1
|
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Cárcamo-Fonfría A, Caballero-Muñoz MDM, Agúndez JAG. Oxidative Stress in Huntington's Disease. Biomolecules 2025; 15:527. [PMID: 40305278 PMCID: PMC12025275 DOI: 10.3390/biom15040527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Although the pathogenesis of the neurodegenerative phenomena of Huntington's disease (HD) is not well known, in the last 30 years, numerous data have been published that suggest a possible role of oxidative stress. The majority of studies regarding this issue were performed in different experimental models of this disease (neurotoxic models such as intraperitoneal injection of 3-nitropropionic acid or intrastriatal injection of quinolinic acid, transgenic animal models for HD, and cell cultures) and, less frequently, in samples of brain tissue, plasma/serum, blood cells, and other tissues from patients with a genetic-molecular diagnosis of presymptomatic and symptomatic HD compared to healthy controls. In this narrative review, we have summarized the data from the main studies in which oxidative stress parameters have been measured both in patients with HD and in experimental models of the same disease, as well as the few studies on gene variants involved in oxidative stress in patients with HD. Most studies addressing this issue in experimental models of HD have shown an increase in markers or oxidative stress, a decrease in antioxidant substances, or both. However, the results of studies on patients with HD have not been conclusive as few studies have been published on the matter. However, a meta-analysis of blood studies on HD patients (including a pool of serum and blood cell studies) has shown an increase in lipid peroxidation markers, OH8dG concentrations, and GPx activity and a decrease in GSH levels. Future prospective and multicenter studies with a long-term follow-up period involving a large number of HD patients and healthy controls are needed to address this topic.
Collapse
Affiliation(s)
- Félix Javier Jiménez-Jiménez
- Section of Neurology, Hospital Universitario del Sureste, Arganda del Rey, 28500 Madrid, Spain; (H.A.-N.); (A.C.-F.); (M.d.M.C.-M.)
| | - Hortensia Alonso-Navarro
- Section of Neurology, Hospital Universitario del Sureste, Arganda del Rey, 28500 Madrid, Spain; (H.A.-N.); (A.C.-F.); (M.d.M.C.-M.)
| | - Elena García-Martín
- University Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| | - Alba Cárcamo-Fonfría
- Section of Neurology, Hospital Universitario del Sureste, Arganda del Rey, 28500 Madrid, Spain; (H.A.-N.); (A.C.-F.); (M.d.M.C.-M.)
| | - María del Mar Caballero-Muñoz
- Section of Neurology, Hospital Universitario del Sureste, Arganda del Rey, 28500 Madrid, Spain; (H.A.-N.); (A.C.-F.); (M.d.M.C.-M.)
| | - José A. G. Agúndez
- University Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| |
Collapse
|
2
|
Bernal López OE, San Juan D, Vidal BE, Collado-Corona MÁ. History of Clinical Neurophysiology in Mexico. J Clin Neurophysiol 2025:00004691-990000000-00214. [PMID: 40105714 DOI: 10.1097/wnp.0000000000001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
SUMMARY In the 18th century, Luigi Galvani proposed the hypothesis of animal electricity, which is produced by the brain and distributed through the nerves to the muscles. This was the cornerstone of what is known today as the modern study of nerve function, earning him the title of the Father of Clinical Neurophysiology. The 19th century was subsequently marked by two major figures: Santiago Ramón y Cajal (Neuron Theory) and Hans Berger, known for describing cerebral electrical activity and recording the first electroencephalograms. In Mexico, Clinical Neurophysiology emerged in the late 19th century and consolidated itself in the first half of the 20th century. In the year of 1938, Dr. Clemente Robles and Teodoro Flores Covarrubias built the first electroencephalograph, marking the beginning of the era of Clinical Neurophysiology. Initially, this diagnostic tool was primarily applied to psychiatric patients, as there was no clear separation between psychiatry and neurology and patients were treated jointly at the largest psychiatric center of that time, "La Castañeda." In 1968, the Mexican Society of Electroencephalography A.C. was founded and later changed its name to the Mexican Society of Clinical Neurophysiology A.C. Simultaneously, its members achieved universal recognition of the medical specialty, which has become established in clinical practice and has shown progressive academic and scientific growth in Mexico.
Collapse
Affiliation(s)
- Oscar Eduardo Bernal López
- Laboratory of Clinical Neurophysiology, National Institute of Medical Sciences and Nutrition Dr. Salvador Zubirán, Mexico City, Mexico
| | - Daniel San Juan
- Epilepsy Clinic, National Institute of Neurology and Neurosurgery Dr. Manuel Velasco Suárez, Mexico City, Mexico
| | - Bruno Estañol Vidal
- Chief of the Laboratory of Clinical Neurophysiology, National Institute of Medical Sciences and Nutrition Dr. Salvador Zubirán, Mexico City, Mexico; and
| | | |
Collapse
|
3
|
Wang N, Jia Y, Zhou X, Wang X, Zhou H, Xiao N. Effects of Repetitive Transcranial Magnetic Stimulation on Pallidum GABAergic Neurons and Motor Function in Rat Models of Kernicterus. Brain Sci 2023; 13:1252. [PMID: 37759853 PMCID: PMC10526431 DOI: 10.3390/brainsci13091252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Kernicterus is a serious complication of hyperbilirubinemia, caused by neuronal injury due to excessive unconjugated bilirubin (UCB) in specific brain areas. This injury induced by this accumulation in the globus pallidus can induce severe motor dysfunction. Repetitive transcranial magnetic stimulation (rTMS) has shown neuroprotective effects in various neurological diseases. This study aimed to investigate the effects of rTMS on pallidal nerve damage and motor dysfunction in a rat model of kernicterus. Rats were divided into a sham group (n = 16), a model group (bilirubin with sham rTMS; n = 16) and an rTMS group (bilirubin with rTMS; n = 16). High-frequency rTMS (10 Hz) was applied starting from 24 h postmodeling for 7 days. The rotarod test, western blotting and immunohistochemical staining were performed to measure motor function and protein expression levels. The rTMS mitigated the negative effects of UCB on the general health of kernicterus-model rats and improved their growth and development. Furthermore, the rTMS alleviated UCB-induced motor dysfunction and increased the expression of GABAergic neuronal marker GAD67 in the globus pallidus. Notably, it also inhibited apoptosis-related protein caspase-3 activation. In conclusion, rTMS could alleviate motor dysfunction by inhibiting apoptosis and increasing globus pallidus GAD67 in kernicterus rat models, indicating that it may be a promising treatment for kernicterus.
Collapse
Affiliation(s)
| | | | | | | | | | - Nong Xiao
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400010, China; (N.W.); (Y.J.); (X.Z.); (X.W.); (H.Z.)
| |
Collapse
|
4
|
Stanojevic JB, Zeljkovic M, Dragic M, Stojanovic IR, Ilic TV, Stevanovic ID, Ninkovic MB. Intermittent theta burst stimulation attenuates oxidative stress and reactive astrogliosis in the streptozotocin-induced model of Alzheimer's disease-like pathology. Front Aging Neurosci 2023; 15:1161678. [PMID: 37273654 PMCID: PMC10233102 DOI: 10.3389/fnagi.2023.1161678] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/10/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction Intracerebroventricularly (icv) injected streptozotocin (STZ) is a widely used model for sporadic Alzheimer's disease (sAD)-like pathology, marked by oxidative stress-mediated pathological progression. Intermittent theta burst stimulation (iTBS) is a noninvasive technique for brain activity stimulation with the ability to induce long-term potentiation-like plasticity and represents a promising treatment for several neurological diseases, including AD. The present study aims to investigate the effect of the iTBS protocol on the animal model of STZ-induced sAD-like pathology in the context of antioxidant, anti-inflammatory, and anti-amyloidogenic effects in the cortex, striatum, hippocampus, and cerebellum. Methods Male Wistar rats were divided into four experimental groups: control (icv normal saline solution), STZ (icv STZ-3 mg/kg), STZ + iTBS (STZ rats subjected to iTBS protocol), and STZ + Placebo (STZ animals subjected to placebo iTBS noise artifact). Biochemical assays and immunofluorescence microscopy were used to evaluate functional and structural changes. Results The icv STZ administration induces oxidative stress and attenuates antioxidative capacity in all examined brain regions. iTBS treatment significantly reduced oxidative and nitrosative stress parameters. Also, iTBS decreased Aβ-1-42 and APP levels. The iTBS enhances antioxidative capacity reported as elevated activity of its enzymatic and non-enzymatic components. In addition, iTBS elevated BDNF expression and attenuated STZ-induced astrogliosis confirmed by decreased GFAP+/VIM+/C3+ cell reactivity in the hippocampus. Discussion Our results provide experimental evidence for the beneficial effects of the applied iTBS protocol in attenuating oxidative stress, increasing antioxidant capacity and decreasing reactive astrogliosis in STZ-administrated rats.
