1
|
Hashmi MU, Ahmed R, Mahmoud S, Ahmed K, Bushra NM, Ahmed A, Elwadie B, Madni A, Saad AB, Abdelrahman N. Exploring Methylene Blue and Its Derivatives in Alzheimer's Treatment: A Comprehensive Review of Randomized Control Trials. Cureus 2023; 15:e46732. [PMID: 38022191 PMCID: PMC10631450 DOI: 10.7759/cureus.46732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Methylene blue (MB) and its compounds are investigated for their potential benefits in the management of Alzheimer's disease (AD). AD is a widely seen neuropathological disorder characterized by the gradual decline of cognitive abilities, ultimately leading to the development of severe dementia. It is anticipated that there will be a significant increase in the prevalence of AD due to the aging population. Histopathologically, AD is distinguished by the presence of intracellular tangles of neurofibrillary tissues (NFTs) and extracellular amyloid plaques within the brain. MB is a thiophenazine dye with FDA approval for treating several illnesses. Its ease in crossing the blood-brain barrier and potential therapeutic use in central nervous system diseases have increased interest in its application for treating AD. The literature review includes randomized clinical trials investigating MB's potential benefits in treating AD. The findings of the studies indicate that the administration of MB has demonstrated enhancements in cognitive function, reductions in the accumulation of plaques containing beta-amyloid, improvements in memory and cognitive function in animal subjects, and possesses antioxidant properties that can mitigate oxidative stress and inflammation within the brain. This review evaluates the modern and latest research on the application of MB for treating AD.
Collapse
Affiliation(s)
| | - Ragda Ahmed
- Internal Medicine, White River Health, Batesville, USA
| | - Sulafa Mahmoud
- Internal Medicine, Michigan State University, East Lansing, USA
| | - Kholood Ahmed
- Internal Medicine, College of Human Medicine, Michigan State University, East Lansing, USA
| | - Noura M Bushra
- Internal Medicine, Michigan State University, East Lansing, USA
| | - Areeg Ahmed
- Nephrology, Harlem Hospital Center, Columbia University, New York, USA
| | - Batran Elwadie
- Internal Medicine, Michigan State University, East Lansing, USA
| | - Amna Madni
- Internal Medicine, Michigan State University, East Lansing, USA
| | - Amel B Saad
- Internal Medicine, Michigan State University, East Lansing, USA
| | - Nadir Abdelrahman
- Family Medicine, College of Human Medicine, Michigan State University, East Lansing, USA
| |
Collapse
|
2
|
Uhelski ML, Johns ME, Horrmann A, Mohamed S, Sohail A, Khasabova IA, Simone DA, Banik RK. Adverse effects of methylene blue in peripheral neurons: An in vitro electrophysiology and cell culture study. Mol Pain 2022; 18:17448069221142523. [PMID: 36408567 PMCID: PMC9730009 DOI: 10.1177/17448069221142523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Methylene blue (MB) is an effective treatment for methemoglobinemia, ifosfamide-induced encephalopathy, cyanide poisoning, and refractory vasoplegia. However, clinical case reports and preclinical studies indicate potentially neurotoxic activity of MB at certain concentrations. The exact mechanisms of MB neurotoxicity are not known, and while the effects of MB on neuronal tissue from different brain regions and myenteric ganglia have been examined, its effects on primary afferent neurons from dorsal root ganglia (DRG) have not been studied. Mouse DRG were exposed to MB (0.3-10 μM) in vitro to assess neurite outgrowth. Increasing concentrations of MB (0.3-10 μM) were associated with neurotoxicity as shown by a substantial loss of cells with neurite formation, particularly at 10 μM. In parallel experiments, cultured rat DRG neurons were treated with MB (100 μM) to examine how MB affects electrical membrane properties of small-diameter sensory neurons. MB decreased peak inward and outward current densities, decreased action potential amplitude, overshoot, afterhyperpolarization, increased action potential rise time, and decreased action potential firing in response to current stimulation. MB induced dose-dependent toxicity in peripheral neurons, in vitro. These findings are consistent with studies in brain and myenteric ganglion neurons showing increased neuronal loss and altered membrane electrical properties after MB application. Further research is needed to parse out the toxicity profile for MB to minimize damage to neuronal structures and reduce side effects in clinical settings.
Collapse
Affiliation(s)
- Megan L Uhelski
- Department of Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Malcolm E Johns
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Alec Horrmann
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sadiq Mohamed
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ayesha Sohail
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Iryna A Khasabova
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Ratan K Banik
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA,Ratan K Banik, Department of Anesthesiology, University of Minnesota, B515 Mayo Memorial Building, 420 Delaware Street S.E., MMC 294, Minneapolis, MN 55455, USA.
| |
Collapse
|
3
|
Morén C, Treder N, Martínez-Pinteño A, Rodríguez N, Arbelo N, Madero S, Gómez M, Mas S, Gassó P, Parellada E. Systematic Review of the Therapeutic Role of Apoptotic Inhibitors in Neurodegeneration and Their Potential Use in Schizophrenia. Antioxidants (Basel) 2022; 11:2275. [PMID: 36421461 PMCID: PMC9686909 DOI: 10.3390/antiox11112275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 09/15/2023] Open
Abstract
Schizophrenia (SZ) is a deleterious brain disorder affecting cognition, emotion and reality perception. The most widely accepted neurochemical-hypothesis is the imbalance of neurotransmitter-systems. Depleted GABAergic-inhibitory function might produce a regionally-located dopaminergic and glutamatergic-storm in the brain. The dopaminergic-release may underlie the positive psychotic-symptoms while the glutamatergic-release could prompt the primary negative symptoms/cognitive deficits. This may occur due to excessive synaptic-pruning during the neurodevelopmental stages of adolescence/early adulthood. Thus, although SZ is not a neurodegenerative disease, it has been suggested that exaggerated dendritic-apoptosis could explain the limited neuroprogression around its onset. This apoptotic nature of SZ highlights the potential therapeutic action of anti-apoptotic drugs, especially at prodromal stages. If dysregulation of apoptotic mechanisms underlies the molecular basis of SZ, then anti-apoptotic molecules could be a prodromal therapeutic option to halt or prevent SZ. In fact, risk alleles related in apoptotic genes have been recently associated to SZ and shared molecular apoptotic changes are common in the main neurodegenerative disorders and SZ. PRISMA-guidelines were considered. Anti-apoptotic drugs are commonly applied in classic neurodegenerative disorders with promising results. Despite both the apoptotic-hallmarks of SZ and the widespread use of anti-apoptotic targets in neurodegeneration, there is a strikingly scarce number of studies investigating anti-apoptotic approaches in SZ. We analyzed the anti-apoptotic approaches conducted in neurodegeneration and the potential applications of such anti-apoptotic therapies as a promising novel therapeutic strategy, especially during early stages.
Collapse
Affiliation(s)
- Constanza Morén
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- U722 Group, Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Carlos III Health Institute, 28029 Madrid, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Nina Treder
- Faculty of Psychology and Neuroscience, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Albert Martínez-Pinteño
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Natàlia Rodríguez
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Néstor Arbelo
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Santiago Madero
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Marta Gómez
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Psychiatry, Servizo Galego de Saúde (SERGAS), 36001 Pontevedra, Spain
| | - Sergi Mas
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Patricia Gassó
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Eduard Parellada
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
4
|
Bužga M, Machytka E, Dvořáčková E, Švagera Z, Stejskal D, Máca J, Král J. Methylene blue: a controversial diagnostic acid and medication? Toxicol Res (Camb) 2022; 11:711-717. [PMID: 36337249 PMCID: PMC9618115 DOI: 10.1093/toxres/tfac050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 09/01/2023] Open
Abstract
A narrative review of the literature was conducted to determine if the administration of methylene blue (MB) in humans has potential risks. Studies were identified from MEDLINE, Web of Science, Scopus, and Cochrane. MB is a diagnostic substance used during some diagnostic procedures and also a part of the treatment of several diseases including methemoglobinemia, vasoplegic syndrome, fosfamide-induced encephalopathy, and cyanide intoxication, and the detection of leaks or position of parathyroid corpuscles during surgery. Although the use of MB is historically justified, and it ought to be safe, because it originated as a diagnostic material, the basic toxicological characteristics of this substance are unknown. Despite reports of severe adverse effects of MB, which could significantly exceed any possible benefits evaluated for the given indication. Therefore, the clinical use of MB currently represents a controversial problem given the heterogeneity of available data and the lack of preclinical data. This is in conflict with standards of safe use of such substances in human medicinal practice. The toxic effects of the application of MB are dose-dependent and include serious symptoms such as hemolysis, methemoglobinemia, nausea and vomitus, chest pain, dyspnoea, and hypertension. Some countries regard MB as harmful because of the resulting skin irritation and triggering of an adverse inflammatory response. MB induced serotoninergic toxicity clinically manifests as neuromuscular hyperactivity. This review aims to summarize the current understanding concerning the indications for MB administration and define the potential adverse effects of MB.
Collapse
Affiliation(s)
- Marek Bužga
- Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava 17. listopadu 1790, OStrava, 70800, Czech Republic
- Department of Physiology and Pathophysiology, Faculty of Medicine, University of Ostrava, Ostrava Syllabova 19, Ostrava Vitkovice, 70030, Czech Republic
| | - Evžen Machytka
- Institute for Clinical and Experimental Medicine, Hepatogastroenterology Department, Prague Videňska 1958/9, Praha, 14021, Czech Republic
| | - Eliška Dvořáčková
- Institute of Pharmacology, 1st Faculty of Medicine, Charles University, Prague Albertov 4, Praha, 12108, Czech Republic
| | - Zdeněk Švagera
- Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava 17. listopadu 1790, OStrava, 70800, Czech Republic
| | - David Stejskal
- Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava 17. listopadu 1790, OStrava, 70800, Czech Republic
| | - Jan Máca
- Department of Physiology and Pathophysiology, Faculty of Medicine, University of Ostrava, Ostrava Syllabova 19, Ostrava Vitkovice, 70030, Czech Republic
| | - Jan Král
- Institute for Clinical and Experimental Medicine, Hepatogastroenterology Department, Prague Videňska 1958/9, Praha, 14021, Czech Republic
| |
Collapse
|
5
|
Wang YF, Luo Y, Hou GL, He RJ, Zhang HY, Yi YL, Zhang Y, Cui ZQ. Pretreatment with Methylene Blue Protects Against Acute Seizure and Oxidative Stress in a Kainic Acid-Induced Status Epilepticus Model. Med Sci Monit 2021; 27:e933469. [PMID: 34628461 PMCID: PMC8513497 DOI: 10.12659/msm.933469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The aim of the present study was to investigate the potential anticonvulsant effect of methylene blue (MB) in a kainic acid (KA)-induced status epilepticus (SE) model. The effects of MB on levels of oxidative stress and glutamate (Glu) also were explored. Material/Methods Sixty C57BL/6 mice were randomly divided into 5 equal-sized groups: (1) controls; (2) KA; (3) MB 0.5 mg/kg+KA; (4) MB 1 mg/kg+KA; and (5) vehicle+KA. The SE model was established by intra-amygdala microinjection of KA. Behavioral observations and simultaneous electroencephalographic records of the seizures in different groups were analyzed to determine the potential anticonvulsant effect of MB. The influences of MB on oxidative stress markers and glutamate were also detected to explore the possible mechanism. Results MB afforded clear protection against KA-induced acute seizure, as measured by the delayed latency of onset of generalized seizures and SE, decreased percentage of SE, and increased survival rate in mice with acute epilepsy. MB markedly increased the latency to first onset of epileptiform activity and decreased the average duration of epileptiform events, as well as the percentage of time during which the epileptiform activity occurred. Administration of MB prevented KA-induced deterioration of oxidative stress markers and Glu. Conclusions MB is protective against acute seizure in SE. This beneficial effect may be at least partially related to its potent antioxidant ability and influence on Glu level.
