1
|
de Souza VS, Medeiros LF, Stein DJ, de Oliveira CL, Medeiros HR, Dussan-Sarria JA, Caumo W, de Souza A, Torres ILS. Transcranial direct current stimulation is more effective than pregabalin in controlling nociceptive and anxiety-like behaviors in a rat fibromyalgia-like model. Scand J Pain 2024; 24:sjpain-2023-0038. [PMID: 38557595 DOI: 10.1515/sjpain-2023-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Despite the fact that fibromyalgia, a widespread disease of the musculoskeletal system, has no specific treatment, patients have shown improvement after pharmacological intervention. Pregabalin has demonstrated efficacy; however, its adverse effects may reduce treatment adherence. In this context, neuromodulatory techniques such as transcranial direct current stimulation (tDCS) may be employed as a complementary pain-relieving method. Consequently, the purpose of this study was to evaluate the effect of pregabalin and tDCS treatments on the behavioral and biomarker parameters of rats submitted to a fibromyalgia-like model. METHODS Forty adult male Wistar rats were divided into two groups: control and reserpine. Five days after the end of the administration of reserpine (1 mg/kg/3 days) to induce a fibromyalgia-like model, rats were randomly assigned to receive either vehicle or pregabalin (30 mg/kg) along with sham or active- tDCS treatments. The evaluated behavioral parameters included mechanical allodynia by von Frey test and anxiety-like behaviors by elevated plus-maze test (time spent in opened and closed arms, number of entries in opened and closed arms, protected head-dipping, unprotected head-dipping [NPHD], grooming, rearing, fecal boluses). The biomarker analysis (brain-derived neurotrophic factor [BDNF] and tumor necrosis factor-α [TNF-α]) was performed in brainstem and cerebral cortex and in serum. RESULTS tDCS reversed the reduction in the mechanical nociceptive threshold and the decrease in the serum BDNF levels induced by the model of fibromyalgia; however, there was no effect of pregabalin in the mechanical threshold. There were no effects of pregabalin or tDCS found in TNF-α levels. The pain model induced an increase in grooming time and a decrease in NPHD and rearing; while tDCS reversed the increase in grooming, pregabalin reversed the decrease in NPHD. CONCLUSIONS tDCS was more effective than pregabalin in controlling nociception and anxiety-like behavior in a rat model-like fibromyalgia. Considering the translational aspect, our findings suggest that tDCS could be a potential non-pharmacological treatment for fibromyalgia.
Collapse
Affiliation(s)
- Vanessa Silva de Souza
- Institute of Basic Health Sciences, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil
| | - Liciane Fernandes Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil
- Universidade La Salle, Canoas, RS, 92010-000, Brazil
- Post graduate program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
| | - Dirson João Stein
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil
- Post graduate Program in Medicine: Medical Science, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
| | - Camila Lino de Oliveira
- Institute of Basic Health Sciences, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil
| | - Helouise Richardt Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil
- Post graduate Program in Medicine: Medical Science, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
| | | | - Wolnei Caumo
- Post graduate Program in Medicine: Medical Science, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
| | - Andressa de Souza
- Institute of Basic Health Sciences, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil
| | - Iraci L S Torres
- Institute of Basic Health Sciences, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil
- Department of Pharmacology, Universidade Federal Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
| |
Collapse
|
2
|
Zin LEF, Vizuete AFK, Callai EMM, Catarina LS, Fróes F, Moreira AP, de Oliveira Marques C, Leal MB, Ponzoni D, Puricelli E, da Silva Torres IL, Gonçalves CA, Quevedo AS. Astroglial Alterations in the Hippocampus of Rats Submitted to a Single Trans-Cranial Direct Current Stimulation Trial. Neurochem Res 2023; 48:3447-3456. [PMID: 37464227 DOI: 10.1007/s11064-023-03990-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/14/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023]
Abstract
Evidence indicates that transcranial direct current stimulation (tDCS) provides therapeutic benefits in different situations, such as epilepsy, depression, inflammatory and neuropathic pain. Despite the increasing use of tDCS, its cellular and molecular basis remains unknown. Astrocytes display a close functional and structural relationship with neurons and have been identified as mediators of neuroprotection in tDCS. Considering the importance of hippocampal glutamatergic neurotransmission in nociceptive pathways, we decided to investigate short-term changes in the hippocampal astrocytes of rats subjected to tDCS, evaluating specific cellular markers (GFAP and S100B), as well as markers of astroglial activity; glutamate uptake, glutamine synthesis by glutamine synthetase (GS) and glutathione content. Data clearly show that a single session of tDCS increases the pain threshold elicited by mechanical and thermal stimuli, as evaluated by von Frey and hot plate tests, respectively. These changes involve inflammatory and astroglial neurochemical changes in the hippocampus, based on specific changes in cell markers, such as S100B and GS. Alterations in S100B were also observed in the cerebrospinal fluid of tDCS animals and, most importantly, specific functional changes (increased glutamate uptake and increased GS activity) were detected in hippocampal astrocytes. These findings contribute to a better understanding of tDCS as a therapeutic strategy for nervous disorders and reinforce the importance of astrocytes as therapeutic targets.