Collapse
Affiliation(s)
- Jelena B. Stanojevic
- Institute for Biochemistry, Faculty of Medicine, University of Niš, Niš, Serbia
- Medical Faculty of Military Medical Academy, University of Defense, Belgrade, Serbia
| | - Milica Zeljkovic
- Laboratory for Neurobiology, Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Milorad Dragic
- Laboratory for Neurobiology, Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Ivana R. Stojanovic
- Institute for Biochemistry, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Tihomir V. Ilic
- Medical Faculty of Military Medical Academy, University of Defense, Belgrade, Serbia
| | - Ivana D. Stevanovic
- Medical Faculty of Military Medical Academy, University of Defense, Belgrade, Serbia
- Institute of Medical Research, Military Medical Academy, Belgrade, Serbia
| | - Milica B. Ninkovic
- Medical Faculty of Military Medical Academy, University of Defense, Belgrade, Serbia
- Institute of Medical Research, Military Medical Academy, Belgrade, Serbia
| |
Collapse
|
5
|
Peña-Toledo MA, Luque E, LaTorre M, Jimena I, Leiva-Cepas F, Ruz-Caracuel I, Agüera E, Peña-Amaro J, Tunez I. The ultrastructure of muscle fibers and satellite cells in experimental autoimmune encephalomyelitis after treatment with transcranial magnetic stimulation. Ultrastruct Pathol 2022; 46:401-412. [PMID: 35994513 DOI: 10.1080/01913123.2022.2112330] [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: 10/15/2022]
Abstract
In this study, we investigated the effect of transcranial magnetic stimulation (TMS) on the ultrastructure of muscle fibers and satellite cells in rats with experimental autoimmune encephalomyelitis (EAE). EAE-induced animals were treated with TMS (60 Hz at 0.7 mT) for 2 hours in the morning, once a day, 5 days a week, for 3 weeks, starting on day 15 post-immunization. The rats were sacrificed on day 36 post-immunization, and the soleus muscles were evaluated by light microscopy and transmission electron microscopy. Findings were compared with a non-treated EAE group. Electron microscopy analysis showed the presence of degenerated mitochondria, autophagic vacuoles, and altered myofibrils in non-treated EAE group. This correlates with the presence of acid phosphatase activity in muscle fibers and core-targetoid lesions with desmin immunohistochemistry. Most myonuclei in the EAE group showed apoptotic features. In contrast, EAE induced-TMS treated animals had less ultrastructural changes in the mitochondria and the myofibrils, together with less frequent apoptotic nuclear features. Peripheral desmin+ protrusions, as a marker of active satellite cells, were significantly increased in TMS-treated group. This correlates ultrastructurally with the presence of active features in satellite cells in the TMS group. In conclusion, the attenuation of ultrastructural alterations in muscle fibers and activation response of satellite cells caused by EAE indicated that skeletal muscle had a regenerative response to TMS.
Collapse
Affiliation(s)
- María Angeles Peña-Toledo
- Dementia and Multiple Sclerosis Unit, Neurology Service, Reina Sofia University Hospital, Cordoba, Spain.,Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain
| | - Evelio Luque
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain
| | - Manuel LaTorre
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain
| | - Ignacio Jimena
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain
| | - Fernando Leiva-Cepas
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain.,Department of Pathology, Reina Sofía University Hospital, Córdoba, Spain
| | - Ignacio Ruz-Caracuel
- Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain.,Department of Pathology, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - Eduardo Agüera
- Dementia and Multiple Sclerosis Unit, Neurology Service, Reina Sofia University Hospital, Cordoba, Spain.,Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain
| | - J Peña-Amaro
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain
| | - Isaac Tunez
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain.,Cooperative Research Thematic Excellent Network on Brain Stimulation (REDESTIM), Ministery for Economy, Industry and Competitiveness, Madrid, Spain
| |
Collapse
|
6
|
Caballero-Villarraso J, Medina FJ, Escribano BM, Agüera E, Santamaría A, Pascual-Leone A, Túnez I. Mechanisms Involved in Neuroprotective Effects of Transcranial Magnetic Stimulation. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:557-573. [PMID: 34370648 DOI: 10.2174/1871527320666210809121922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/05/2021] [Accepted: 04/25/2021] [Indexed: 11/22/2022]
Abstract
Transcranial Magnetic Stimulation (TMS) is widely used in neurophysiology to study cortical excitability. Research over the last few decades has highlighted its added value as a potential therapeutic tool in the treatment of a broad range of psychiatric disorders. More recently, a number of studies have reported beneficial and therapeutic effects for TMS in neurodegenerative conditions and strokes. Yet, despite its recognised clinical applications and considerable research using animal models, the molecular and physiological mechanisms through which TMS exerts its beneficial and therapeutic effects remain unclear. They are thought to involve biochemical-molecular events affecting membrane potential and gene expression. In this aspect, the dopaminergic system plays a special role. This is the most directly and selectively modulated neurotransmitter system, producing an increase in the flux of dopamine (DA) in various areas of the brain after the application of repetitive TMS (rTMS). Other neurotransmitters, such as glutamate and gamma-aminobutyric acid (GABA) have shown a paradoxical response to rTMS. In this way, their levels increased in the hippocampus and striatum but decreased in the hypothalamus and remained unchanged in the mesencephalon. Similarly, there are sufficient evidence that TMS up-regulates the gene expression of BDNF (one of the main brain neurotrophins). Something similar occurs with the expression of genes such as c-Fos and zif268 that encode trophic and regenerative action neuropeptides. Consequently, the application of TMS can promote the release of molecules involved in neuronal genesis and maintenance. This capacity may mean that TMS becomes a useful therapeutic resource to antagonize processes that underlie the previously mentioned neurodegenerative conditions.
Collapse
Affiliation(s)
- Javier Caballero-Villarraso
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Cordoba, Cordoba, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.,UGC Análisis Clínicos, Hospital Universitario Reina Sofía, Córdoba, Cordoba, Spain
| | - Francisco J Medina
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
| | - Begoña M Escribano
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.,Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Veterinaria, Universidad de Córdoba, Cordoba, Spain
| | - Eduardo Agüera
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.,UGC Neurología, Hospital Universitario Reina Sofía, Córdoba, Cordoba, Spain
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A. Mexico City, Mexico
| | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Instituto Guttman de Neurorrehabilitación, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Isaac Túnez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Cordoba, Cordoba, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
| |
Collapse
|
7
|
Uzair M, Abualait T, Arshad M, Yoo WK, Mir A, Bunyan RF, Bashir S. Transcranial magnetic stimulation in animal models of neurodegeneration. Neural Regen Res 2022; 17:251-265. [PMID: 34269184 PMCID: PMC8464007 DOI: 10.4103/1673-5374.317962] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/08/2020] [Accepted: 12/24/2020] [Indexed: 11/13/2022] Open
Abstract
Brain stimulation techniques offer powerful means of modulating the physiology of specific neural structures. In recent years, non-invasive brain stimulation techniques, such as transcranial magnetic stimulation (TMS) and transcranial direct current stimulation, have emerged as therapeutic tools for neurology and neuroscience. However, the possible repercussions of these techniques remain unclear, and there are few reports on the incisive recovery mechanisms through brain stimulation. Although several studies have recommended the use of non-invasive brain stimulation in clinical neuroscience, with a special emphasis on TMS, the suggested mechanisms of action have not been confirmed directly at the neural level. Insights into the neural mechanisms of non-invasive brain stimulation would unveil the strategies necessary to enhance the safety and efficacy of this progressive approach. Therefore, animal studies investigating the mechanisms of TMS-induced recovery at the neural level are crucial for the elaboration of non-invasive brain stimulation. Translational research done using animal models has several advantages and is able to investigate knowledge gaps by directly targeting neuronal levels. In this review, we have discussed the role of TMS in different animal models, the impact of animal studies on various disease states, and the findings regarding brain function of animal models after TMS in pharmacology research.
Collapse
Affiliation(s)
- Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Turki Abualait
- College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Muhammad Arshad
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Woo-Kyoung Yoo
- Department of Physical Medicine and Rehabilitation, Hallym University College of Medicine, Anyang, South Korea
- Hallym Institute for Translational Genomics & Bioinformatics, Hallym University College of Medicine, Anyang, South Korea
| | - Ali Mir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| | - Reem Fahd Bunyan
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
8
|
Bocci T, Baloscio D, Ferrucci R, Sartucci F, Priori A. Cerebellar Direct Current Stimulation (ctDCS) in the Treatment of Huntington's Disease: A Pilot Study and a Short Review of the Literature. Front Neurol 2020; 11:614717. [PMID: 33343504 PMCID: PMC7744723 DOI: 10.3389/fneur.2020.614717] [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: 10/06/2020] [Accepted: 11/11/2020] [Indexed: 11/13/2022] Open
Abstract
Introduction: In recent years, a growing body of literature has investigated the use of non-invasive brain stimulation (NIBS) techniques as a putative treatment in Huntington's Disease (HD). Our aim was to evaluate the effects of cerebellar transcranial Direct Current Simulation (ctDCS) on the motor outcome in patients affected by HD, encompassing at the same time the current knowledge about the effects of NIBS both on motor and non-motor dysfunctions in HD. Materials and Methods: Four patients (two females) were enrolled and underwent ctDCS (both anodal or sham, elapsed by at least 3 months: 2.0 mA, 20 min per day, 5 days a week). Clinical scores were assessed by using the Unified Huntington's Disease Rating Scale - part I (UHDRS-I), immediately before ctDCS (T0), at the end of the 5-days treatment (T1) and 4 weeks later (T2). Results: Anodal ctDCS improved motor scores compared to baseline (p = 0.0046), whereas sham stimulation left them unchanged (p = 0.33, Friedman test). In particular, following anodal ctDCS, UHDRS-I score significantly improved, especially regarding the subitem "dystonia," both at T1 and T2 compared to sham condition (p < 0.05; Wilcoxon matched-pairs signed test). Conclusions: ctDCS improved motor scores in HD, with effects lasting for about 4 weeks after tDCS completion. This is the first study discussing the putative role of cerebellar non-invasive simulation for the treatment of HD.