Collapse
Affiliation(s)
- Yong-Feng Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China (mainland)
| | - Yan Luo
- Department of Reproductive Genetic, Hebei General Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Gao-Lei Hou
- Department of Neurosurgery, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China (mainland)
| | - Rui-Jing He
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China (mainland)
| | - Hao-Yun Zhang
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China (mainland)
| | - Yan-Li Yi
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China (mainland)
| | - Ying Zhang
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China (mainland)
| | - Zhi-Qiang Cui
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, China (mainland)
| |
Collapse
|
6
|
Ye Z, Li X, Zheng D, Pei S, Cheng P, Zhang L, Zhu L. Intravitreally Injected Methylene Blue Protects Retina against Acute Ocular Hypertension in Rats. Curr Eye Res 2021; 47:91-101. [PMID: 34165383 DOI: 10.1080/02713683.2021.1948062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose: To assess the neuroprotective effects of methylene blue (MB) in a rat model of acute ocular hypertension (AOH) and explore its possible mechanisms.Methods: Our AOH rat model was obtained with anterior chamber perfusion for 60 min. After that, 100 μM MB was injected into the vitreous cavity immediately after injury. Electroretinogram, fundus photography, optical coherence tomography (OCT) and retina morphology examination were utilized to quantify retinal damage before surgery, as well as 7, 14 and 28 days after. The average number of surviving retinal ganglion cells (RGCs) was counted after fluorescent retrograde labelling with 4% DiI. And TUNEL assay was used to investigate retinal cell apoptosis at 24 hours after AOH. Nrf2 and BACE1 in the retina were determined by RT-qPCR analysis.Results: AOH did produce a severe degeneration effect on the whole retinal layer. Intravitreally injected MB maintained certain retinal thickness after AOH, reduced the destruction of electroretinograms, and enhanced RGCs survival. The average number of TUNEL-labelled cells statistically reduced in the MB-treated retina tissue compared with retina treated with normal saline. The relative mRNA level of Nrf2 was also much higher in the MB-treated retinas after AOH, and the expression of BACE1 had a decline in the AOH + MB group.Conclusions: MB can protect the retina from AOH injury and the possible mechanism might involve the inhibition of BACE1 expression and the activation of Nrf2 antioxidant pathway.
Collapse
Affiliation(s)
- Zhiqiang Ye
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoli Li
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, Henan, China
| | - Dongliang Zheng
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuaili Pei
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pei Cheng
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lishu Zhang
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lin Zhu
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
7
|
Methylene blue can act as an antidote to pesticide poisoning of bumble bee mitochondria. Sci Rep 2021; 11:14710. [PMID: 34282204 PMCID: PMC8289979 DOI: 10.1038/s41598-021-94231-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/07/2021] [Indexed: 11/09/2022] Open
Abstract
The population of bumble bees and other pollinators has considerably declined worldwide, probably, due to the toxic effect of pesticides used in agriculture. Inexpensive and available antidotes can be one of the solutions for the problem of pesticide toxicity for pollinators. We studied the properties of the thiazine dye Methylene blue (MB) as an antidote against the toxic action of pesticides in the bumble bee mitochondria and found that MB stimulated mitochondrial respiration mediated by Complex I of the electron transport chain (ETC) and increased respiration of the mitochondria treated with mitochondria-targeted (chlorfenapyr, hydramethylnon, pyridaben, tolfenpyrad, and fenazaquin) and non-mitochondrial (deltamethrin, metribuzin, and penconazole) pesticides. MB also restored the mitochondrial membrane potential dissipated by the pesticides affecting the ETC. The mechanism of MB action is most probably related to its ability to shunt electron flow in the mitochondrial ETC.
Collapse
|
8
|
Méndez M, Fidalgo C, Arias JL, Arias N. Methylene blue and photobiomodulation recover cognitive impairment in hepatic encephalopathy through different effects on cytochrome c-oxidase. Behav Brain Res 2021; 403:113164. [PMID: 33549685 DOI: 10.1016/j.bbr.2021.113164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/02/2021] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
Mitochondrial dysfunction plays a central role in hepatic encephalopathy (HE), due to changes in enzyme cytochrome c-oxidase (CCO), causing a decline in brain metabolism. We used an HE animal model and applied intracranial administration of methylene blue (MB) and transcranial photobiomodulation (PBM), both targeting CCO, to determine their differential effects on recovering cognition. Five groups of rats were used: sham-operated group + saline (SHAM + SAL, n = 6), hepatic encephalopathy + SAL (HE + SAL, n = 7), SHAM + methylene blue (SHAM + MB, n = 7), HE + MB (n = 7), HE + PBM (n = 7). PBM animals were exposed transcranially to 670 +/- 10 nm LED light at a dose of 9 J/cm2 once a day for 7 days, and the MB and SAL groups were injected with 2.2 μg/0.5 μL in the accumbens. Cognitive dysfunction was evaluated on a striatal stimulus-response task using the Morris water maze. Our results showed cognitive improvement in the HE group when treated with MB. This improvement was accompanied by a decrease in CCO activity in the prefrontal cortex, dorsal striatum, and dorsal hippocampus. When comparing MB and PBM, we found that, although both treatments effectively improved the HE-memory deficit, there was a differential effect on CCO. A general decrease in CCO activity was found in the prefrontal and entorhinal cortices, dorsal striatum, and hippocampus when PBM, compared to MB, was applied. Our results suggest that mitochondrial dysfunction and brain metabolic decline in HE might involve CCO alteration and can be improved by administering MB and PBM.
Collapse
Affiliation(s)
- Marta Méndez
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad de Oviedo, Plaza Feijoo s/n, Oviedo, 33003, Spain; INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| | - Camino Fidalgo
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain; Departamento de Psicología y Sociología, IIS Aragón, Universidad de Zaragoza, Ciudad Escolar s/n, Teruel, 44003, Spain
| | - Jorge L Arias
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad de Oviedo, Plaza Feijoo s/n, Oviedo, 33003, Spain; INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| | - Natalia Arias
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain; UK Dementia Research Institute, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK.
| |
Collapse
|
9
|
Fung JCL, Cho EYP. Methylene blue promotes survival and GAP-43 expression of retinal ganglion cells after optic nerve transection. Life Sci 2020; 262:118462. [PMID: 32961228 DOI: 10.1016/j.lfs.2020.118462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 11/28/2022]
Abstract
AIMS Neurodegeneration of the optic nerve and retinal ganglion cells (RGCs) leads to progressive vision loss. As part of the central nervous system, RGCs show limited ability to regenerate and there is extensive search for neuroprotective agents for optic nerve damage. Methylene blue (MB) exhibits beneficial effects against various neurodegenerative diseases of the central nervous system. However, the mechanisms associated with its putative protection on neuronal survival and regeneration remain obscure. This study used the optic nerve transection model to examine the effects of MB on RGC survival, the expression of regenerative marker GAP-43 in RGCs and microglial activation. MAIN METHODS Axons of RGCs were injured by cutting the optic nerve. MB was injected intravitreally either immediately post-injury or delayed to 3 days post-injury. Using immunohistochemical staining, surviving RGCs, GAP-43-positive RGCs and microglial cells were quantified in wholemount retinas 7 days post-injury. KEY FINDINGS Both immediate and delayed (a more clinically realistic situation) intravitreal injection of MB promoted RGC survival. MB also increased the number of GAP-43-positive RGCs, suggesting an enhanced ability of RGCs to regenerate. This was exemplified by the regenerative sprouting of axon-like processes from injured RGCs after MB treatment. The increase in RGC survival and GAP-43 expression correlated with an increase in the number of microglial cells. SIGNIFICANCE These results reveal that MB has survival-promoting and growth-promoting effects on RGCs after optic nerve injury. Together with the established safety profile of MB in humans, MB is a promising treatment for neurodegeneration and injury of the optic nerve.
Collapse
Affiliation(s)
- Jacqueline C L Fung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Eric Y P Cho
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
10
|
Fernández JC, Peláez R, Rey-Funes M, Soliño M, Contartese DS, Dorfman VB, López-Costa JJ, Larrayoz IM, Loidl CF, Martínez A. Methylene Blue Prevents Retinal Damage Caused by Perinatal Asphyxia in the Rat. Front Cell Neurosci 2020; 14:157. [PMID: 32581722 PMCID: PMC7289067 DOI: 10.3389/fncel.2020.00157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/12/2020] [Indexed: 01/24/2023] Open
Abstract
Perinatal asphyxia (PA) is responsible for a large proportion of neonatal deaths and numerous neurological sequelae, including visual dysfunction and blindness. In PA, the retina is exposed to ischemia/reoxygenation, which results in nitric oxide (NO) overproduction and neurotoxicity. We hypothesized that methylene blue (MB), a guanylyl cyclase inhibitor, and free-radical scavenger currently used in the clinic, may block this pathway and prevent PA-induced retinal degeneration. Male rat pups were subjected to an experimental model of PA. Four groups were studied: normally delivered (CTL), normally delivered treated with 2 mg Kg-1 MB (MB), exposed to PA for 20 min at 37°C (PA), and exposed to PA and, then, treated with MB (PA-MB). Scotopic electroretinography performed 45 days after birth showed that PA animals had significant defects in the a- and b-waves and oscillatory potentials (OP). The same animals presented a significant increase in the thickness of the inner retina and a large number of TUNEL-positive cells. All these physiological and morphological parameters were significantly prevented by the treatment with MB. Gene expression analysis demonstrated significant increases in iNOS, MMP9, and VEGF in the eyes of PA animals, which were prevented by MB treatment. In conclusion, MB regulates key players of inflammation, matrix remodeling, gliosis, and angiogenesis in the eye and could be used as a treatment to prevent the deleterious visual consequences of PA. Given its safety profile and low cost, MB may be used clinically in places where alternative treatments may be unavailable.