Collapse
Affiliation(s)
- Lisandra Eda Fusinato Zin
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
- Atitus Educação, Campus Santa Teresinha, Passo Fundo, Brazil
| | - Adriana Fernanda K Vizuete
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
| | | | | | - Fernanda Fróes
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
| | - Ana Paula Moreira
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
| | | | - Miriara B Leal
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
| | - Deise Ponzoni
- Dentistry Graduate Program, UFRGS, Porto Alegre, Brazil
| | | | | | - Carlos-Alberto Gonçalves
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil.
- Neuroscience Graduate Program, UFRGS, Porto Alegre, Brazil.
| | - Alexandre Silva Quevedo
- Biochemistry Graduate Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, 2600, 90035-003, Brazil
- Dentistry Graduate Program, UFRGS, Porto Alegre, Brazil
- Neuroscience Graduate Program, UFRGS, Porto Alegre, Brazil
| |
Collapse
|
3
|
Zancanaro M, Stein DJ, Lopes BC, de Souza A, Ströher Toledo R, de Souza AH, Oliveira SM, Visioli F, Sanches PRS, Fregni F, Caumo W, Torres ILS. Preemptive transcranial direct current stimulation induces analgesia, prevents chronic inflammation and fibrosis, and promotes tissue repair in a rat model of postoperative pain. Neurosci Lett 2023; 813:137407. [PMID: 37499743 DOI: 10.1016/j.neulet.2023.137407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
This study evaluated the effects of previous exposure to Transcranial Direct Current Stimulation (tDCS) on nociceptive, neuroinflammatory, and neurochemical parameters, in rats subjected to an incisional pain model. Forty adult male Wistar rats (60 days old; weighing ∼ 250 g) were divided into five groups: 1. control (C); 2. drugs (D); 3. surgery (S); 4. surgery + sham-tDCS (SsT) and 5. surgery + tDCS (ST). Bimodal tDCS (0.5 mA) was applied for 20 min/day/8 days before the incisional model. Mechanical allodynia (von Frey) was evaluated at different time points after surgery. Cytokines and BDNF levels were evaluated in the cerebral cortex, hippocampus, brainstem, and spinal cord. Histology and activity of myeloperoxidase (MPO) and N-acetyl-β-D-glucosaminidase (NAGase) were evaluated in the surgical lesion sites in the right hind paw. The results demonstrate that the surgery procedure increased BDNF and IL-6 levels in the spinal cord levels in the hippocampus, and decreased IL-1β and IL-6 levels in the cerebral cortex, IL-6 levels in the hippocampus, and IL-10 levels in the brainstem and hippocampus. In addition, preemptive tDCS was effective in controlling postoperative pain, increasing BDNF, IL-6, and IL-10 levels in the spinal cord and brainstem, increasing IL-1β in the spinal cord, and decreasing IL-6 levels in the cerebral cortex and hippocampus, IL-1β and IL-10 levels in the hippocampus. Preemptive tDCS also contributes to tissue repair, preventing chronic inflammation, and consequent fibrosis. Thus, these findings imply that preemptive methods for postoperative pain management should be considered an interesting pain management strategy, and may contribute to the development of clinical applications for tDCS in surgical situations.