Collapse
Affiliation(s)
- Tommaso Bocci
- "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan & Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo, Milan, Italy
| | - Davide Baloscio
- Section of Neurophysiopathology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberta Ferrucci
- "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan & Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo, Milan, Italy
| | - Ferdinando Sartucci
- Section of Neurophysiopathology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alberto Priori
- "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan & Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo, Milan, Italy
| |
Collapse
|
9
|
Agüera E, Caballero-Villarraso J, Feijóo M, Escribano BM, Conde C, Bahamonde MC, Giraldo AI, Paz-Rojas E, Túnez I. Clinical and Neurochemical Effects of Transcranial Magnetic Stimulation (TMS) in Multiple Sclerosis: A Study Protocol for a Randomized Clinical Trial. Front Neurol 2020; 11:750. [PMID: 32849212 PMCID: PMC7431867 DOI: 10.3389/fneur.2020.00750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Transcranial Magnetic Stimulation (TMS) is a technique based on the principles of electromagnetic induction. It applies pulses of magnetic radiation that penetrate the brain tissue, and it is a non-invasive, painless, and practically innocuous procedure. Previous studies advocate the therapeutic capacity of TMS in several neurodegenerative and psychiatric processes, both in animal models and in human studies. Its uses in Parkinson's disease, Alzheimer's disease and in Huntington's chorea have shown improvement in the symptomatology and in the molecular profile, and even in the cellular density of the brain. Consequently, the extrapolation of these TMS results in the aforementioned neurodegenerative disease to other entities with etiopathogenic and clinical analogy would raise the relevance and feasibility of its use in multiple sclerosis (MS). The overall objective will be to demonstrate the effectiveness of the TMS in terms of safety and clinical improvement, as well as to observe the molecular changes in relation to the treatment. Methods and Design: Phase II clinical trial, unicentric, controlled, randomized, single blind. A total of 90 patients diagnosed with relapsing-remitting multiple sclerosis (RRMS) who meet all the inclusion criteria and do not present any of the exclusion criteria that are established and from which clinically evaluable results can be obtained. The patients included will be assigned under the 1:1:1 randomization formula, constituting three groups for the present study: 30 patients treated with natalizumab + white (placebo) + 30 patients treated with natalizumab + TMS (1 Hz) + 30 patients treated with natalizumab + TMS (5 Hz). Discussion: Results of this study will inform on the efficiency of the TMS for the treatment of MS. The expected results are that TMS is a useful therapeutic resource to improve clinical status (main parameters) and neurochemical profile (surrogate parameters); both types of parameters will be checked. Ethics and Dissemination: The study is approved by the Local Ethics Committee and registered in https://clinicaltrials.gov (NCT04062331). Dissemination will include submission to a peer-reviewed journal, patients, associations of sick people and family members, healthcare magazines and congress presentations. Trial Registration:ClinicalTrials.gov ID: NCT04062331 (registration date: 19th/ August/2019). Version Identifier: EMTr-EMRR, ver-3, 21/11/2017.
Collapse
Affiliation(s)
- Eduardo Agüera
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Unidad de Gestión Clínica de Neurología, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Javier Caballero-Villarraso
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departmento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Córdoba, Spain.,Unidad de Gestión Clínica de Análisis Clínicos, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Montserrat Feijóo
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departmento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Córdoba, Spain
| | - Begoña M Escribano
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
| | - Cristina Conde
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - María C Bahamonde
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Unidad de Gestión Clínica de Neurología, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Ana I Giraldo
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departmento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Córdoba, Spain
| | - Elier Paz-Rojas
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Canvax Biotech S.L., Córdoba, Spain
| | - Isaac Túnez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departmento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Córdoba, Spain
| |
Collapse
|
10
|
Wei Q, Wu G, Xing J, Mao D, Hutz RJ, Shi F. Roles of poly (ADP-ribose) polymerase 1 activation and cleavage in induction of multi-oocyte ovarian follicles in the mouse by 3-nitropropionic acid. Reprod Fertil Dev 2020; 31:1017-1032. [PMID: 30836053 DOI: 10.1071/rd18406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/07/2019] [Indexed: 01/08/2023] Open
Abstract
3-nitropropionic acid (3-NPA) is known to be a mitochondrial toxin produced by plants and fungi, which may produce DNA damage in cells. However, studies of its reproductive toxicology are lacking. We know that poly(ADP-ribose) polymerase (PARP) plays an important role in a large variety of physiological processes and is involved in DNA repair pathways. The present study was therefore aimed at exploring the involvement of PARP-1 activation and cleavage after 3-NPA stimulation in female mice. We observed an increased number of atretic follicles and multi-oocyte follicles (MOFs) after treatment with 3-NPA and serum concentrations of 17β-oestradiol and progesterone were significantly reduced. Our results provide evidence that PARP-1 cleavage and activational signals are involved in pathological ovarian processes stimulated by 3-NPA. In addition, total superoxide dismutase, glutathione peroxidase and catalase activities were significantly increased, whereas succinate dehydrogenase was decreased in a dose-dependent manner. Results from our in vitro study similarly indicated that 3-NPA inhibited the proliferation of mouse granulosa cells and increased apoptosis in a dose-dependent manner. In summary, 3-NPA induces granulosa cell apoptosis, follicle atresia and MOFs in the ovaries of female mice and causes oxidative stress so as to disrupt endogenous hormonal systems, possibly acting through PARP-1 signalling.
Collapse
Affiliation(s)
- Quanwei Wei
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Guoyun Wu
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jun Xing
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; and Department of Animal Husbandry and Veterinary Medicine, Jiangsu Polytechnic College of Agriculture and Forestry, Jurong 212400, China
| | - Dagan Mao
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Reinhold J Hutz
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Lapham Hall, Milwaukee, WI 53211-0413, USA
| | - Fangxiong Shi
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; and Corresponding author.
| |
Collapse
|
11
|
Calabrese EJ, Bhatia TN, Calabrese V, Dhawan G, Giordano J, Hanekamp YN, Kapoor R, Kozumbo WJ, Leak RK. Cytotoxicity models of Huntington’s disease and relevance of hormetic mechanisms: A critical assessment of experimental approaches and strategies. Pharmacol Res 2019; 150:104371. [DOI: 10.1016/j.phrs.2019.104371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022]
|
12
|
Lai H. Exposure to Static and Extremely-Low Frequency Electromagnetic Fields and Cellular Free Radicals. Electromagn Biol Med 2019; 38:231-248. [PMID: 31450976 DOI: 10.1080/15368378.2019.1656645] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This paper summarizes studies on changes in cellular free radical activities from exposure to static and extremely-low frequency (ELF) electromagnetic fields (EMF), particularly magnetic fields. Changes in free radical activities, including levels of cellular reactive oxygen (ROS)/nitrogen (RNS) species and endogenous antioxidant enzymes and compounds that maintain physiological free radical concentrations in cells, is one of the most consistent effects of EMF exposure. These changes have been reported to affect many physiological functions such as DNA damage; immune response; inflammatory response; cell proliferation and differentiation; wound healing; neural electrical activities; and behavior. An important consideration is the effects of EMF-induced changes in free radicals on cell proliferation and differentiation. These cellular processes could affect cancer development and proper growth and development in organisms. On the other hand, they could cause selective killing of cancer cells, for instance, via the generation of the highly cytotoxic hydroxyl free radical by the Fenton Reaction. This provides a possibility of using these electromagnetic fields as a non-invasive and low side-effect cancer therapy. Static- and ELF-EMF probably play important roles in the evolution of living organisms. They are cues used in many critical survival functions, such as foraging, migration, and reproduction. Living organisms can detect and respond immediately to low environmental levels of these fields. Free radical processes are involved in some of these mechanisms. At this time, there is no credible hypothesis or mechanism that can adequately explain all the observed effects of static- and ELF-EMF on free radical processes. We are actually at the impasse that there are more questions than answers.
Collapse
Affiliation(s)
- Henry Lai
- Department of Bioengineering, University of Washington , Seattle , WA , USA
| |
Collapse
|
13
|
RNA Aptamers Rescue Mitochondrial Dysfunction in a Yeast Model of Huntington's Disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:45-56. [PMID: 30195782 PMCID: PMC6023792 DOI: 10.1016/j.omtn.2018.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 03/27/2018] [Accepted: 04/25/2018] [Indexed: 01/27/2023]
Abstract
Huntington’s disease (HD) is associated with the misfolding and aggregation of mutant huntingtin harboring an elongated polyglutamine stretch at its N terminus. A distinguishing pathological hallmark of HD is mitochondrial dysfunction. Any strategy that can restore the integrity of the mitochondrial environment should have beneficial consequences for the disease. Specific RNA aptamers were selected that were able to inhibit aggregation of elongated polyglutamine stretch containing mutant huntingtin fragment (103Q-htt). They were successful in reducing the calcium overload, which leads to mitochondrial membrane depolarization in case of HD. In one case, the level of Ca2+ was restored to the level of cells not expressing 103Q-htt, suggesting complete recovery. The presence of aptamers was able to increase mitochondrial mass in cells expressing 103Q-htt, along with rescuing loss of mitochondrial genome. The oxidative damage to the proteome was prevented, which led to increased viability of cells, as monitored by flow cytometry. Thus, the presence of aptamers was able to inhibit aggregation of mutant huntingtin fragment and restore mitochondrial dysfunction in the HD cell model, confirming the advantage of the strategy in a disease-relevant parameter.
Collapse
|
14
|
Medina-Fernández FJ, Escribano BM, Padilla-Del-Campo C, Drucker-Colín R, Pascual-Leone Á, Túnez I. Transcranial magnetic stimulation as an antioxidant. Free Radic Res 2018; 52:381-389. [PMID: 29385851 DOI: 10.1080/10715762.2018.1434313] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the last decades, different transcranial magnetic stimulation protocols have been developed as a therapeutic tool against neurodegenerative and psychiatric diseases, although the biochemical, molecular and cellular mechanisms underlying these effects are not well known. Recent data show that those magnetic stimulation protocols showing beneficial effects could trigger an anti-oxidant action that would favour, at least partially, their therapeutic effect. We have aimed to review the molecular effects related to oxidative damage induced by this therapeutic strategy, as well as from them addressing a broader definition of the anti-oxidant concept.