Collapse
Affiliation(s)
- Juan Carlos Fernández
- Instituto de Biología Celular y Neurociencia "Prof. E. de Robertis", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Primera Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rafael Peláez
- Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Manuel Rey-Funes
- Instituto de Biología Celular y Neurociencia "Prof. E. de Robertis", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Manuel Soliño
- Instituto de Biología Celular y Neurociencia "Prof. E. de Robertis", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela S Contartese
- Instituto de Biología Celular y Neurociencia "Prof. E. de Robertis", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Verónica B Dorfman
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Buenos Aires, Argentina
| | - Juan José López-Costa
- Instituto de Biología Celular y Neurociencia "Prof. E. de Robertis", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - César F Loidl
- Instituto de Biología Celular y Neurociencia "Prof. E. de Robertis", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alfredo Martínez
- Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| |
Collapse
|
11
|
Auchter AM, Barrett DW, Monfils MH, Gonzalez-Lima F. Methylene Blue Preserves Cytochrome Oxidase Activity and Prevents Neurodegeneration and Memory Impairment in Rats With Chronic Cerebral Hypoperfusion. Front Cell Neurosci 2020; 14:130. [PMID: 32508596 PMCID: PMC7251060 DOI: 10.3389/fncel.2020.00130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 04/20/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic cerebral hypoperfusion in neurocognitive disorders diminishes cytochrome oxidase activity leading to neurodegenerative effects and impairment of learning and memory. Methylene blue at low doses stimulates cytochrome oxidase activity and may thus counteract the adverse effects of cerebral hypoperfusion. However, the effects of methylene blue on cytochrome oxidase activity during chronic cerebral hypoperfusion have not been described before. To test this hypothesis, rats underwent bilateral carotid artery occlusion or sham surgery, received daily 4 mg/kg methylene blue or saline injections, and learned a visual water task. Brain mapping of cytochrome oxidase activity was done by quantitative enzyme histochemistry. Permanent carotid occlusion for 1 month resulted in decreased cytochrome oxidase activity in visual cortex, prefrontal cortex, perirhinal cortex, hippocampus and amygdala, and weaker interregional correlation of cytochrome oxidase activity between these regions. Methylene blue preserved cytochrome oxidase activity in regions affected by carotid occlusion and strengthened their interregional correlations of cytochrome oxidase activity, which prevented neurodegenerative effects and facilitated task-specific learning and memory. Brain-behavior correlations revealed positive correlations between performance and brain regions in which cytochrome oxidase activity was preserved by methylene blue. These results are the first to demonstrate that methylene blue prevents neurodegeneration and memory impairment by preserving cytochrome oxidase activity and interregional correlation of cytochrome oxidase activity in brain regions susceptible to chronic hypoperfusion. This demonstration provides further support for the hypothesis that lower cerebral blood flow results in an Alzheimer's-like syndrome and that stimulating cytochrome oxidase activity with low-dose methylene blue is neuroprotective.
Collapse
Affiliation(s)
| | | | | | - F. Gonzalez-Lima
- Department of Psychology, Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
12
|
Sasaoka M, Ota T, Kageyama M. Rotenone-induced inner retinal degeneration via presynaptic activation of voltage-dependent sodium and L-type calcium channels in rats. Sci Rep 2020; 10:969. [PMID: 31969611 PMCID: PMC6976703 DOI: 10.1038/s41598-020-57638-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/31/2019] [Indexed: 01/11/2023] Open
Abstract
Rotenone, a mitochondrial complex I inhibitor, causes retinal degeneration via unknown mechanisms. To elucidate the molecular mechanisms of its action, we further characterized a rat model of rotenone-induced retinal degeneration. Intravitreal injection of rotenone (2 nmol/eye) damaged mainly the inner retinal layers, including cell loss in the ganglion cell and inner nuclear layers, which were very similar to those induced by 10 nmol/eye N-methyl-D-aspartate (NMDA). These morphological changes were accompanied by the reduced b-wave amplitude of electroretinogram, and increased immunostaining of 2,4-dinitrophenyl, an oxidative stress marker. Rotenone also downregulated expression of neurofilament light-chain gene (Nfl) as a retinal ganglion cell (RGC) marker. This effect was prevented by simultaneous injection of rotenone with antioxidants or NMDA receptor antagonists. More importantly, voltage-dependent sodium and L-type calcium channel blockers and intracellular calcium signaling modulators remarkably suppressed rotenone-induced Nfl downregulation, whereas none of these agents modified NMDA-induced Nfl downregulation. These results suggest that rotenone-induced inner retinal degeneration stems from indirect postsynaptic NMDA stimulation that is triggered by oxidative stress-mediated presynaptic intracellular calcium signaling via activation of voltage-dependent sodium and L-type calcium channels.
Collapse
Affiliation(s)
- Masaaki Sasaoka
- Global Alliances and External Research, Santen Pharmaceutical Co., Ltd., Ikoma-shi, Nara, 630-0101, Japan
| | - Takashi Ota
- Global Alliances and External Research, Santen Pharmaceutical Co., Ltd., Ikoma-shi, Nara, 630-0101, Japan
| | - Masaaki Kageyama
- Global Alliances and External Research, Santen Pharmaceutical Co., Ltd., Ikoma-shi, Nara, 630-0101, Japan.
| |
Collapse
|
13
|
Berrocal M, Caballero-Bermejo M, Gutierrez-Merino C, Mata AM. Methylene Blue Blocks and Reverses the Inhibitory Effect of Tau on PMCA Function. Int J Mol Sci 2019; 20:ijms20143521. [PMID: 31323781 PMCID: PMC6678728 DOI: 10.3390/ijms20143521] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/05/2019] [Accepted: 07/11/2019] [Indexed: 12/24/2022] Open
Abstract
Methylene blue (MB) is a synthetic phenothiazine dye that, in the last years, has generated much debate about whether it could be a useful therapeutic drug for tau-related pathologies, such as Alzheimer’s disease (AD). However, the molecular mechanism of action is far from clear. Recently we reported that MB activates the plasma membrane Ca2+-ATPase (PMCA) in membranes from human and pig tissues and from cells cultures, and that it could protect against inactivation of PMCA by amyloid β-peptide (Aβ). The purpose of the present study is to further examine whether the MB could also modulate the inhibitory effect of tau, another key molecular marker of AD, on PMCA activity. By using kinetic assays in membranes from several tissues and cell cultures, we found that this phenothiazine was able to block and even to completely reverse the inhibitory effect of tau on PMCA. The results of this work point out that MB could mediate the toxic effect of tau related to the deregulation of calcium homeostasis by blocking the impairment of PMCA activity by tau. We then could conclude that MB could interfere with the toxic effects of tau by restoring the function of PMCA pump as a fine tuner of calcium homeostasis.
Collapse
Affiliation(s)
- Maria Berrocal
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura and Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Montaña Caballero-Bermejo
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura and Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Carlos Gutierrez-Merino
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura and Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Ana M Mata
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura and Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain.
| |
Collapse
|
14
|
Gureev AP, Shaforostova EA, Laver DA, Khorolskaya VG, Syromyatnikov MY, Popov VN. Methylene blue elicits non-genotoxic H 2O 2 production and protects brain mitochondria from rotenone toxicity. J Appl Biomed 2019; 17:107-114. [DOI: 10.32725/jab.2019.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
|
15
|
Mekala NK, Kurdys J, Depuydt MM, Vazquez EJ, Rosca MG. Apoptosis inducing factor deficiency causes retinal photoreceptor degeneration. The protective role of the redox compound methylene blue. Redox Biol 2018; 20:107-117. [PMID: 30300862 PMCID: PMC6175772 DOI: 10.1016/j.redox.2018.09.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 01/06/2023] Open
Abstract
Dysfunction in mitochondrial oxidative phosphorylation (OXPHOS) underlies a wide spectrum of human ailments known as mitochondrial diseases. Deficiencies in complex I of the electron transport chain (ETC) contribute to 30–40% of all cases of mitochondrial diseases, and leads to eye disease including optic nerve atrophy and retinal degeneration. The mechanisms responsible for organ damage in mitochondrial defects may include energy deficit, oxidative stress, and an increase in the NADH/NAD+ redox ratio due to decreased NAD+ regeneration. Currently, there is no effective treatment to alleviate human disease induced by complex I defect. Photoreceptor cells have the highest energy demand and dependence on OXPHOS for survival, and the lowest reserve capacity indicating that they are sensitive to OXPHOS defects. We investigated the effect of mitochondrial OXPHOS deficiency on retinal photoreceptors in a model of mitochondrial complex I defect (apoptosis inducing factor, AIF-deficient mice, Harlequin mice), and tested the protective effect of a mitochondrial redox compound (methylene blue, MB) on mitochondrial and photoreceptor integrity. MB prevented the reduction in the retinal thickness and protein markers for photoreceptor outer segments, Muller and ganglion cells, and altered mitochondrial integrity and function induced by AIF deficiency. In rotenone-induced complex I deficient 661 W cells (an immortalized mouse photoreceptor cell line) MB decreased the NADH/NAD+ ratio and oxidative stress without correcting the energy deficit, and improved cell survival. MB deactivated the mitochondrial stress response pathways, the unfolding protein response and mitophagy. In conclusion, preserving mitochondrial structure and function alleviates retinal photoreceptor degeneration in mitochondrial complex I defect. Mitochondrial complex I causes damage of the retinal photoreceptor cells and their outer segments. Methylene blue decreases the NADH/ NAD+ ratio and oxidative stress induced by complex I defect. Methylene blue deactivates the mitochondrial stress response pathways. Methylene blue maintains mitochondrial integrity and function. Methylene blue improves photoreceptor cell survival and outer segment integrity.
Collapse
Affiliation(s)
- Naveen K Mekala
- Department of Foundational Sciences at Central Michigan University College of Medicine, Mount Pleasant, MI, United States
| | - Jacob Kurdys
- Department of Foundational Sciences at Central Michigan University College of Medicine, Mount Pleasant, MI, United States
| | - Mikayla M Depuydt
- Department of Foundational Sciences at Central Michigan University College of Medicine, Mount Pleasant, MI, United States
| | - Edwin J Vazquez
- Department of Foundational Sciences at Central Michigan University College of Medicine, Mount Pleasant, MI, United States
| | - Mariana G Rosca
- Department of Foundational Sciences at Central Michigan University College of Medicine, Mount Pleasant, MI, United States.
| |
Collapse
|
16
|
Cui ZQ, Li WL, Luo Y, Yang JP, Qu ZZ, Zhao WQ. Methylene Blue Exerts Anticonvulsant and Neuroprotective Effects on Self-Sustaining Status Epilepticus (SSSE) Induced by Prolonged Basolateral Amygdala Stimulation in Wistar Rats. Med Sci Monit 2018; 24:161-169. [PMID: 29307885 PMCID: PMC5771162 DOI: 10.12659/msm.907758] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/18/2017] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND This study was designed to investigate the potential anticonvulsant and neuroprotective effects of methylene blue (MB) on self-sustaining status epilepticus (SSSE) induced by prolonged basolateral amygdala stimulation (BLA) in Wistar rats. MATERIAL AND METHODS The rats were randomly divided into 4 groups: (1) the Control group (rats without any treatment); (2) the Sham group (rats received electrode implantation but without electrical stimulation); (3) the SSSE group (rats received electrode implantation and additional electrical stimulation); and (4) the SSSE+MB group (rats received 1 mg/kg MB intraperitoneal injection 5 min after SSSE). SSSE models were established by prolonged BLA stimulation. The severities of SSSE were assessed by the number of separate seizures and the accumulated time of seizures. The variations of malondialdehyde/glutathione (MDA/GSH) were assessed 24 h after the establishment of SSSE. Nissl staining was performed to detect the surviving neurons in hippocampal CA1 and CA3 regions, and Western blotting assays were used to detect Caspase-3 (CASP3), B cell lymphoma 2 (BCL2), and BCL2-associated X protein (BAX). RESULTS Compared with the SSSE group, treatment with MB (1) markedly reduced the number and accumulated time of seizure activities; (2) significantly attenuated the increase of MDA and the decrease of GSH hippocampal levels; (3) markedly improved the cell morphology and alleviated the neuronal loss in hippocampal CA1 and CA3 regions; (4) significantly attenuated the increase of CASP3 and BAX and the decrease of BCL2 hippocampal levels. CONCLUSIONS MB has a protective effect in the SSSE model and may be useful as an adjuvant for preventing or treating epilepsy in humans.
