischemia (ACI) injury was unclear. In this study, we investigated the effect of MB on this injury and revealed that MB protected against ACI injury by augmenting mitophagy. Using a rat middle cerebral artery occlusion (MCAO) model, we demonstrated that MB improved neurological function, and reduced the infarct volume and the necrosis after ACI injury. These improvements depended on the effect of MB on mitochondrial structure and function. ACI caused the disorder of and disintegration of mitochondrial structure, while MB ameliorated the destruction of mitochondria. In addition, mitophagy was inhibited at 24 h after stroke and MB augmented mitophagy. In an oxygen-glucose deprivation (OGD) model in vitro, we further revealed that the elevation of mitochondrial membrane potential (MMP) by MB under OGD conditions mediated the augmented mitophagy. In contrast, exacerbating the decline of MMP during OGD abolished the MB-induced activation of mitophagy. Taken together, MB promotes mitophagy by maintaining the MMP at a relatively high level, which contributes to a decrease in necrosis and an improvement in neurological function, thereby protecting against ACI injury.