Collapse
Affiliation(s)
- Francisco J Medina-Fernández
- a Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería , Universidad de Córdoba , Córdoba , Spain.,b Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC) , Córdoba , Spain
| | - Begoña M Escribano
- a Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería , Universidad de Córdoba , Córdoba , Spain.,c Departamento de Biología Celular, Fisiología e Inmunología , Universidad de Córdoba , Córdoba , Spain
| | | | - René Drucker-Colín
- e Departmento de Neuropatología Molecular , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM) , Ciudad de México , DF , México
| | - Álvaro Pascual-Leone
- f Division of Cognitive Neurology, Department of Neurology , Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston , MA , USA
| | - Isaac Túnez
- a Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería , Universidad de Córdoba , Córdoba , Spain.,b Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC) , Córdoba , Spain
| |
Collapse
|
15
|
Medina-Fernandez FJ, Escribano BM, Luque E, Caballero-Villarraso J, Gomez-Chaparro JL, Feijoo M, Garcia-Maceira FI, Pascual-Leone A, Drucker-Colin R, Tunez I. Comparative of transcranial magnetic stimulation and other treatments in experimental autoimmune encephalomyelitis. Brain Res Bull 2018; 137:140-145. [DOI: 10.1016/j.brainresbull.2017.11.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/19/2017] [Accepted: 11/28/2017] [Indexed: 11/16/2022]
|
16
|
Elgohary HM, Tantawy SA. Pulsed electromagnetic field with or without exercise therapy in the treatment of benign prostatic hyperplasia. J Phys Ther Sci 2017; 29:1305-1310. [PMID: 28878453 PMCID: PMC5574357 DOI: 10.1589/jpts.29.1305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/09/2017] [Indexed: 11/24/2022] Open
Abstract
[Purpose] To investigate the effect of pulsed electromagnetic field with or without
exercise therapy in the treatment of benign prostatic hyperplasia. [Subjects and Methods]
Sixty male patients aged 55–65 years with benign prostatic hyperplasia were invited to
participate in this study. Patients were randomly assigned to Group A (n=20; patients who
received pulsed electromagnetic field in addition to pelvic floor and aerobic exercises),
Group B (n=20; patients who received pulsed electromagnetic field), and Group C (n=20;
patients who received placebo electromagnetic field). The assessments included post-void
residual urine, urine flow rate, prostate specific antigen, white blood cells count, and
International Prostate Symptom Score were weighed, before and after a 4-week intervention.
[Results] There were significant differences in Group A and B in all parameters. Group C
showed non-significant differences in all measured variables except for International
Prostate Symptom Score. Among groups, all parameters showed highly significant differences
in favor of Group A. There were non-significant differences between Group A and B and
significant difference between Groups A and C and between Groups B and C. [Conclusion] The
present study demonstrated that electromagnetic field had a significant impact on the
treatment of benign prostatic hyperplasia. Accordingly, electromagnetic field can be
utilized alone or in combination with other physiotherapy modalities. Moreover, clinicians
should have the capacity to perceive the advantages accomplished using extra treatment
alternatives. Electromagnetic field is a safe, noninvasive method and can be used for the
treatment of benign prostatic hyperplasia.
Collapse
Affiliation(s)
- Hany M Elgohary
- Department of Physical Therapy for Surgery, Faculty of Physical Therapy, Cairo University, Egypt
| | - Sayed A Tantawy
- Physiotherapy Department, Center of Radiation, Oncology and Nuclear Medicine, Cairo University, Egypt.,Department of Physiotherapy, College of Medical & Health Sciences, Ahlia University, Kingdom of Bahrain
| |
Collapse
|
17
|
Medina-Fernandez FJ, Escribano BM, Agüera E, Aguilar-Luque M, Feijoo M, Luque E, Garcia-Maceira FI, Pascual-Leone A, Drucker-Colin R, Tunez I. Effects of transcranial magnetic stimulation on oxidative stress in experimental autoimmune encephalomyelitis. Free Radic Res 2017; 51:460-469. [DOI: 10.1080/10715762.2017.1324955] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
| | - Begoña M. Escribano
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain
- Faculty of Veterinary Medicine, Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
| | - Eduardo Agüera
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain
- Department of Neurology, Reina Sofia University Hospital, Cordoba, Spain
| | - Macarena Aguilar-Luque
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain
| | - Montserrat Feijoo
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cordoba, Cordoba, Spain
| | - Evelio Luque
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cordoba, Cordoba, Spain
- Histology Section, Faculty of Medicine, Department of Morphological Sciences, University of Cordoba, Cordoba, Spain
| | | | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - René Drucker-Colin
- Department of Molecular Neuropathology, Institute of Cell Physiology, National Autonomous University of Mexico (UNAM), Ciudad de Mexico, D.F, Mexico
| | - Isaac Tunez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Research in Biomedicine of Cordoba (IMIBIC), Cordoba, Spain
| |
Collapse
|
18
|
Transcranial magnetic stimulation modifies astrocytosis, cell density and lipopolysaccharide levels in experimental autoimmune encephalomyelitis. Life Sci 2017; 169:20-26. [DOI: 10.1016/j.lfs.2016.11.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/09/2023]
|
19
|
Mancic B, Stevanovic I, Ilic TV, Djuric A, Stojanovic I, Milanovic S, Ninkovic M. Transcranial theta-burst stimulation alters GLT-1 and vGluT1 expression in rat cerebellar cortex. Neurochem Int 2016; 100:120-127. [DOI: 10.1016/j.neuint.2016.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/31/2016] [Accepted: 09/09/2016] [Indexed: 12/13/2022]
|
20
|
Urnukhsaikhan E, Mishig-Ochir T, Kim SC, Park JK, Seo YK. Neuroprotective Effect of Low Frequency-Pulsed Electromagnetic Fields in Ischemic Stroke. Appl Biochem Biotechnol 2016; 181:1360-1371. [PMID: 27761795 DOI: 10.1007/s12010-016-2289-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 10/10/2016] [Indexed: 11/30/2022]
Abstract
Low frequency-pulsed electromagnetic fields (LF-PEMFs) affect many biological processes; however, the fundamental mechanisms responsible for these effects remain unclear. Our study aimed to investigate the effect of LF-PEMFs on neuroprotection after ischemic stroke. C57B6 mice were exposed to LF-PEMF (F = 60 Hz, Bm = 10 mT) after photothrombotic occlusion. We measured the BDNF/TrkB/Akt signaling pathway, pro-apoptotic and pro-survival protein and gene expressions, and the expression of inflammatory mediators and performed behavioral tests in both LF-PEMF-treated and untreated ischemic stroke mice. Our results showed that LF-PEMF treatment promotes activation of the BDNF/TrkB/Akt signaling pathway. Subsequently, pro-survival proteins were significantly increased, while pro-apoptotic proteins and inflammatory mediators were decreased in ischemic stroke mice after LF-PEMF treatment. The results demonstrated that LF-PEMF exposure has a neuroprotective effect after ischemic stroke in mice during the recovery process.
Collapse
Affiliation(s)
| | | | - Soo-Chan Kim
- Graduate School of Bio and Information Technology, Hankyong National University, Anseong-si, Kyonggi-do, South Korea
| | - Jung-Keug Park
- Department of Medical Biotechnology, Dongguk University, Seoul, Republic of Korea
| | - Young-Kwon Seo
- Department of Medical Biotechnology, Dongguk University, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Long-term exposure to ELF-MF ameliorates cognitive deficits and attenuates tau hyperphosphorylation in 3xTg AD mice. Neurotoxicology 2016; 53:290-300. [PMID: 26945731 DOI: 10.1016/j.neuro.2016.02.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/24/2016] [Accepted: 02/29/2016] [Indexed: 12/30/2022]
Abstract
Although numerous studies have reported the influence of extremely low frequency magnetic field (ELF-MF) exposure on human health, its effects on cognitive deficits in Alzheimer's disease (AD) have remained under debate. Moreover, the influence of ELF-MF on hyperphosphorylated tau, which is one of the most common pathological hallmarks of AD, has not been reported to date. Therefore, transgenic mice (3xTg) were used in the present study. 3xTg mice, which express an APP/PS1 mutation combined with a tau (P301L) mutation and that develop cognitive deficits at 6 months of age, were subjected to ELF-MF (50Hz, 500μT) exposure or sham exposure daily for 3 months. We discovered that ELF-MF exposure ameliorated cognitive deficits and increased synaptic proteins in 3xTg mice. The protective effects of ELF-MF exposure may have also been caused by the inhibition of apoptosis and/or decreased oxidative stress levels that were observed in the hippocampus tissues of treated mice. Furthermore, tau hyperphosphorylation was decreased in vivo because of ELF-MF exposure, and this decrease was induced by the inhibition of GSK3β and CDK5 activities and activation of PP2Ac. We are the first to report that exposure to ELF-MF can attenuate tau phosphorylation. These findings suggest that ELF-MF exposure could act as a valid therapeutic strategy for ameliorating cognitive deficits and attenuating tau hyperphosphorylation in AD.