Collapse
Affiliation(s)
- Zhi-qiang Cui
- Faculty of Graduate Studies, Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Wen-ling Li
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Yan Luo
- Department of Reproductive Genetic, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Ji-peng Yang
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Zhen-zhen Qu
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Wen-qing Zhao
- Faculty of Graduate Studies, Hebei Medical University, Shijiazhuang, Hebei, P.R. China
- Department of Functional Neurosurgery, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| |
Collapse
|
17
|
Hypothermia Prevents Retinal Damage Generated by Optic Nerve Trauma in the Rat. Sci Rep 2017; 7:6966. [PMID: 28761115 PMCID: PMC5537267 DOI: 10.1038/s41598-017-07294-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/26/2017] [Indexed: 11/11/2022] Open
Abstract
Ocular and periocular traumatisms may result in loss of vision. Hypothermia provides a beneficial intervention for brain and heart conditions and, here, we study whether hypothermia can prevent retinal damage caused by traumatic neuropathy. Intraorbital optic nerve crush (IONC) or sham manipulation was applied to male rats. Some animals were subjected to hypothermia (8 °C) for 3 h following surgery. Thirty days later, animals were subjected to electroretinography and behavioral tests. IONC treatment resulted in amplitude reduction of the b-wave and oscillatory potentials of the electroretinogram, whereas the hypothermic treatment significantly (p < 0.05) reversed this process. Using a descending method of limits in a two-choice visual task apparatus, we demonstrated that hypothermia significantly (p < 0.001) preserved visual acuity. Furthermore, IONC-treated rats had a lower (p < 0.0001) number of retinal ganglion cells and a higher (p < 0.0001) number of TUNEL-positive cells than sham-operated controls. These numbers were significantly (p < 0.0001) corrected by hypothermic treatment. There was a significant (p < 0.001) increase of RNA-binding motif protein 3 (RBM3) and of BCL2 (p < 0.01) mRNA expression in the eyes exposed to hypothermia. In conclusion, hypothermia constitutes an efficacious treatment for traumatic vision-impairing conditions, and the cold-shock protein pathway may be involved in mediating the beneficial effects shown in the retina.
Collapse
|
18
|
Gureev AP, Syromyatnikov MY, Gorbacheva TM, Starkov AA, Popov VN. Methylene blue improves sensorimotor phenotype and decreases anxiety in parallel with activating brain mitochondria biogenesis in mid-age mice. Neurosci Res 2016; 113:19-27. [DOI: 10.1016/j.neures.2016.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/19/2016] [Accepted: 07/29/2016] [Indexed: 01/31/2023]
|
19
|
Bittner GD, Spaeth CS, Poon AD, Burgess ZS, McGill CH. Repair of traumatic plasmalemmal damage to neurons and other eukaryotic cells. Neural Regen Res 2016; 11:1033-42. [PMID: 27630671 PMCID: PMC4994430 DOI: 10.4103/1673-5374.187019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The repair (sealing) of plasmalemmal damage, consisting of small holes to complete transections, is critical for cell survival, especially for neurons that rarely regenerate cell bodies. We first describe and evaluate different measures of cell sealing. Some measures, including morphological/ultra-structural observations, membrane potential, and input resistance, provide very ambiguous assessments of plasmalemmal sealing. In contrast, measures of ionic current flow and dye barriers can, if appropriately used, provide more accurate assessments. We describe the effects of various substances (calcium, calpains, cytoskeletal proteins, ESCRT proteins, mUNC-13, NSF, PEG) and biochemical pathways (PKA, PKC, PLC, Epac, cytosolic oxidation) on plasmalemmal sealing probability, and suggest that substances, pathways, and cellular events associated with plasmalemmal sealing have undergone a very conservative evolution. During sealing, calcium ion influx mobilizes vesicles and other membranous structures (lysosomes, mitochondria, etc.) in a continuous fashion to form a vesicular plug that gradually restricts diffusion of increasingly smaller molecules and ions over a period of seconds to minutes. Furthermore, we find no direct evidence that sealing occurs through the collapse and fusion of severed plasmalemmal leaflets, or in a single step involving the fusion of one large wound vesicle with the nearby, undamaged plasmalemma. We describe how increases in perikaryal calcium levels following axonal transection account for observations that cell body survival decreases the closer an axon is transected to the perikaryon. Finally, we speculate on relationships between plasmalemmal sealing, Wallerian degeneration, and the ability of polyethylene glycol (PEG) to seal cell membranes and rejoin severed axonal ends – an important consideration for the future treatment of trauma to peripheral nerves. A better knowledge of biochemical pathways and cytoplasmic structures involved in plasmalemmal sealing might provide insights to develop treatments for traumatic nerve injuries, stroke, muscular dystrophy, and other pathologies.
Collapse
Affiliation(s)
- George D Bittner
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | | | - Andrew D Poon
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Zachary S Burgess
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | | |
Collapse
|
20
|
Lopez Sanchez M, Crowston J, Mackey D, Trounce I. Emerging Mitochondrial Therapeutic Targets in Optic Neuropathies. Pharmacol Ther 2016; 165:132-52. [DOI: 10.1016/j.pharmthera.2016.06.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Indexed: 12/14/2022]
|
21
|
Gueven N, Nadikudi M, Daniel A, Chhetri J. Targeting mitochondrial function to treat optic neuropathy. Mitochondrion 2016; 36:7-14. [PMID: 27476756 DOI: 10.1016/j.mito.2016.07.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 01/03/2023]
Abstract
Many reports have illustrated a tight connection between vision and mitochondrial function. Not only are most mitochondrial diseases associated with some form of vision impairment, many ophthalmological disorders such as glaucoma, age-related macular degeneration and diabetic retinopathy also show signs of mitochondrial dysfunction. Despite a vast amount of evidence, vision loss is still only treated symptomatically, which is only partially a consequence of resistance to acknowledge that mitochondria could be the common denominator and hence a promising therapeutic target. More importantly, clinical support of this concept is only emerging. Moreover, only a few drug candidates and treatment strategies are in development or approved that selectively aim to restore mitochondrial function. This review rationalizes the currently developed therapeutic approaches that target mitochondrial function by discussing their proposed mode(s) of action and provides an overview on their development status with regards to optic neuropathies.
Collapse
Affiliation(s)
- Nuri Gueven
- Pharmacy, School of Medicine, University of Tasmania, Hobart, TAS, Australia.
| | - Monila Nadikudi
- Pharmacy, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Abraham Daniel
- Pharmacy, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Jamuna Chhetri
- Pharmacy, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
22
|
Rodriguez P, Zhao J, Milman B, Tiwari YV, Duong TQ. Methylene blue and normobaric hyperoxia combination therapy in experimental ischemic stroke. Brain Behav 2016; 6:e00478. [PMID: 27458543 PMCID: PMC4951618 DOI: 10.1002/brb3.478] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/11/2016] [Accepted: 03/18/2016] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Ischemic stroke is a global burden that contributes to the disability and mortality of millions of patients. This study aimed to evaluate the efficacy of combined MB (methylene blue) and NBO (normobaric hyperoxia) therapy in experimental ischemic stroke. METHODS Rats with transient (60 min) MCAO (middle cerebral artery occlusion) were treated with: (1) air + vehicle (N = 8), (2) air + MB (N = 8), (3) NBO + vehicle (N = 7), and (4) NBO + MB (N = 9). MB (1 mg/kg) was administered at 30 min, again on days 2, 7, and 14 after stroke. NBO was given during MRI (30-150 min) on day 0, and again 1 h each during MRI on subsequent days. Serial diffusion, perfusion and T2 MRI were performed to evaluate lesion volumes. Foot-fault and cylinder tests were performed to evaluate sensorimotor function. RESULTS The major findings were: (1) NBO + MB therapy showed a greater decrease in infarct volume compared to NBO alone, but similar infarct volume compared to MB alone, (2) NBO + MB therapy accelerated sensorimotor functional recovery compared to NBO or MB alone, (3) Infarct volumes on day 2 did not change significantly from those on day 28 for all four groups, but behavioral function continued to show improved recovery in the NBO + MB group. CONCLUSIONS These findings support the hypothesis that combined NBO + MB further improves functional outcome and reduces infarct volume compared to either treatment alone and these improvements extended up to 28 days.
Collapse
Affiliation(s)
- Pavel Rodriguez
- Research Imaging InstituteUniversity of Texas Health Science CenterSan AntonioTexas
- Department of RadiologyUniversity of Texas Health Science CenterSan AntonioTexas
| | - Jiang Zhao
- Research Imaging InstituteUniversity of Texas Health Science CenterSan AntonioTexas
- Department of Anatomy and EmbryologyPeking University Health Science CenterBeijingChina
| | - Brian Milman
- Research Imaging InstituteUniversity of Texas Health Science CenterSan AntonioTexas
| | - Yash Vardhan Tiwari
- Research Imaging InstituteUniversity of Texas Health Science CenterSan AntonioTexas
- Department of Biomedical EngineeringUniversity of TexasSan AntonioTexas
| | - Timothy Q. Duong
- Research Imaging InstituteUniversity of Texas Health Science CenterSan AntonioTexas
| |
Collapse
|
23
|
Rey-Funes M, Larrayoz IM, Fernández JC, Contartese DS, Rolón F, Inserra PIF, Martínez-Murillo R, López-Costa JJ, Dorfman VB, Martínez A, Loidl CF. Methylene blue prevents retinal damage in an experimental model of ischemic proliferative retinopathy. Am J Physiol Regul Integr Comp Physiol 2016; 310:R1011-9. [PMID: 26984891 DOI: 10.1152/ajpregu.00266.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 03/11/2016] [Indexed: 11/22/2022]
Abstract
Perinatal asphyxia induces retinal lesions, generating ischemic proliferative retinopathy, which may result in blindness. Previously, we showed that the nitrergic system was involved in the physiopathology of perinatal asphyxia. Here we analyze the application of methylene blue, a well-known soluble guanylate cyclase inhibitor, as a therapeutic strategy to prevent retinopathy. Male rats (n = 28 per group) were treated in different ways: 1) control group comprised born-to-term animals; 2) methylene blue group comprised animals born from pregnant rats treated with methylene blue (2 mg/kg) 30 and 5 min before delivery; 3) perinatal asphyxia (PA) group comprised rats exposed to perinatal asphyxia (20 min at 37°C); and 4) methylene blue-PA group comprised animals born from pregnant rats treated with methylene blue (2 mg/kg) 30 and 5 min before delivery, and then the pups were subjected to PA as above. For molecular studies, mRNA was obtained at different times after asphyxia, and tissue was collected at 30 days for morphological and biochemical analysis. Perinatal asphyxia produced significant gliosis, angiogenesis, and thickening of the inner retina. Methylene blue treatment reduced these parameters. Perinatal asphyxia resulted in a significant elevation of the nitrergic system as shown by NO synthase (NOS) activity assays, Western blotting, and (immuno)histochemistry for the neuronal isoform of NOS and NADPH-diaphorase activity. All these parameters were also normalized by the treatment. In addition, methylene blue induced the upregulation of the anti-angiogenic peptide, pigment epithelium-derived factor. Application of methylene blue reduced morphological and biochemical parameters of retinopathy. This finding suggests the use of methylene blue as a new treatment to prevent or decrease retinal damage in the context of ischemic proliferative retinopathy.