Collapse
|
22
|
Guerriero F, Ricevuti G. Extremely low frequency electromagnetic fields stimulation modulates autoimmunity and immune responses: a possible immuno-modulatory therapeutic effect in neurodegenerative diseases. Neural Regen Res 2016; 11:1888-1895. [PMID: 28197174 PMCID: PMC5270416 DOI: 10.4103/1673-5374.195277] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Increasing evidence shows that extremely low frequency electromagnetic fields (ELF-EMFs) stimulation is able to exert a certain action on autoimmunity and immune cells. In the past, the efficacy of pulsed ELF-EMFs in alleviating the symptoms and the progression of multiple sclerosis has been supported through their action on neurotransmission and on the autoimmune mechanisms responsible for demyelination. Regarding the immune system, ELF-EMF exposure contributes to a general activation of macrophages, resulting in changes of autoimmunity and several immunological reactions, such as increased reactive oxygen species-formation, enhanced phagocytic activity and increased production of chemokines. Transcranial electromagnetic brain stimulation is a non-invasive novel technique used recently to treat different neurodegenerative disorders, in particular Alzheimer's disease. Despite its proven value, the mechanisms through which EMF brain-stimulation exerts its beneficial action on neuronal function remains unclear. Recent studies have shown that its beneficial effects may be due to a neuroprotective effect on oxidative cell damage. On the basis of in vitro and clinical studies on brain activity, modulation by ELF-EMFs could possibly counteract the aberrant pro-inflammatory responses present in neurodegenerative disorders reducing their severity and their onset. The objective of this review is to provide a systematic overview of the published literature on EMFs and outline the most promising effects of ELF-EMFs in developing treatments of neurodegenerative disorders. In this regard, we review data supporting the role of ELF-EMF in generating immune-modulatory responses, neuromodulation, and potential neuroprotective benefits. Nonetheless, we reckon that the underlying mechanisms of interaction between EMF and the immune system are still to be completely understood and need further studies at a molecular level.
Collapse
Affiliation(s)
- Fabio Guerriero
- Department of Internal Medicine and Medical Therapy, Section of Geriatrics, University of Pavia, Pavia, Italy; Azienda di Servizi alla Persona, Istituto di Cura Santa Margherita of Pavia, Pavia, Italy
| | - Giovanni Ricevuti
- Department of Internal Medicine and Medical Therapy, Section of Geriatrics, University of Pavia, Pavia, Italy; Azienda di Servizi alla Persona, Istituto di Cura Santa Margherita of Pavia, Pavia, Italy
| |
Collapse
|
23
|
Mitochondrial modulators in experimental Huntington’s disease: reversal of mitochondrial dysfunctions and cognitive deficits. Neurobiol Aging 2015; 36:2186-200. [DOI: 10.1016/j.neurobiolaging.2015.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 12/25/2014] [Accepted: 02/05/2015] [Indexed: 01/19/2023]
|
24
|
Masoudian N, Riazi GH, Afrasiabi A, Modaresi SMS, Dadras A, Rafiei S, Yazdankhah M, Lyaghi A, Jarah M, Ahmadian S, Seidkhani H. Variations of glutamate concentration within synaptic cleft in the presence of electromagnetic fields: an artificial neural networks study. Neurochem Res 2015; 40:629-42. [PMID: 25577979 DOI: 10.1007/s11064-014-1509-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/20/2014] [Accepted: 12/26/2014] [Indexed: 12/31/2022]
Abstract
Glutamate is an excitatory neurotransmitter that is released by the majority of central nervous system synapses and is involved in developmental processes, cognitive functions, learning and memory. Excessive elevated concentrations of Glu in synaptic cleft results in neural cell apoptosis which is called excitotoxicity causing neurodegenerative diseases. Hence, we investigated the possibility of extremely low frequency electromagnetic fields (ELF-EMF) as a risk factor which is able to change Glu concentration in synaptic clef. Synaptosomes as a model of nervous terminal were exposed to ELF-EMF for 15-55 min in flux intensity range from 0.1 to 2 mT and frequency range from 50 to 230 Hz. Finally, all raw data by INForm v4.02 software as an artificial neural network program was analyzed to predict the effect of whole mentioned range spectra. The results showed the tolerance of all effects between the ranges from -35 to +40 % compared to normal state when glutamatergic systems exposed to ELF-EMF. It indicates that glutamatergic system attempts to compensate environmental changes though release or reuptake in order to keep the system safe. Regarding to the wide range of ELF-EMF acquired in this study, the obtained outcomes have potential for developing treatments based on ELF-EMF for some neurological diseases; however, in vivo experiments on the cross linking responses between glutamatergic and cholinergic systems in the presence of ELF-EMF would be needed.
Collapse
Affiliation(s)
- Neda Masoudian
- Institute of Biochemistry and Biophysics (I.B.B.), University of Tehran, Tehran, Iran
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Reininghaus E, Lackner N. Relationship satisfaction and sexuality in Huntington's disease. HANDBOOK OF CLINICAL NEUROLOGY 2015; 130:325-34. [PMID: 26003252 DOI: 10.1016/b978-0-444-63247-0.00018-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Huntington's disease (HD) is a chronic disabling disease that inflicts a considerable burden on patients and their families for a variety of reasons. These reasons include cognitive impairment and motor dysfunction, personality changes, and knowledge of possible genetic transmission of the disease to their children. Thus, the decision to take a genetic test for individuals at risk for HD is often associated with family planning and relationship stress. However, for most individuals, a positive genetic test does not alter family planning with regard to their decision to have children. HD has also been associated with abnormal sexual behaviors, although only a few studies have explored sexuality and sexual dysfunction in HD. Up to 85% of men and 75% of women experience sexual problems, including hypoactive sexual disorder in some cases and increased sexual interest and paraphilia in others. Psychologic support should involve the communication of realistic expectations about the progression of the disorder and potential consequences on the children.
Collapse
Affiliation(s)
- Eva Reininghaus
- Department of Psychiatry, Medical University of Graz, Graz, Austria.
| | - Nina Lackner
- Department of Psychiatry, Medical University of Graz, Graz, Austria
| |
Collapse
|
26
|
Wilhelm EA, Bortolatto CF, Jesse CR, Luchese C. Ebselen protects against behavioral and biochemical toxicities induced by 3-nitropropionic acid in rats: correlations between motor coordination, reactive species levels, and succinate dehydrogenase activity. Biol Trace Elem Res 2014; 162:200-10. [PMID: 25277606 DOI: 10.1007/s12011-014-0137-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/18/2014] [Indexed: 01/01/2023]
Abstract
The protective effect of ebselen was investigated against 3-nitropropionic acid (3-NP)-induced behavioral and biochemical toxicities in rats. Ebselen (10 or 25 mg/kg, intragastrically) was administered to rats 30 min before 3-NP (20 mg/kg, intraperitoneally) once a day for a period of 4 days. Locomotor activity, motor coordination, and body weight gain were determined. The striatal content of reactive oxygen species (ROS), reduced glutathione (GSH), ascorbic acid (AA), and protein carbonyl as well as catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), and glutathione-S-transferase (GST) activities was determined 24 h after the last dose of 3-NP. Na(+)/ K(+)-ATPase, succinate dehydrogenase (SDH), and δ-aminolevulinic dehydratase (δ-ALA-D) activities were also determined. The results demonstrated that ebselen at a dose of 25 mg/kg, but not at 10 mg/kg, protected against (1) a decrease in locomotor activity, motor coordination impairment, and body weight loss; (2) striatal oxidative damage, which was characterized by an increase in ROS levels, protein carbonyl content, and GR activity, an inhibition of CAT and GPx activities, and a decrease in GSH levels; and (3) an inhibition of SDH and Na(+)/K(+)-ATPase activities, induced by 3-NP. GST activity and AA levels were not modified by ebselen or 3-NP. Ebselen was not effective against the inhibition of δ-ALA-D activity induced by 3-NP. The results revealed a significant correlation between SDH activity and ROS levels, and SDH activity and latency to fall (rotarod test). The present study highlighted the protective effect of ebselen against 3-NP-induced toxicity in rats.
Collapse
Affiliation(s)
- Ethel A Wilhelm
- Programa de Pós-graduação em Bioquímica e Bioprospecção (PPGBioq), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, CEP 96010-900, RS, Brazil,
| | | | | | | |
Collapse
|
27
|
Reale M, Kamal MA, Patruno A, Costantini E, D'Angelo C, Pesce M, Greig NH. Neuronal cellular responses to extremely low frequency electromagnetic field exposure: implications regarding oxidative stress and neurodegeneration. PLoS One 2014; 9:e104973. [PMID: 25127118 PMCID: PMC4134243 DOI: 10.1371/journal.pone.0104973] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/15/2014] [Indexed: 11/19/2022] Open
Abstract
Neurodegenerative diseases comprise both hereditary and sporadic conditions characterized by an identifying progressive nervous system dysfunction and distinctive neuopathophysiology. The majority are of non-familial etiology and hence environmental factors and lifestyle play key roles in their pathogenesis. The extensive use of and ever increasing worldwide demand for electricity has stimulated societal and scientific interest on the environmental exposure to low frequency electromagnetic fields (EMFs) on human health. Epidemiological studies suggest a positive association between 50/60-Hz power transmission fields and leukemia or lymphoma development. Consequent to the association between EMFs and induction of oxidative stress, concerns relating to development of neurodegenerative diseases, such as Alzheimer disease (AD), have been voiced as the brain consumes the greatest fraction of oxygen and is particularly vulnerable to oxidative stress. Exposure to extremely low frequency (ELF)-EMFs are reported to alter animal behavior and modulate biological variables, including gene expression, regulation of cell survival, promotion of cellular differentiation, and changes in cerebral blood flow in aged AD transgenic mice. Alterations in inflammatory responses have also been reported, but how these actions impact human health remains unknown. We hence evaluated the effects of an electromagnetic wave (magnetic field intensity 1mT; frequency, 50-Hz) on a well-characterized immortalized neuronal cell model, human SH-SY5Y cells. ELF-EMF exposure elevated the expession of NOS and O2−, which were countered by compensatory changes in antioxidant catylase (CAT) activity and enzymatic kinetic parameters related to CYP-450 and CAT activity. Actions of ELF-EMFs on cytokine gene expression were additionally evaluated and found rapidly modified. Confronted with co-exposure to H2O2-induced oxidative stress, ELF-EMF proved not as well counteracted and resulted in a decline in CAT activity and a rise in O2− levels. Together these studies support the further evaluation of ELF-EMF exposure in cellular and in vivo preclinical models to define mechanisms potentially impacted in humans.