Collapse
Affiliation(s)
- Manuel Rey-Funes
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Ignacio M Larrayoz
- Angiogenesis Study Group, Center for Biomedical Research of La Rioja, Logroño, Spain;
| | - Juan C Fernández
- Primera Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela S Contartese
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Federico Rolón
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Pablo I F Inserra
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| | - Ricardo Martínez-Murillo
- Neurovascular Research Group, Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Consejo Superior Investigaciones Científicas, Madrid, Spain; and
| | - Juan J López-Costa
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Verónica B Dorfman
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| | - Alfredo Martínez
- Angiogenesis Study Group, Center for Biomedical Research of La Rioja, Logroño, Spain
| | - César F Loidl
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina; Laboratorio de Neurociencia, Facultad de Ciencias Médicas, Universidad Católica de Cuyo, San Juan, Argentina
| |
Collapse
|
24
|
Kuliaviene I, Baniene R, Virketyte S, Kincius M, Jansen E, Gulbinas A, Kupcinskas L, Trumbeckaite S, Borutaite V. Methylene blue attenuates mitochondrial dysfunction of rat kidney during experimental acute pancreatitis. J Dig Dis 2016; 17:186-92. [PMID: 26861116 DOI: 10.1111/1751-2980.12328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/22/2016] [Accepted: 02/05/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The disturbance of mitochondrial functions has been considered as one of the mechanisms of pathogenesis of acute pancreatitis (AP) followed by kidney failure. This study was aimed to investigate the effects of methylene blue (MB) on pancreas and kidney mitochondrial respiratory functions during experimental acute pancreatitis in rats. METHODS AP was induced by administrating sodium taurocholate into the pancreatic duct of male Wistar rats. The rats were divided into three groups: the MB group, MB (5 mg/kg) was injected intravenously 10 min prior to AP induction; the AP group, saline solution was injected intravenously 10 min prior to AP induction; and the sham operation group, isotonic sodium chlorine was used instead of sodium taurocholate. The animals were sacrificed after 24 h. The pancreas and kidney were removed for mitochondrial assay by oxygraphic and spectrophotometric methods. RESULTS Intravenous injection of MB did not prevent AP-induced inhibition of pancreatic mitochondrial respiration; however, MB significantly improved kidney mitochondrial respiratory functions with complex I-dependent substrates glutamate and malate. The activity of complex I of mitochondria isolated from AP-damaged kidney was increased after pretreatment with MB. However, MB did not affect AP-inhibited kidney mitochondrial respiration with succinate. MB had no protective effects on amylase activity or on urea content in serum in AP. CONCLUSION The disturbances of kidney mitochondrial energy metabolism in experimental model of severe AP can be ameliorated by MB administration.
Collapse
Affiliation(s)
- Irma Kuliaviene
- Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rasa Baniene
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Simona Virketyte
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Marius Kincius
- Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Eugene Jansen
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Antanas Gulbinas
- Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Limas Kupcinskas
- Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Sonata Trumbeckaite
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
25
|
Ghergherehchi CL, Bittner GD, Hastings RL, Mikesh M, Riley DC, Trevino RC, Schallert T, Thayer WP, Bhupanapadu Sunkesula SR, Ha TAN, Munoz N, Pyarali M, Bansal A, Poon AD, Mazal AT, Smith TA, Wong NS, Dunne PJ. Effects of extracellular calcium and surgical techniques on restoration of axonal continuity by polyethylene glycol fusion following complete cut or crush severance of rat sciatic nerves. J Neurosci Res 2016; 94:231-45. [PMID: 26728662 DOI: 10.1002/jnr.23704] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/12/2015] [Accepted: 12/03/2015] [Indexed: 01/10/2023]
Abstract
Complete crush or cut severance of sciatic nerve axons in rats and other mammals produces immediate loss of axonal continuity. Loss of locomotor functions subserved by those axons is restored only after months, if ever, by outgrowths regenerating at ∼1 mm/day from the proximal stumps of severed axonal segments. The distal stump of a severed axon typically begins to degenerate in 1-3 days. We recently developed a polyethylene glycol (PEG) fusion technology, consisting of sequential exposure of severed axonal ends to hypotonic Ca(2+) -free saline, methylene blue, PEG in distilled water, and finally Ca(2+) -containing isotonic saline. This study examines factors that affect the PEG fusion restoration of axonal continuity within minutes, as measured by conduction of action potentials and diffusion of an intracellular fluorescent dye across the lesion site of rat sciatic nerves completely cut or crush severed in the midthigh. Also examined are factors that affect the longer-term PEG fusion restoration of lost behavioral functions within days to weeks, as measured by the sciatic functional index. We report that exposure of cut-severed axonal ends to Ca(2+) -containing saline prior to PEG fusion and stretch/tension of proximal or distal axonal segments of cut-severed axons decrease PEG fusion success. Conversely, trimming cut-severed ends in Ca(2+) -free saline just prior to PEG fusion increases PEG fusion success. PEG fusion prevents or retards the Wallerian degeneration of cut-severed axons, as assessed by measures of axon diameter and G ratio. PEG fusion may produce a paradigm shift in the treatment of peripheral nerve injuries. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - George D Bittner
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | | | - Michelle Mikesh
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - D Colton Riley
- Department of Neuroscience, University of Texas at Austin, Austin, Texas.,Department of Plastic Surgery, Vanderbilt School of Medicine, Nashville, Tennessee
| | - Richard C Trevino
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Tim Schallert
- Department of Psychology, University of Texas at Austin, Austin, Texas
| | - Wesley P Thayer
- Department of Plastic Surgery, Vanderbilt School of Medicine, Nashville, Tennessee
| | | | - Tu-Anh N Ha
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Nicolas Munoz
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Monika Pyarali
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Aakarshita Bansal
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Andrew D Poon
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Alexander T Mazal
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Tyler A Smith
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Nicole S Wong
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - Patrick J Dunne
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| |
Collapse
|
26
|
Abstract
Stroke is a leading cause of death and long-term disability. Methylene blue, 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 methylene blue in experimental stroke models.
Collapse
Affiliation(s)
- Zhao Jiang
- Research Imaging Institute, Radiology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Timothy Q Duong
- Department of Ophthalmology, Radiology and Physiology, University of Texas Health Science Center, San Antonio, Texas, USA
| |
Collapse
|
27
|
Study of the transition to higher iodide in the malonic acid Briggs–Rauscher oscillator. REACTION KINETICS MECHANISMS AND CATALYSIS 2015. [DOI: 10.1007/s11144-015-0967-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Bittner GD, Rokkappanavar KK, Peduzzi JD. Application and implications of polyethylene glycol-fusion as a novel technology to repair injured spinal cords. Neural Regen Res 2015; 10:1406-8. [PMID: 26604897 PMCID: PMC4625502 DOI: 10.4103/1673-5374.162772] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- George D Bittner
- Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Kiran K Rokkappanavar
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jean D Peduzzi
- Department of Anatomy & Cell Biology, Department of Physical Medicine and Rehabilitation at Oakwood, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
29
|
Yang SH, Li W, Sumien N, Forster M, Simpkins JW, Liu R. Alternative mitochondrial electron transfer for the treatment of neurodegenerative diseases and cancers: Methylene blue connects the dots. Prog Neurobiol 2015; 157:273-291. [PMID: 26603930 DOI: 10.1016/j.pneurobio.2015.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/10/2015] [Accepted: 10/20/2015] [Indexed: 12/21/2022]
Abstract
Brain has exceptional high requirement for energy metabolism with glucose as the exclusive energy source. Decrease of brain energy metabolism and glucose uptake has been found in patients of Alzheimer's, Parkinson's and other neurodegenerative diseases, providing a clear link between neurodegenerative disorders and energy metabolism. On the other hand, cancers, including glioblastoma, have increased glucose uptake and rely on aerobic glycolysis for energy metabolism. The switch of high efficient oxidative phosphorylation to low efficient aerobic glycolysis pathway (Warburg effect) provides macromolecule for biosynthesis and proliferation. Current research indicates that methylene blue, a century old drug, can receive electron from NADH in the presence of complex I and donates it to cytochrome c, providing an alternative electron transfer pathway. Methylene blue increases oxygen consumption, decrease glycolysis, and increases glucose uptake in vitro. Methylene blue enhances glucose uptake and regional cerebral blood flow in rats upon acute treatment. In addition, methylene blue provides protective effect in neuron and astrocyte against various insults in vitro and in rodent models of Alzheimer's, Parkinson's, and Huntington's disease. In glioblastoma cells, methylene blue reverses Warburg effect by enhancing mitochondrial oxidative phosphorylation, arrests glioma cell cycle at s-phase, and inhibits glioma cell proliferation. Accordingly, methylene blue activates AMP-activated protein kinase, inhibits downstream acetyl-coA carboxylase and cyclin-dependent kinases. In summary, there is accumulating evidence providing a proof of concept that enhancement of mitochondrial oxidative phosphorylation via alternative mitochondrial electron transfer may offer protective action against neurodegenerative diseases and inhibit cancers proliferation.
Collapse
Affiliation(s)
- Shao-Hua Yang
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Wenjun Li
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Nathalie Sumien
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Michael Forster
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - James W Simpkins
- Department of Physiology and Pharmacology, Center for Neuroscience, Health Science Center, West Virginia University, Medical Center Drive, Morgantown, WV 26506, USA
| | - Ran Liu
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
30
|
Bittner GD, Sengelaub DR, Trevino RC, Peduzzi JD, Mikesh M, Ghergherehchi CL, Schallert T, Thayer WP. The curious ability of polyethylene glycol fusion technologies to restore lost behaviors after nerve severance. J Neurosci Res 2015; 94:207-30. [PMID: 26525605 DOI: 10.1002/jnr.23685] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 09/17/2015] [Accepted: 10/12/2015] [Indexed: 01/09/2023]
Abstract
Traumatic injuries to PNS and CNS axons are not uncommon. Restoration of lost behaviors following severance of mammalian peripheral nerve axons (PNAs) relies on regeneration by slow outgrowths and is typically poor or nonexistent when after ablation or injuries close to the soma. Behavioral recovery after severing spinal tract axons (STAs) is poor because STAs do not naturally regenerate. Current techniques to enhance PNA and/or STA regeneration have had limited success and do not prevent the onset of Wallerian degeneration of severed distal segments. This Review describes the use of a recently developed polyethylene glycol (PEG) fusion technology combining concepts from biochemical engineering, cell biology, and clinical microsurgery. Within minutes after microsuturing carefully trimmed cut ends and applying a well-specified sequence of solutions, PEG-fused axons exhibit morphological continuity (assessed by intra-axonal dye diffusion) and electrophysiological continuity (assessed by conduction of action potentials) across the lesion site. Wallerian degeneration of PEG-fused PNAs is greatly reduced as measured by counts of sensory and/or motor axons and maintenance of axonal diameters and neuromuscular synapses. After PEG-fusion repair, cut-severed, crush-severed, or ablated PNAs or crush-severed STAs rapidly (within days to weeks), more completely, and permanently restore PNA- or STA-mediated behaviors compared with nontreated or conventionally treated animals. PEG-fusion success is enhanced or decreased by applying antioxidants or oxidants, trimming cut ends or stretching axons, and exposure to Ca(2+) -free or Ca(2+) -containing solutions, respectively. PEG-fusion technology employs surgical techniques and chemicals already used by clinicians and has the potential to produce a paradigm shift in the treatment of traumatic injuries to PNAs and STAs.
Collapse
Affiliation(s)
- G D Bittner
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - D R Sengelaub
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - R C Trevino
- Department of Orthopedic Surgery, Wellspan Health, York, Pennsylvania
| | - J D Peduzzi
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan
| | - M Mikesh
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - C L Ghergherehchi
- Department of Neuroscience, University of Texas at Austin, Austin, Texas
| | - T Schallert
- Department of Psychology, University of Texas at Austin, Austin, Texas
| | - W P Thayer
- Department of Plastic Surgery, Vanderbilt School of Medicine, Nashville, Tennessee
| |
Collapse
|
31
|
Lu Q, Tucker D, Dong Y, Zhao N, Zhang Q. Neuroprotective and Functional Improvement Effects of Methylene Blue in Global Cerebral Ischemia. Mol Neurobiol 2015; 53:5344-55. [PMID: 26433378 DOI: 10.1007/s12035-015-9455-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/25/2015] [Indexed: 11/25/2022]
Abstract
Transient global cerebral ischemia (GCI) causes delayed neuronal cell death in the vulnerable hippocampus CA1 subfield, as well as behavioral deficits. Ischemia reperfusion (I/R) produces excessive reactive oxygen species and plays a key role in brain injury. The mitochondrial electron respiratory chain is the main cellular source of free radical generation, and dysfunction of mitochondria has a significant impact on the neuronal cell death in ischemic brain. The aim of the present study is to investigate the potential beneficial effects of methylene blue (MB) in a four-vessel occlusion (4VO) GCI model on adult male rats. MB was delivered at a dose of 0.5 mg/kg/day for 7 days, through a mini-pump implanted subcutaneously after GCI. We first found that MB significantly improved ischemic neuronal survival in the hippocampal CA1 region as measured by cresyl violet staining as well as NeuN staining. We also found that MB has the ability to rescue ischemia-induced decreases of cytochrome c oxidase activity and ATP generation in the CA1 region following I/R. Further analysis with labeling of MitoTracker® Red revealed that the depolarization of mitochondrial membrane potential (MMP) was markedly attenuated following MB treatment. In addition, the induction of caspase-3, caspase-8, and caspase-9 activities and the increased numbers of TUNEL-positive cells of the CA1 region were significantly reduced by MB application. Correspondingly, Barnes maze tests showed that the deterioration of spatial learning and memory performance following GCI was significantly improved in the MB-treatment group compared to the ischemic control group. In summary, our study suggests that MB may be a promising therapeutic agent targeting neuronal cell death and cognitive deficits following transient global cerebral ischemia.
Collapse
Affiliation(s)
- Qing Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA
| | - Donovan Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA
| | - Ningjun Zhao
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA.
| |
Collapse
|
32
|
Talley Watts L, Long JA, Boggs RC, Manga H, Huang S, Shen Q, Duong TQ. Delayed Methylene Blue Improves Lesion Volume, Multi-Parametric Quantitative Magnetic Resonance Imaging Measurements, and Behavioral Outcome after Traumatic Brain Injury. J Neurotrauma 2015; 33:194-202. [PMID: 25961471 DOI: 10.1089/neu.2015.3904] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) remains a primary cause of death and disability in both civilian and military populations worldwide. There is a critical need for the development of neuroprotective agents that can circumvent damage and provide functional recovery. We previously showed that methylene blue (MB), a U.S. Food and Drug Administration-grandfathered drug with energy-enhancing and antioxidant properties, given 1 and 3 h post-TBI, had neuroprotective effects in rats. This study aimed to further investigate the neuroprotection of delayed MB treatment (24 h postinjury) post-TBI as measured by lesion volume and functional outcomes. Comparisons were made with vehicle and acute MB treatment. Multi-modal magnetic resonance imaging and behavioral studies were performed at 1 and 3 h and 2, 7, and 14 days after an impact to the primary forelimb somatosensory cortex. We found that delaying MB treatment 24 h postinjury still minimized lesion volume and functional deficits, compared to vehicle-treated animals. The data further support the potential for MB as a neuroprotective treatment, especially when medical teatment is not readily available. MB has an excellent safety profile and is clinically approved for other indications. MB clinical trials on TBI can thus be readily explored.
Collapse
Affiliation(s)
- Lora Talley Watts
- 1 Research Imaging Institute, University of Texas Health Science Center , San Antonio, Texas.,2 Departments of Cellular and Structure Biology, University of Texas Health Science Center , San Antonio, Texas.,3 Department of Neurology, University of Texas Health Science Center , San Antonio, Texas
| | - Justin Alexander Long
- 1 Research Imaging Institute, University of Texas Health Science Center , San Antonio, Texas
| | - Robert Cole Boggs
- 1 Research Imaging Institute, University of Texas Health Science Center , San Antonio, Texas
| | - Hemanth Manga
- 1 Research Imaging Institute, University of Texas Health Science Center , San Antonio, Texas
| | - Shiliang Huang
- 1 Research Imaging Institute, University of Texas Health Science Center , San Antonio, Texas
| | - Qiang Shen
- 1 Research Imaging Institute, University of Texas Health Science Center , San Antonio, Texas
| | - Timothy Q Duong
- 1 Research Imaging Institute, University of Texas Health Science Center , San Antonio, Texas.,3 Department of Neurology, University of Texas Health Science Center , San Antonio, Texas.,4 Department of Ophthalmology, University of Texas Health Science Center , San Antonio, Texas.,5 Research Division, South Texas Veterans Health Care System , San Antonio, Texas
| |
Collapse
|
33
|
Gonzalez-Lima F, Auchter A. Protection against neurodegeneration with low-dose methylene blue and near-infrared light. Front Cell Neurosci 2015; 9:179. [PMID: 26029050 PMCID: PMC4428125 DOI: 10.3389/fncel.2015.00179] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 04/24/2015] [Indexed: 12/21/2022] Open
Affiliation(s)
- F Gonzalez-Lima
- Department of Psychology and Institute for Neuroscience, University of Texas at Austin Austin, TX, USA
| | - Allison Auchter
- Department of Psychology and Institute for Neuroscience, University of Texas at Austin Austin, TX, USA
| |
Collapse
|
34
|
Tretter L, Horvath G, Hölgyesi A, Essek F, Adam-Vizi V. Enhanced hydrogen peroxide generation accompanies the beneficial bioenergetic effects of methylene blue in isolated brain mitochondria. Free Radic Biol Med 2014; 77:317-30. [PMID: 25277417 DOI: 10.1016/j.freeradbiomed.2014.09.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 09/02/2014] [Accepted: 09/18/2014] [Indexed: 12/21/2022]
Abstract
The redox dye methylene blue (MB) is proven to have beneficial effects in various models of neurodegenerative diseases. Here we investigated the effects of MB (100 nM, 300 nM, and 1 μM) on key bioenergetic parameters and on H2O2 production/elimination in isolated guinea pig brain mitochondria under normal as well as respiration-impaired conditions. As measured by high-resolution Oxygraph the rate of resting oxygen consumption was increased, but the ADP-stimulated respiration was unaffected by MB with any of the substrates (glutamate malate, succinate, or α-glycerophosphate) used for supporting mitochondrial respiration. In mitochondria treated with inhibitors of complex I or complex III MB moderately but significantly increased the rate of ATP production, restored ΔΨm, and increased the rate of Ca(2+) uptake. The effects of MB are consistent with transferring electrons from upstream components of the electron transport chain to cytochrome c, which is energetically favorable when the flow of electrons in the respiratory chain is compromised. On the other hand, MB significantly increased the production of H2O2 measured by Amplex UltraRed fluorimetry under all conditions, in resting, ATP-synthesizing, and respiration-impaired mitochondria, with each substrate combination supporting respiration. Furthermore, it also decreased the elimination of H2O2. Generation of H2O2 without superoxide formation, observed in the presence of MB, is interpreted as a result of reduction of molecular oxygen to H2O2 by the reduced MB. The elevated generation and impaired elimination of H2O2 should be considered for the overall oxidative state of mitochondria treated with MB.
Collapse
Affiliation(s)
- L Tretter
- MTA-SE Laboratory for Neurobiochemistry, Department of Medical Biochemistry, Semmelweis University, Budapest H-1094, Hungary
| | - G Horvath
- MTA-SE Laboratory for Neurobiochemistry, Department of Medical Biochemistry, Semmelweis University, Budapest H-1094, Hungary
| | - A Hölgyesi
- MTA-SE Laboratory for Neurobiochemistry, Department of Medical Biochemistry, Semmelweis University, Budapest H-1094, Hungary
| | - F Essek
- MTA-SE Laboratory for Neurobiochemistry, Department of Medical Biochemistry, Semmelweis University, Budapest H-1094, Hungary
| | - V Adam-Vizi
- MTA-SE Laboratory for Neurobiochemistry, Department of Medical Biochemistry, Semmelweis University, Budapest H-1094, Hungary.
| |
Collapse
|
35
|
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. [PMID: 25218555 DOI: 10.1016/j.brainres.2014.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/12/2014] [Accepted: 09/02/2014] [Indexed: 11/29/2022]
Abstract
Stroke is a leading cause of morbidity and mortality in the world. Low-dose methylene blue (MB), which has been used safely to treat methemoglobinemia and cyanide poisoning in humans, has energy enhancing and antioxidant properties. We tested the hypothesis that methylene blue 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. Serial MRI was used to evaluate MB treatment efficacy. The major findings were: (i) MB significantly prolonged the perfusion-diffusion mismatch, (ii) MB mildly increased the CBF in the hypoperfused tissue, (iii) MB did not change the final infarct volume in permanent ischemic stroke, and (iv) there were no dose-dependent effects on mismatch progression for the 1 and 3mg/kg doses studied. This neuroprotective effect is likely the result of sustained ATP production and increased CBF to tissue at risk. This work has the potential to readily lead to clinical stroke trials given MB's excellent safety profile.