Collapse
Affiliation(s)
- Marcella Reale
- Department of Experimental and Clinical Sciences, University “G. d'Annunzio, Chieti, Italy
- * E-mail: (MR); (NG)
| | - Mohammad A. Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Antonia Patruno
- Department of Medicine and Aging Science, University 'G. d'Annunzio' of Chieti-Pescara, Chieti, Italy
| | - Erica Costantini
- Department of Experimental and Clinical Sciences, University “G. d'Annunzio, Chieti, Italy
| | - Chiara D'Angelo
- Department of Experimental and Clinical Sciences, University “G. d'Annunzio, Chieti, Italy
| | - Miko Pesce
- Department of Medicine and Aging Science, University 'G. d'Annunzio' of Chieti-Pescara, Chieti, Italy
| | - Nigel H. Greig
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail: (MR); (NG)
| |
Collapse
|
28
|
Ribeiro M, Silva AC, Rodrigues J, Naia L, Rego AC. Oxidizing effects of exogenous stressors in Huntington's disease knock-in striatal cells--protective effect of cystamine and creatine. Toxicol Sci 2013; 136:487-99. [PMID: 24008831 DOI: 10.1093/toxsci/kft199] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Huntington's disease (HD) is a polyglutamine-expansion disease associated to degeneration of striatal and cortical neurons. Previously, we showed that oxidative stress occurs in HD knock-in striatal cells, but little is known regarding cell antioxidant response against exogenous stimuli. Therefore, in the present study we analyzed cellular antioxidant profile following hydrogen peroxide (H2O2) and staurosporine (STS) exposure and tested the protective effect of cystamine and creatine in striatal cells expressing mutant huntingtin with 111 glutamines (STHdh (Q111/Q111); mutant cells) versus wild-type cells (STHdh (Q7/Q7)). Mutant cells displayed increased mitochondrial reactive oxygen species (ROS) and decreased NADPH oxidase and xanthine oxidase (XO) activities, reflecting lower superoxide cytosolic generation, along with increased superoxide dismutases (SODs) and components of glutathione redox cycle. Exposure to H2O2 and STS enhanced ROS in mutant cells and largely increased XO activity; STS further boosted the generation of mitochondrial ROS and caspase-3 activity. Both stimuli slightly increased SOD1 activity, without affecting SOD2 activity, and decreased glutathione reductase with a consequent rise in oxidized glutathione or glutathione disulfide in mutant cells, whereas H2O2 only increased glutathione peroxidase activity. Additionally, creatine and cystamine increased mutant cells viability and prevented ROS formation in HD cells subjected to H2O2 and STS. These results indicate that elevation of the antioxidant systems accompanies mitochondrial-driven ROS generation in mutant striatal cells and that exposure to noxious stimuli induces a higher susceptibility to oxidative stress by increasing XO activity and lowering the antioxidant response. Furthermore, creatine and cystamine are efficient in preventing H2O2- and STS-evoked ROS formation in HD striatal cells.
Collapse
|
29
|
Shen LK, Huang HM, Yang PC, Huang YK, Wang PDY, Leung TK, Chen CJ, Chang WJ. A static magnetic field attenuates lipopolysaccharide-induced neuro-inflammatory response via IL-6-mediated pathway. Electromagn Biol Med 2013; 33:132-8. [PMID: 23781996 DOI: 10.3109/15368378.2013.794734] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
An effective method for controlling brain damage and neurodegeneration caused by inflammation remains elusive. Down-expression of the lipopolysaccharide (LPS)-induced inflammatory cytokines resulting in endotoxin tolerance is reported as an alternative anti-infection treatment. Nonetheless, because the dosage and action site are hard to control, endotoxin tolerance caused by low-dose LPS injection in brain tissue may induce side effects. The aim of this study was to test the hypothesis that static magnetic fields (SMF) stimulate endotoxin tolerance in brain tissue. In this study, survival rate and pathological changes in brain tissues of LPS-challenged mice were examined with and without SMF treatment. In addition, the effects of SMF exposure on growth rate and cytokine expression of LPS-challenged BV-2 microglia cells were monitored. Our results showed that SMF pre-exposure had positive effects on the survival rate and histological outcomes of LPS-treated mice. Furthermore, SMF exposure significantly decreased IL-6 expression in BV-2 cells (p < 0.05) by a phenomenon similar to endotoxin tolerance. We suggest that SMF has potential as an alternative simulation source for controlling LPS-induced excess neuro-inflammatory response.
Collapse
Affiliation(s)
- Li-Kuo Shen
- Department of Radiology, Shuang Ho Hospital, Taipei Medical University , New Taipei City , Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Bertolino G, Dutra Souza HC, de Araujo JE. Neuropathology and behavioral impairments in Wistar rats with a 6-OHDA lesion in the substantia nigra compacta and exposure to a static magnetic field. Electromagn Biol Med 2013; 32:527-35. [DOI: 10.3109/15368378.2012.751394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Colle D, Santos DB, Hartwig JM, Godoi M, Braga AL, Farina M. Succinobucol versus probucol: Higher efficiency of succinobucol in mitigating 3-NP-induced brain mitochondrial dysfunction and oxidative stress in vitro. Mitochondrion 2013; 13:125-33. [DOI: 10.1016/j.mito.2013.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 01/08/2013] [Accepted: 01/08/2013] [Indexed: 02/07/2023]
|
32
|
Tasset I, Pérez-Herrera A, Medina FJ, Arias-Carrión Ó, Drucker-Colín R, Túnez I. Extremely low-frequency electromagnetic fields activate the antioxidant pathway Nrf2 in a Huntington's disease-like rat model. Brain Stimul 2013; 6:84-6. [DOI: 10.1016/j.brs.2012.03.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 03/18/2012] [Accepted: 03/21/2012] [Indexed: 10/28/2022] Open
|
33
|
Medina FJ, Túnez I. Mechanisms and pathways underlying the therapeutic effect of transcranial magnetic stimulation. Rev Neurosci 2013; 24:507-25. [DOI: 10.1515/revneuro-2013-0024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/22/2013] [Indexed: 11/15/2022]
|
34
|
Ribeiro M, Rosenstock TR, Cunha-Oliveira T, Ferreira IL, Oliveira CR, Rego AC. Glutathione redox cycle dysregulation in Huntington's disease knock-in striatal cells. Free Radic Biol Med 2012; 53:1857-67. [PMID: 22982598 DOI: 10.1016/j.freeradbiomed.2012.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 09/04/2012] [Accepted: 09/06/2012] [Indexed: 01/06/2023]
Abstract
Huntington's disease (HD) is a CAG repeat disorder affecting the HD gene, which encodes for huntingtin (Htt) and is characterized by prominent cell death in the striatum. Oxidative stress was previously implicated in HD neurodegeneration, but the role of the major endogenous antioxidant system, the glutathione redox cycle, has been less studied following expression of full-length mutant Htt (FL-mHtt). Thus, in this work we analyzed the glutathione system in striatal cells derived from HD knock-in mice expressing mutant Htt versus wild-type cells. Mutant cells showed increased intracellular reactive oxygen species (ROS) and caspase-3 activity, which were significantly prevented following treatment with glutathione ethyl ester. Interestingly, mutant cells exhibited an increase in intracellular levels of both reduced and oxidized forms of glutathione, and enhanced activities of glutathione peroxidase (GPx) and glutathione reductase (GRed). Furthermore, glutathione-S-transferase (GST) and γ-glutamyl transpeptidase (γ-GT) activities were also increased in mutant cells. Nevertheless, glutamate-cysteine ligase (GCL) and glutathione synthetase (GS) activities and levels of GCL catalytic subunit were decreased in cells expressing FL-mHtt, highly suggesting decreased de novo synthesis of glutathione. Enhanced intracellular total glutathione, despite decreased synthesis, could be explained by decreased extracellular glutathione in mutant cells. This occurred concomitantly with decreased mRNA expression levels and activity of the multidrug resistance protein 1 (Mrp1), a transport protein that mediates cellular export of glutathione disulfide and glutathione conjugates. Additionally, inhibition of Mrp1 enhanced intracellular GSH in wild-type cells only. These data suggest that FL-mHtt affects the export of glutathione by decreasing the expression of Mrp1. Data further suggest that boosting of GSH-related antioxidant defense mechanisms induced by FL-mHtt is insufficient to counterbalance increased ROS formation and emergent apoptotic features in HD striatal cells.
Collapse
Affiliation(s)
- Márcio Ribeiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | |
Collapse
|
35
|
Consales C, Merla C, Marino C, Benassi B. Electromagnetic fields, oxidative stress, and neurodegeneration. Int J Cell Biol 2012; 2012:683897. [PMID: 22991514 PMCID: PMC3444040 DOI: 10.1155/2012/683897] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 06/19/2012] [Accepted: 06/19/2012] [Indexed: 12/21/2022] Open
Abstract
Electromagnetic fields (EMFs) originating both from both natural and manmade sources permeate our environment. As people are continuously exposed to EMFs in everyday life, it is a matter of great debate whether they can be harmful to human health. On the basis of two decades of epidemiological studies, an increased risk for childhood leukemia associated with Extremely Low Frequency fields has been consistently assessed, inducing the International Agency for Research on Cancer to insert them in the 2B section of carcinogens in 2001. EMFs interaction with biological systems may cause oxidative stress under certain circumstances. Since free radicals are essential for brain physiological processes and pathological degeneration, research focusing on the possible influence of the EMFs-driven oxidative stress is still in progress, especially in the light of recent studies suggesting that EMFs may contribute to the etiology of neurodegenerative disorders. This review synthesizes the emerging evidences about this topic, highlighting the wide data uncertainty that still characterizes the EMFs effect on oxidative stress modulation, as both pro-oxidant and neuroprotective effects have been documented. Care should be taken to avoid methodological limitations and to determine the patho-physiological relevance of any alteration found in EMFs-exposed biological system.
Collapse
Affiliation(s)
- Claudia Consales
- Unit of Radiation Biology and Human Health, ENEA-Casaccia, Rome 00123, Italy
| | | | | | - Barbara Benassi
- Unit of Radiation Biology and Human Health, ENEA-Casaccia, Rome 00123, Italy
| |
Collapse
|
36
|
Pilato F, Profice P, Ranieri F, Capone F, Di Iorio R, Florio L, Di Lazzaro V. Synaptic plasticity in neurodegenerative diseases evaluated and modulated by in vivo neurophysiological techniques. Mol Neurobiol 2012; 46:563-71. [PMID: 22821187 DOI: 10.1007/s12035-012-8302-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 07/09/2012] [Indexed: 12/12/2022]
Abstract
Several studies demonstrated in experimental models and in humans synaptic plasticity impairment in some neurodegenerative and neuropsychiatric diseases such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and schizophrenia. Recently new neurophysiological tools, such as repetitive transcranial magnetic stimulation and transcranial direct current stimulation, have been introduced in experimental and clinical settings for studying physiology of the brain and modulating cortical activity. These techniques use noninvasive transcranial electrical or magnetic stimulation to modulate neurons activity in the human brain. Cortical stimulation might enhance or inhibit the activity of cortico-subcortical networks, depending on stimulus frequency and intensity, current polarity, and other stimulation parameters such as the configuration of the induced electric field and stimulation protocols. On this basis, in the last two decades, these techniques have rapidly become valuable tools to investigate physiology of the human brain and have been applied to treat drug-resistant neurological and psychiatric diseases. Here we describe these techniques and discuss the mechanisms that may explain these effects.
Collapse
Affiliation(s)
- F Pilato
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
37
|
Vahabzadeh-Hagh AM, Muller PA, Gersner R, Zangen A, Rotenberg A. Translational neuromodulation: approximating human transcranial magnetic stimulation protocols in rats. Neuromodulation 2012; 15:296-305. [PMID: 22780329 PMCID: PMC5764706 DOI: 10.1111/j.1525-1403.2012.00482.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Transcranial magnetic stimulation (TMS) is a well-established clinical protocol with numerous potential therapeutic and diagnostic applications. Yet, much work remains in the elucidation of TMS mechanisms, optimization of protocols, and in development of novel therapeutic applications. As with many technologies, the key to these issues lies in the proper experimentation and translation of TMS methods to animal models, among which rat models have proven popular. A significant increase in the number of rat TMS publications has necessitated analysis of their relevance to human work. We therefore review the essential principles for the approximation of human TMS protocols in rats as well as specific methods that addressed these issues in published studies. MATERIALS AND METHODS We performed an English language literature search combined with our own experience and data. We address issues that we see as important in the translation of human TMS methods to rat models and provide a summary of key accomplishments in these areas. RESULTS An extensive literature review illustrated the growth of rodent TMS studies in recent years. Current advances in the translation of single, paired-pulse, and repetitive stimulation paradigms to rodent models are presented. The importance of TMS in the generation of data for preclinical trials is also highlighted. CONCLUSIONS Rat TMS has several limitations when considering parallels between animal and human stimulation. However, it has proven to be a useful tool in the field of translational brain stimulation and will likely continue to aid in the design and implementation of stimulation protocols for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Andrew M. Vahabzadeh-Hagh
- Department of Neurology, Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Paul A. Muller
- Department of Neurology, Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Roman Gersner
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Abraham Zangen
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander Rotenberg
- Department of Neurology, Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
38
|
Neuroprotective effects of extremely low-frequency electromagnetic fields on a Huntington's disease rat model: effects on neurotrophic factors and neuronal density. Neuroscience 2012; 209:54-63. [DOI: 10.1016/j.neuroscience.2012.02.034] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 02/09/2012] [Accepted: 02/17/2012] [Indexed: 01/30/2023]
|
39
|
Deshmukh RS, Chaudhary RK, Roy I. Effect of pesticides on the aggregation of mutant huntingtin protein. Mol Neurobiol 2012; 45:405-14. [PMID: 22415443 DOI: 10.1007/s12035-012-8252-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 02/28/2012] [Indexed: 01/08/2023]
Abstract
The classical reports on neurodegeneration concentrate on studying disruption of signalling cascades. Although it is now well recognized that misfolding and aggregation of specific proteins are associated with a majority of these diseases, their role in aggravating the symptoms is not so well understood. Huntington's disease (HD) is a neurodegenerative disorder that results from damage to complex II of mitochondria. In this work, we have studied the effect of mitochondrial complex I inhibitors, viz. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and rotenone, and complex II inhibitor, viz. 3-nitropropionic acid, on the aggregation of mutant huntingtin (mthtt) protein, whose misfolding and aggregation results in cellular abnormalities characteristic of HD. All three inhibitors were found to accelerate the aggregation of mthtt in vitro, although the amounts of aggregates formed were different in all cases. Thus, apart from their effect on mitochondrial viability, these neurotoxins are capable of interfering with the protein aggregation process and thus, hastening the onset of the disease.
Collapse
Affiliation(s)
- Ruhi S Deshmukh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | | | | |
Collapse
|
40
|
Bhateja DK, Dhull DK, Gill A, Sidhu A, Sharma S, Reddy BK, Padi SS. Peroxisome proliferator-activated receptor-α activation attenuates 3-nitropropionic acid induced behavioral and biochemical alterations in rats: Possible neuroprotective mechanisms. Eur J Pharmacol 2012; 674:33-43. [DOI: 10.1016/j.ejphar.2011.10.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 10/07/2011] [Accepted: 10/15/2011] [Indexed: 01/01/2023]
|
41
|
Xiong Y, Uys JD, Tew KD, Townsend DM. S-glutathionylation: from molecular mechanisms to health outcomes. Antioxid Redox Signal 2011; 15:233-70. [PMID: 21235352 PMCID: PMC3110090 DOI: 10.1089/ars.2010.3540] [Citation(s) in RCA: 244] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Redox homeostasis governs a number of critical cellular processes. In turn, imbalances in pathways that control oxidative and reductive conditions have been linked to a number of human disease pathologies, particularly those associated with aging. Reduced glutathione is the most prevalent biological thiol and plays a crucial role in maintaining a reduced intracellular environment. Exposure to reactive oxygen or nitrogen species is causatively linked to the disease pathologies associated with redox imbalance. In particular, reactive oxygen species can differentially oxidize certain cysteine residues in target proteins and the reversible process of S-glutathionylation may mitigate or mediate the damage. This post-translational modification adds a tripeptide and a net negative charge that can lead to distinct structural and functional changes in the target protein. Because it is reversible, S-glutathionylation has the potential to act as a biological switch and to be integral in a number of critical oxidative signaling events. The present review provides a comprehensive account of how the S-glutathionylation cycle influences protein structure/function and cellular regulatory events, and how these may impact on human diseases. By understanding the components of this cycle, there should be opportunities to intervene in stress- and aging-related pathologies, perhaps through prevention and diagnostic and therapeutic platforms.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Pharmaceutical Sciences, Medical University of South Carolina, Charleston, 29425, USA
| | | | | | | |
Collapse
|
42
|
Numakawa T, Matsumoto T, Numakawa Y, Richards M, Yamawaki S, Kunugi H. Protective Action of Neurotrophic Factors and Estrogen against Oxidative Stress-Mediated Neurodegeneration. J Toxicol 2011; 2011:405194. [PMID: 21776259 PMCID: PMC3135156 DOI: 10.1155/2011/405194] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 02/28/2011] [Accepted: 03/29/2011] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress is involved in the pathogenesis of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Low levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) are important for maintenance of neuronal function, though elevated levels lead to neuronal cell death. A complex series of events including excitotoxicity, Ca(2+) overload, and mitochondrial dysfunction contributes to oxidative stress-mediated neurodegeneration. As expected, many antioxidants like phytochemicals and vitamins are known to reduce oxidative toxicity. Additionally, growing evidence indicates that neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and estrogens significantly prevent neuronal damage caused by oxidative stress. Here, we review and discuss recent studies addressing the protective mechanisms of neurotrophic factors and estrogen within this system.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| | - Tomoya Matsumoto
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
- Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yumiko Numakawa
- Peptide-prima Co., Ltd., 1-25-81, Nuyamazu, Kumamoto 861-2102, Japan
| | - Misty Richards
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- The Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208, USA
| | - Shigeto Yamawaki
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
- Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| |
Collapse
|
43
|
Antioxidant-like effects and protective action of transcranial magnetic stimulation in depression caused by olfactory bulbectomy. Neurochem Res 2010; 35:1182-7. [PMID: 20428940 DOI: 10.1007/s11064-010-0172-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2010] [Indexed: 12/23/2022]
Abstract
We studied the effects of transcranial magnetic stimulation (TMS, 60 Hz and 0.7 mT for 4 h/day for 14 days) on oxidative and cell damage caused by olfactory bulbectomy (OBX) in Wistar rats. The levels of lipid peroxidation products and caspase-3 were enhanced by OBX, whereas it prompted a reduction in reduced glutathione (GSH) content and antioxidative enzymes activities. The treatment with TMS reverted towards normality the biomarkers indicative of oxidative stress and apoptosis. In conclusion, our data show that TMS induced a protection against cell and oxidative damage induced by OBX, as well as they support the hypothesis that oxidative stress may play an important role in depression.