Collapse
Affiliation(s)
- Pavel Rodriguez
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Zhao Jiang
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States; Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Shiliang Huang
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Qiang Shen
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Timothy Q Duong
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.
| |
Collapse
|
36
|
Attenuation of noise-induced hearing loss using methylene blue. Cell Death Dis 2014; 5:e1200. [PMID: 24763057 PMCID: PMC4001318 DOI: 10.1038/cddis.2014.170] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 01/05/2023]
Abstract
The overproduction of reactive oxygen species (ROS) and reactive nitrogen species (RNS) has been known to contribute to the pathogenesis of noise-induced hearing loss. In this study, we discovered that in BALB/c mice pretreatment with methylene blue (MB) for 4 consecutive days significantly protected against cochlear injury by intense broad-band noise for 3 h. It decreased both compound threshold shift and permanent threshold shift and, further, reduced outer hair cell death in the cochlea. MB also reduced ROS and RNS formation after noise exposure. Furthermore, it protected against rotenone- and antimycin A-induced cell death and also reversed ATP generation in the in vitro UB-OC1 cell system. Likewise, MB effectively attenuated the noise-induced impairment of complex IV activity in the cochlea. In addition, it increased the neurotrophin-3 (NT-3) level, which could affect the synaptic connections between hair cells and spiral ganglion neurons in the noise-exposed cochlea, and also promoted the conservation of both efferent and afferent nerve terminals on the outer and inner hair cells. These findings suggest that the amelioration of impaired mitochondrial electron transport and the potentiation of NT-3 expression by treatment with MB have a significant therapeutic value in preventing ROS-mediated sensorineural hearing loss.
Collapse
|
37
|
Gonzalez-Lima F, Barksdale BR, Rojas JC. Mitochondrial respiration as a target for neuroprotection and cognitive enhancement. Biochem Pharmacol 2014; 88:584-93. [DOI: 10.1016/j.bcp.2013.11.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/16/2013] [Accepted: 11/18/2013] [Indexed: 10/25/2022]
|
38
|
Willis GL, Moore C, Armstrong SM. Parkinson's disease, lights and melanocytes: looking beyond the retina. Sci Rep 2014; 4:3921. [PMID: 24473093 PMCID: PMC5379242 DOI: 10.1038/srep03921] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 01/13/2014] [Indexed: 12/11/2022] Open
Abstract
Critical analysis of recent research suggesting that light pollution causes Parkinson's disease (PD) reveals that such a hypothesis is unsustainable in the context of therapeutic use of light in treating various neuropsychiatric conditions. Reinterpretation of their findings suggests that retinal damage caused by prolonged light exposure may have contributed to the observed enhancement of experimental PD. To test this hypothesis further, forty-two Sprague Dawley rats received microinjections of 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenyl-2, 4, 6-tetrahydropyridine (MPTP), paraquat or rotenone into the vitreal mass in doses so minute that the effects could not be attributed to diffusion into brain. Significant changes in five motor parameters consistent with symptoms of experimental PD were observed. These findings support the interpretation that the retina is involved in the control of motor function and in the aetiology of PD.
Collapse
Affiliation(s)
- Gregory L. Willis
- Neurosciences Section, The Bronowski Institute of Behavioural Neuroscience, Coliban Medical Centre, 19 Jennings Street, Kyneton, Victoria 3444, Australia
| | - Cleo Moore
- Neurosciences Section, The Bronowski Institute of Behavioural Neuroscience, Coliban Medical Centre, 19 Jennings Street, Kyneton, Victoria 3444, Australia
| | - Stuart Maxwell Armstrong
- Neurosciences Section, The Bronowski Institute of Behavioural Neuroscience, Coliban Medical Centre, 19 Jennings Street, Kyneton, Victoria 3444, Australia
| |
Collapse
|
39
|
Neurological and psychological applications of transcranial lasers and LEDs. Biochem Pharmacol 2013; 86:447-57. [PMID: 23806754 DOI: 10.1016/j.bcp.2013.06.012] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/14/2013] [Accepted: 06/15/2013] [Indexed: 11/23/2022]
Abstract
Transcranial brain stimulation with low-level light/laser therapy (LLLT) is the use of directional low-power and high-fluency monochromatic or quasimonochromatic light from lasers or LEDs in the red-to-near-infrared wavelengths to modulate a neurobiological function or induce a neurotherapeutic effect in a nondestructive and non-thermal manner. The mechanism of action of LLLT is based on photon energy absorption by cytochrome oxidase, the terminal enzyme in the mitochondrial respiratory chain. Cytochrome oxidase has a key role in neuronal physiology, as it serves as an interface between oxidative energy metabolism and cell survival signaling pathways. Cytochrome oxidase is an ideal target for cognitive enhancement, as its expression reflects the changes in metabolic capacity underlying higher-order brain functions. This review provides an update on new findings on the neurotherapeutic applications of LLLT. The photochemical mechanisms supporting its cognitive-enhancing and brain-stimulatory effects in animal models and humans are discussed. LLLT is a potential non-invasive treatment for cognitive impairment and other deficits associated with chronic neurological conditions, such as large vessel and lacunar hypoperfusion or neurodegeneration. Brain photobiomodulation with LLLT is paralleled by pharmacological effects of low-dose USP methylene blue, a non-photic electron donor with the ability to stimulate cytochrome oxidase activity, redox and free radical processes. Both interventions provide neuroprotection and cognitive enhancement by facilitating mitochondrial respiration, with hormetic dose-response effects and brain region activational specificity. This evidence supports enhancement of mitochondrial respiratory function as a generalizable therapeutic principle relevant to highly adaptable systems that are exquisitely sensitive to energy availability such as the nervous system.
Collapse
|
40
|
Richardson TE, Kelly HN, Yu AE, Simpkins JW. Therapeutic strategies in Friedreich's ataxia. Brain Res 2013; 1514:91-7. [PMID: 23587934 PMCID: PMC4461031 DOI: 10.1016/j.brainres.2013.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 04/02/2013] [Accepted: 04/02/2013] [Indexed: 10/26/2022]
Abstract
First established as a diagnosis by Nikolaus Friedreich in 1863, Friedreich's ataxia (FA) is an autosomal recessive progressive neurodegenerative disorder cause by a trinucleotide repeat expansion. FA begins with the functional absence of the FXN gene product frataxin, a protein whose exact function still remains unknown. This absence results in impaired intracellular antioxidant defenses, dysregulation of iron-sulfur cluster proteins, depression of aerobic electron transport chain respiration, massive mitochondrial dysfunction, and ultimately cell death in the brain, spinal cord and heart. Herein, we review the molecular and cellular pathogenesis leading to widespread organ system dysfunction, as well as current therapeutic research aimed at preventing the debilitating effects of frataxin loss and preventing the signs and symptoms associated of FA. We also discuss the ongoing treatment strategies employed by our laboratory to prevent mitochondrial damage using synergistic effects of 17β-estradiol and methylene blue, previously shown by our group and others to have protective effects in human FA fibroblasts. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- Timothy E. Richardson
- Institute for Aging and Alzheimer’s Disease Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Heather N. Kelly
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Amanda E. Yu
- Institute for Aging and Alzheimer’s Disease Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - James W. Simpkins
- Institute for Aging and Alzheimer’s Disease Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
41
|
Poteet E, Winters A, Yan LJ, Shufelt K, Green KN, Simpkins JW, Wen Y, Yang SH. Neuroprotective actions of methylene blue and its derivatives. PLoS One 2012; 7:e48279. [PMID: 23118969 PMCID: PMC3485214 DOI: 10.1371/journal.pone.0048279] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 09/21/2012] [Indexed: 01/08/2023] Open
Abstract
Methylene blue (MB), the first lead chemical structure of phenothiazine and other derivatives, is commonly used in diagnostic procedures and as a treatment for methemoglobinemia. We have previously demonstrated that MB could function as an alternative mitochondrial electron transfer carrier, enhance cellular oxygen consumption, and provide protection in vitro and in rodent models of Parkinson's disease and stroke. In the present study, we investigated the structure-activity relationships of MB in vitro using MB and six structurally related compounds. MB reduces mitochondrial superoxide production via alternative electron transfer that bypasses mitochondrial complexes I-III. MB mitigates reactive free radical production and provides neuroprotection in HT-22 cells against glutamate, IAA and rotenone toxicity. Distinctly, MB provides no protection against direct oxidative stress induced by glucose oxidase. Substitution of a side chain at MB's 10-nitrogen rendered a 1000-fold reduction of the protective potency against glutamate neurototoxicity. Compounds without side chains at positions 3 and 7, chlorophenothiazine and phenothiazine, have distinct redox potentials compared to MB and are incapable of enhancing mitochondrial electron transfer, while obtaining direct antioxidant actions against glutamate, IAA, and rotenone insults. Chlorophenothiazine exhibited direct antioxidant actions in mitochondria lysate assay compared to MB, which required reduction by NADH and mitochondria. MB increased complex IV expression and activity, while 2-chlorphenothiazine had no effect. Our study indicated that MB could attenuate superoxide production by functioning as an alternative mitochondrial electron transfer carrier and as a regenerable anti-oxidant in mitochondria.
Collapse
Affiliation(s)
- Ethan Poteet
- Department of Pharmacology and Neuroscience, Institute for Alzheimer’s Disease and Aging Research, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States of America
| | - Ali Winters
- Department of Pharmacology and Neuroscience, Institute for Alzheimer’s Disease and Aging Research, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States of America
| | - Liang-Jun Yan
- Department of Pharmacology and Neuroscience, Institute for Alzheimer’s Disease and Aging Research, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States of America
| | - Kyle Shufelt
- Department of Chemistry, Texas Christian University, Fort Worth, Texas, United States of America
| | - Kayla N. Green
- Department of Chemistry, Texas Christian University, Fort Worth, Texas, United States of America
| | - James W. Simpkins
- Department of Pharmacology and Neuroscience, Institute for Alzheimer’s Disease and Aging Research, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States of America
| | - Yi Wen
- Department of Pharmacology and Neuroscience, Institute for Alzheimer’s Disease and Aging Research, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States of America
| | - Shao-Hua Yang
- Department of Pharmacology and Neuroscience, Institute for Alzheimer’s Disease and Aging Research, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States of America
- * E-mail:
| |
Collapse
|
42
|
Methylene blue treatment for vasoplegia and resultant isoelectric processed EEG (Bispectral Index). J Clin Anesth 2012; 24:511-3. [DOI: 10.1016/j.jclinane.2011.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 09/07/2011] [Accepted: 09/09/2011] [Indexed: 11/23/2022]
|
43
|
Ex vivo models to evaluate the role of ocular melanin in trans-scleral drug delivery. Eur J Pharm Sci 2012; 46:475-83. [DOI: 10.1016/j.ejps.2012.03.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 03/22/2012] [Accepted: 03/25/2012] [Indexed: 11/19/2022]
|
44
|
Fabian E, Reglodi D, Mester L, Szabo A, Szabadfi K, Tamas A, Toth G, Kovacs K. Effects of PACAP on intracellular signaling pathways in human retinal pigment epithelial cells exposed to oxidative stress. J Mol Neurosci 2012; 48:493-500. [PMID: 22644900 DOI: 10.1007/s12031-012-9812-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 05/08/2012] [Indexed: 12/20/2022]
Abstract
The integrity of retinal pigment epithelial cells is critical for photoreceptor survival and vision. Pituitary adenylate cyclase activating polypeptide (PACAP) exerts retinoprotective effects against several types of injuries in vivo, including optic nerve transection, retinal ischemia, excitotoxic injuries, UVA-induced lesion, and diabetic retinopathy. In a recent study, we have proven that PACAP is also protective in oxidative stress-induced injury in human pigment epithelial cells (ARPE-19 cells). The aim of the present study was to investigate the possible mechanisms of this protection. ARPE cells were exposed to a 24-h hydrogen peroxide treatment. Expressions of kinases and apoptotic markers were studied by complex array kits and Western blot. Oxidative stress induced the activation of several apoptotic markers, including Bad, Bax, HIF-1α, several heat shock proteins, TNF-related apoptosis-inducing ligand, and Fas-associated protein with death domain, while PACAP treatment decreased them. The changes in the expression of MAP kinases showed that PACAP activated the protective ERK1/2 and downstream CREB, and decreased the activation of the pro-apoptotic p38MAPK and c-Jun N-terminal kinase, an effect opposite to that observed with only oxidative stress. Furthermore, PACAP increased the activation of the protective Akt pathway. In addition, the effects of oxidative stress on several other signaling molecules were counteracted by PACAP treatment (Chk2, Yes, Lyn, paxillin, p53, PLC, STAT4, RSK). These play a role in cell death, cell cycle, inflammation, adhesion, differentiation and proliferation. In summary, PACAP, acting at several levels, influences the balance between pro- and anti-apoptotic factors in favor of anti-apoptosis, thereby providing protection in oxidative stress-induced injury of human retinal pigment epithelial cells.