Collapse
|
44
|
Túnez I, Tasset I, Pérez-De La Cruz V, Santamaría A. 3-Nitropropionic acid as a tool to study the mechanisms involved in Huntington's disease: past, present and future. Molecules 2010; 15:878-916. [PMID: 20335954 PMCID: PMC6263191 DOI: 10.3390/molecules15020878] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 01/12/2010] [Accepted: 02/01/2010] [Indexed: 11/17/2022] Open
Abstract
Huntington's disease (HD) is an inheritable autosomal-dominant disorder whose causal mechanisms remain unknown. Experimental models have begun to uncover these pathways, thus helping to understand the mechanisms implicated and allowing for the characterization of potential targets for new therapeutic strategies. 3-Nitropropionic acid is known to produce in animals behavioural, biochemical and morphologic changes similar to those occurring in HD. For this reason, this phenotypic model is gaining attention as a valuable tool to mimick this disorder and further developing new therapies. In this review, we will focus on the past and present research of this molecule, to finally bring a perspective on what will be next in this promising field of study.
Collapse
Affiliation(s)
- Isaac Túnez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Maimónides de Investigaciones Biomédicas de Córdoba, Universidad de Córdoba, Av. Menéndez Pidal s/n, 14004 Córdoba, Spain.
| | | | | | | |
Collapse
|
45
|
Belikova NA, Tyurina YY, Borisenko G, Tyurin V, Samhan Arias AK, Yanamala N, Furtmüller PG, Klein-Seetharaman J, Obinger C, Kagan VE. Heterolytic reduction of fatty acid hydroperoxides by cytochrome c/cardiolipin complexes: antioxidant function in mitochondria. J Neurochem 2009; 131:11288-11289. [PMID: 19627079 DOI: 10.1111/j.1471-4159.2009.06331.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cytochrome c (cyt c), a mitochondrial intermembrane electron shuttle between complexes III and IV, can, upon binding with an anionic phospholipid, cardiolipin (CL), act as a peroxidase that catalyzes cardiolipin oxidation. H(2)O(2) was considered as a source of oxidative equivalents for this reaction, which is essential for programmed cell death. Here we report that peroxidase cyt c/CL complexes can utilize free fatty acid hydroperoxides (FFA-OOH) at exceptionally high rates that are approximately 3 orders of magnitude higher than for H(2)O(2). Similarly, peroxidase activity of murine liver mitochondria was high with FFA-OOH. Using EPR spin trapping and LC-MS techniques, we have demonstrated that cyt c/CL complexes split FFA-OOH predominantly via a heterolytic mechanism, yielding hydroxy-fatty acids, whereas H(2)O(2) (and tert-butyl hydroperoxide, t-BuOOH) undergo homolytic cleavage. Computer simulations have revealed that Arg(38) and His(33) are important for the heterolytic mechanism at potential FFA-OOH binding sites of cyt c (but not for H(2)O(2) or t-BuOOH). Regulation of FFA-OOH metabolism may be an important function of cyt c that is associated with elimination of toxic FFA-OOH and synthesis of physiologically active hydroxy-fatty acids in mitochondria.
Collapse
Affiliation(s)
- Natalia A Belikova
- Center for Free Radical and Antioxidant Health and Department of Environmental and Occupational Health, University of Pittsburgh, 100 Technology Drive, Suite 350, Pittsburgh, Pennsylvania 15219-3130, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Improvements in limb kinetic apraxia by repetition of a newly designed facilitation exercise in a patient with corticobasal degeneration. Int J Rehabil Res 2009; 32:178-83. [PMID: 19238090 DOI: 10.1097/mrr.0b013e32831e4546] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Corticobasal degeneration is a progressive neurological disorder characterized by a combination of parkinsonism and cortical dysfunction such as limb kinetic apraxia, alien limb phenomenon, and dementia. To study the effect of repetitive facilitation exercise (RFE) in a patient with corticobasal degeneration, we used a newly designed facilitation exercise designed to elicit movements isolated from the synergy in hemiplegia. This exercise included movements of each isolated finger using stretch reflex and skin-muscle reflex and repetitive movements demanded in activities of daily living (ADL) and manipulating objects. To evaluate improvements in hand functions by RFE, 1-week RFE sessions for the hand were administered alternatively to the left or right hand. The number of finger taps by the hand increased during each 1-week RFE session for the hand, but did not increase during 1-week sessions without RFE. After 1 month of treatment, the patient's difficulties in ADL, including wearing clothes, manipulating objects and cooking, decreased. Our results suggest the importance of the repetition of facilitation exercises and movements in ADL for recovery in patients with degenerative neurogenic diseases.
Collapse
|
47
|
Silva-Adaya D, Pérez-De La Cruz V, Herrera-Mundo MN, Mendoza-Macedo K, Villeda-Hernández J, Binienda Z, Ali SF, Santamaría A. Excitotoxic damage, disrupted energy metabolism, and oxidative stress in the rat brain: antioxidant and neuroprotective effects of L-carnitine. J Neurochem 2008; 105:677-89. [PMID: 18194214 DOI: 10.1111/j.1471-4159.2007.05174.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Excitotoxicity and disrupted energy metabolism are major events leading to nerve cell death in neurodegenerative disorders. These cooperative pathways share one common aspect: triggering of oxidative stress by free radical formation. In this work, we evaluated the effects of the antioxidant and energy precursor, levocarnitine (L-CAR), on the oxidative damage and the behavioral, morphological, and neurochemical alterations produced in nerve tissue by the excitotoxin and free radical precursor, quinolinic acid (2,3-pyrindin dicarboxylic acid; QUIN), and the mitochondrial toxin, 3-nitropropionic acid (3-NP). Oxidative damage was assessed by the estimation of reactive oxygen species formation, lipid peroxidation, and mitochondrial dysfunction in synaptosomal fractions. Behavioral, morphological, and neurochemical alterations were evaluated as markers of neurotoxicity in animals systemically administered with L-CAR, chronically injected with 3-NP and/or intrastriatally infused with QUIN. At micromolar concentrations, L-CAR reduced the three markers of oxidative stress stimulated by both toxins alone or in combination. L-CAR also prevented the rotation behavior evoked by QUIN and the hypokinetic pattern induced by 3-NP in rats. Morphological alterations produced by both toxins (increased striatal glial fibrillary acidic protein-immunoreactivity for QUIN and enhanced neuronal damage in different brain regions for 3-NP) were reduced by L-CAR. In addition, L-CAR prevented the synergistic action of 3-NP and QUIN to increase motor asymmetry and depleted striatal GABA levels. Our results suggest that the protective properties of L-CAR in the neurotoxic models tested are mostly mediated by its characteristics as an antioxidant agent.
Collapse
Affiliation(s)
- Daniela Silva-Adaya
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, México, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Túnez I, Montilla P, del Carmen Muñoz M, Medina FJ, Drucker-Colín R. Effect of transcranial magnetic stimulation on oxidative stress induced by 3-nitropropionic acid in cortical synaptosomes. Neurosci Res 2006; 56:91-5. [PMID: 16837092 DOI: 10.1016/j.neures.2006.05.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2006] [Revised: 05/02/2006] [Accepted: 05/30/2006] [Indexed: 11/16/2022]
Abstract
This study evaluates the effect of transcranial magnetic stimulation (TMS; 60 Hz and 0.7 mT) treatment on 3-nitropropionic acid (20 mg/kg i.p./day for 4 days)-induced oxidative stress in cortical synaptosomes of Wistar rats. The oxidative derangement was confirmed by a high level of lipid peroxidation products and protein carbonyls, together with a decreased in reduced glutathione (GSH) content, catalase and GSH-peroxidase (GSH-Px) activities. Additionally, it was observed a reduction in succinate dehydrogenase (SDH) activity. All changes were partially prevented or reversed by administration of TMS. These results show that TMS reduces oxidative stress in cortical synaptosomes, and suggest that TMS may protect neuronal and maintain synaptic integrity.
Collapse
Affiliation(s)
- Isaac Túnez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Córdoba, Spain.
| | | | | | | | | |
Collapse
|
49
|
del Hoyo P, García-Redondo A, de Bustos F, Molina JA, Sayed Y, Alonso-Navarro H, Caballero L, Arenas J, Jiménez-Jiménez FJ. Oxidative Stress in Skin Fibroblasts Cultures of Patients with Huntington’s Disease. Neurochem Res 2006; 31:1103-9. [PMID: 16944322 DOI: 10.1007/s11064-006-9110-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2006] [Indexed: 10/24/2022]
Abstract
Oxidative stress and mitochondrial dysfunction should play a role in the neurodegeneration in Huntington's disease (HD). The most consistent finding is decreased activity of the mitochondrial complexes II/III and IV of the respiratory chain in the striatum. We assessed enzymatic activities of respiratory chain enzymes and other enzymes involved in oxidative processes in skin fibroblasts cultures of patients with HD. We studied respiratory chain enzyme activities, activities of total, Cu/Zn- and Mn-superoxide-dismutase, glutathione-peroxidase (GPx) and catalase, and coenzyme Q(10) (CoQ(10)) levels in skin fibroblasts cultures from 13 HD patients and 13 age- and sex-matched healthy controls. When compared with controls, HD patients showed significantly lower specific activities for catalase corrected by protein concentrations (P < 0.01). Oxidized, reduced and total CoQ(10) levels (both corrected by citrate synthase (CS) and protein concentrations), and activities of total, Cu/Zn- and Mn-superoxide-dismutase, and gluthatione-peroxidase, did not differ significantly between HD-patients and control groups. Values for enzyme activities in the HD group did not correlate with age at onset and of the disease and with the CAG triplet repeats. The primary finding of this study was the decreased activity of catalase in HD patients, suggesting a possible contribution of catalase, but not of other enzymes related with oxidative stress, to the pathogenesis of this disease.
Collapse
Affiliation(s)
- Pilar del Hoyo
- Departamento de Bioquímica-Investigación, Hospital Universitario Doce de Octubre, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|