Collapse
Affiliation(s)
- E Fabian
- Department of Anatomy, PTE-MTA Lendulet PACAP Research Team, University of Pecs, 7624 Pecs, Szigeti u 12, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Congdon EE, Wu JW, Myeku N, Figueroa YH, Herman M, Marinec PS, Gestwicki JE, Dickey CA, Yu WH, Duff KE. Methylthioninium chloride (methylene blue) induces autophagy and attenuates tauopathy in vitro and in vivo. Autophagy 2012; 8:609-22. [PMID: 22361619 DOI: 10.4161/auto.19048] [Citation(s) in RCA: 224] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
More than 30 neurodegenerative diseases including Alzheimer disease (AD), frontotemporal lobe dementia (FTD), and some forms of Parkinson disease (PD) are characterized by the accumulation of an aggregated form of the microtubule-binding protein tau in neurites and as intracellular lesions called neurofibrillary tangles. Diseases with abnormal tau as part of the pathology are collectively known as the tauopathies. Methylthioninium chloride, also known as methylene blue (MB), has been shown to reduce tau levels in vitro and in vivo and several different mechanisms of action have been proposed. Herein we demonstrate that autophagy is a novel mechanism by which MB can reduce tau levels. Incubation with nanomolar concentrations of MB was sufficient to significantly reduce levels of tau both in organotypic brain slice cultures from a mouse model of FTD, and in cell models. Concomitantly, MB treatment altered the levels of LC3-II, cathepsin D, BECN1, and p62 suggesting that it was a potent inducer of autophagy. Further analysis of the signaling pathways induced by MB suggested a mode of action similar to rapamycin. Results were recapitulated in a transgenic mouse model of tauopathy administered MB orally at three different doses for two weeks. These data support the use of this drug as a therapeutic agent in neurodegenerative diseases.
Collapse
Affiliation(s)
- Erin E Congdon
- Taub Institute/Department of Pathology, Columbia University and Department of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bittner G, Keating C, Kane J, Britt J, Spaeth C, Fan J, Zuzek A, Wilcott R, Thayer W, Winograd J, Gonzalez-Lima F, Schallert T. Rapid, effective, and long-lasting behavioral recovery produced by microsutures, methylene blue, and polyethylene glycol after completely cutting rat sciatic nerves. J Neurosci Res 2012; 90:967-80. [DOI: 10.1002/jnr.23023] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 12/04/2011] [Accepted: 12/13/2011] [Indexed: 01/05/2023]
|
47
|
Spaeth CS, Robison T, Fan JD, Bittner GD. Cellular mechanisms of plasmalemmal sealing and axonal repair by polyethylene glycol and methylene blue. J Neurosci Res 2012; 90:955-66. [PMID: 22302626 DOI: 10.1002/jnr.23022] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 12/04/2011] [Accepted: 12/15/2011] [Indexed: 11/09/2022]
Abstract
Mammalian neurons and all other eukaryotic cells endogenously repair traumatic injury within minutes by a Ca²⁺-induced accumulation of vesicles that interact and fuse with each other and the plasmalemma to seal any openings. We have used uptake or exclusion of extracellular fluorescent dye to measure the ability of rat hippocampal B104 cells or rat sciatic nerves to repair (seal) transected neurites in vitro or transected axons ex vivo. We report that endogenous sealing in both preparations is enhanced by Ca²⁺-containing solutions and is decreased by Ca²⁺-free solutions containing antioxidants such as dithiothreitol (DTT), melatonin (MEL), methylene blue (MB), and various toxins that decrease vesicular interactions. In contrast, the fusogen polyethylene glycol (PEG) at 10-50 mM artificially seals the cut ends of B104 cells and rat sciatic axons within seconds and is not affected by Ca²⁺ or any of the substances that affect endogenous sealing. At higher concentrations, PEG decreases sealing of transected axons and disrupts the plasmalemma of intact cells. These PEG-sealing data are consistent with the hypothesis that lower concentrations of PEG directly seal a damaged plasmalemma. We have considered these and other data to devise a protocol using a well-specified series of solutions that vary in tonicity, Ca²⁺, MB, and PEG content. These protocols rapidly and consistently repair (PEG-fuse) rat sciatic axons in completely cut sciatic nerves in vivo rapidly and dramatically to restore long-lasting morphological continuity, action potential conduction, and behavioral functions.
Collapse
Affiliation(s)
- C S Spaeth
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| | | | | | | |
Collapse
|
48
|
Rojas JC, Bruchey AK, Gonzalez-Lima F. Neurometabolic mechanisms for memory enhancement and neuroprotection of methylene blue. Prog Neurobiol 2011; 96:32-45. [PMID: 22067440 DOI: 10.1016/j.pneurobio.2011.10.007] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 10/26/2011] [Accepted: 10/27/2011] [Indexed: 12/21/2022]
Abstract
This paper provides the first review of the memory-enhancing and neuroprotective metabolic mechanisms of action of methylene blue in vivo. These mechanisms have important implications as a new neurobiological approach to improve normal memory and to treat memory impairment and neurodegeneration associated with mitochondrial dysfunction. Methylene blue's action is unique because its neurobiological effects are not determined by regular drug-receptor interactions or drug-response paradigms. Methylene blue shows a hormetic dose-response, with opposite effects at low and high doses. At low doses, methylene blue is an electron cycler in the mitochondrial electron transport chain, with unparalleled antioxidant and cell respiration-enhancing properties that affect the function of the nervous system in a versatile manner. A major role of the respiratory enzyme cytochrome oxidase on the memory-enhancing effects of methylene blue is supported by available data. The memory-enhancing effects have been associated with improvement of memory consolidation in a network-specific and use-dependent fashion. In addition, low doses of methylene blue have also been used for neuroprotection against mitochondrial dysfunction in humans and experimental models of disease. The unique auto-oxidizing property of methylene blue and its pleiotropic effects on a number of tissue oxidases explain its potent neuroprotective effects at low doses. The evidence reviewed supports a mechanistic role of low-dose methylene blue as a promising and safe intervention for improving memory and for the treatment of acute and chronic conditions characterized by increased oxidative stress, neurodegeneration and memory impairment.
Collapse
Affiliation(s)
- Julio C Rojas
- Departments of Psychology, Pharmacology and Toxicology, University of Texas at Austin, 1 University Station A8000, Austin, TX 78712, USA
| | | | | |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW The concept of neuroprotective therapy for glaucoma is that damage to retinal ganglion cells (RGCs) may be prevented by intervening in neuronal death pathways. This review focuses on strategies for neuroprotection and summarizes preclinical studies that have investigated potential agents over the last 2 years. RECENT FINDINGS Part of the challenge of studies in neuroprotection has been the utilization of an animal model that resembles human glaucoma. Several models have been utilized including acute and chronic intraocular pressure elevation, the DBA/2J mouse, optic nerve axotomy and crush. NMDA inhibitors continued to be explored however with limited success in human trials. Memantine failed to demonstrate neuroprotection in phase III clinical trials. Although its mechanism of neuroprotection has not been fully elaborated, topical brimonidine has shown some neuroprotective benefits. Exogeneous neurotrophins delay, but do not prevent, RGC death. Bioenergetic neuroprotection that is enhancing the energy supply to RGC has been explored with benefits in animal models. Other strategies include TNF-α, modulation of the immune system and inflammation, and blocking apoptotic signals and stem cells. SUMMARY Animal models of glaucoma and neuroprotective strategies continue to be refined. Establishing consensus guidelines for the execution and design of translational research in neuroprotection may optimize the facilitation of neuroprotection research.
Collapse
|
50
|
Riha PD, Rojas JC, Gonzalez-Lima F. Beneficial network effects of methylene blue in an amnestic model. Neuroimage 2010; 54:2623-34. [PMID: 21087672 DOI: 10.1016/j.neuroimage.2010.11.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 11/03/2010] [Accepted: 11/08/2010] [Indexed: 11/29/2022] Open
Abstract
Posterior cingulate/retrosplenial cortex (PCC) hypometabolism is a common feature in amnestic mild cognitive impairment and Alzheimer's disease. In rats, PCC hypometabolism induced by mitochondrial dysfunction induces oxidative damage, neurodegeneration and memory deficits. USP methylene blue (MB) is a diaminophenothiazine drug with antioxidant and metabolic-enhancing properties. In rats, MB facilitates memory and prevents neurodegeneration induced by mitochondrial dysfunction. This study tested the memory-enhancing properties of systemic MB in rats that received an infusion of sodium azide, a cytochrome oxidase inhibitor, directly into the PCC. Lesion volumes were estimated with unbiased stereology. MB's network-level mechanism of action was analyzed using graph theory and structural equation modeling based on cytochrome oxidase histochemistry-derived metabolic mapping data. Sodium azide infusions induced PCC hypometabolism and impaired visuospatial memory in a holeboard food-search task. Isolated PCC cytochrome oxidase inhibition disrupted the cingulo-thalamo-hippocampal effective connectivity, decreased the PCC functional networks and created functional redundancy within the thalamus. An intraperitoneal dose of 4 mg/kg MB prevented the memory impairment, reduced the PCC metabolic lesion volume and partially restored the cingulo-thalamo-hippocampal network effects. The effects of MB were dependent upon the local sub-network necessary for memory retrieval. The data support that MB's metabolic-enhancing effects are contingent upon the neural context, and that MB is able to boost coherent and orchestrated adaptations in response to physical alterations to the network involved in visuospatial memory. These results implicate MB as a candidate intervention to improve memory. Because of its neuroprotective properties, MB may have disease-modifying effects in amnestic conditions associated with hypometabolism.
Collapse
Affiliation(s)
- Penny D Riha
- Departments of Psychology, Pharmacology and Toxicology, University of Texas at Austin, Austin, TX 78712, USA
| | | | | |
Collapse
|