Collapse
Affiliation(s)
- Mayra Zancanaro
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-003, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Dirson J Stein
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-003, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Bettega C Lopes
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-003, Brazil
| | - Andressa de Souza
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-003, Brazil
| | - Roberta Ströher Toledo
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-003, Brazil
| | - Alessandra H de Souza
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-003, Brazil
| | - Sara M Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Fernanda Visioli
- Departamento de Odontologia Conservadora, Faculdade de Odontologia, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | | | - Felipe Fregni
- Laboratory of Neuromodulation, Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital and Massachusetts General Hospital, Harvard University, Boston, United States
| | - Wolnei Caumo
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Iraci L S Torres
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-003, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
4
|
Ye Y, Yan X, Wang L, Xu J, Li T. Transcranial direct current stimulation attenuates chronic pain in knee osteoarthritis by modulating BDNF/TrkB signaling in the descending pain modulation system. Neurosci Lett 2023; 810:137320. [PMID: 37295640 DOI: 10.1016/j.neulet.2023.137320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/15/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
Knee osteoarthritis (KOA) is the most common cause of chronic pain, but its pain mechanisms are complex and may be closely related to the descending pain modulation system. Transcranial direct current stimulation (tDCS) is used for relieving pain, but its analgesic mechanisms are still being explored. The purpose of this study was to investigate the role of BDNF/TrkB signaling in chronic pain in KOA and to investigate whether this signaling is related to the analgesic effect of tDCS. Rats were injected with monosodium iodoacetate (MIA) into the left knee joint to establish a chronic pain model and then received 20 min of tDCS for 8 days. Rats were respectively administered the TrkB inhibitor ANA-12 after MIA modeling and exogenous BDNF after tDCS treatment. Behaviors testing was assessed by hot plate and von Frey hairs using the up-down method. In addition, the expression levels of BDNF and TrkB on the periaqueductal gray (PAG)-the rostral ventromedial medulla (RVM)-the spinal dorsal horn (SDH) axis were detected by Western blot and Immunohistochemistry staining. Behavioral results show that tDCS treatment and ANA-12 injection reversed MIA-induced allodynia while reducing BDNF and TrkB expression levels. Furthermore, injection of exogenous BDNF reversed the therapeutic effect of tDCS on pain. These results indicate that upregulation of the BDNF/TrkB signaling in the descending pain modulation system may play an important role in KOA-induced chronic pain in rats, and tDCS may reduce KOA-induced chronic pain by inhibiting the BDNF/TrkB signaling in the descending pain modulation system.
Collapse
Affiliation(s)
- Yinshuang Ye
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiao Yan
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Lin Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jiawei Xu
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Tieshan Li
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|
5
|
da Silva LS, Toledo RS, Stein DJ, de Castro JM, Caumo W, Torres ILS. Transcranial Direct Current Stimulation (tDCS) antinociceptive effect is not altered by isoflurane anesthesia in neuropathic pain rats. BRAZILIAN JOURNAL OF ANESTHESIOLOGY (ELSEVIER) 2023; 73:514-518. [PMID: 36924939 PMCID: PMC10362433 DOI: 10.1016/j.bjane.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Affiliation(s)
- Lisiane Santos da Silva
- Hospital de Clínicas de Porto Alegre, Laboratório de Farmacologia e Neuromodulação da Dor: Investigações Pré-clínicas, Porto Alegre, RS, Brazil; Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil
| | - Roberta Ströher Toledo
- Hospital de Clínicas de Porto Alegre, Laboratório de Farmacologia e Neuromodulação da Dor: Investigações Pré-clínicas, Porto Alegre, RS, Brazil
| | - Dirson João Stein
- Hospital de Clínicas de Porto Alegre, Laboratório de Farmacologia e Neuromodulação da Dor: Investigações Pré-clínicas, Porto Alegre, RS, Brazil; Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil; Hospital de Clínicas de Porto Alegre, Núcleo Translacional: Farmacologia da Dor e Neuromodulação, Porto Alegre, RS, Brazil
| | - Josimar Macedo de Castro
- Hospital de Clínicas de Porto Alegre, Laboratório de Farmacologia e Neuromodulação da Dor: Investigações Pré-clínicas, Porto Alegre, RS, Brazil; Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil; Hospital de Clínicas de Porto Alegre, Núcleo Translacional: Farmacologia da Dor e Neuromodulação, Porto Alegre, RS, Brazil
| | - Wolnei Caumo
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil; Hospital de Clínicas de Porto Alegre, Núcleo Translacional: Farmacologia da Dor e Neuromodulação, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Hospital de Clínicas de Porto Alegre, Laboratório de Farmacologia e Neuromodulação da Dor: Investigações Pré-clínicas, Porto Alegre, RS, Brazil; Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil; Hospital de Clínicas de Porto Alegre, Núcleo Translacional: Farmacologia da Dor e Neuromodulação, Porto Alegre, RS, Brazil.
| |
Collapse
|
6
|
Xiong HY, Cao YQ, Du SH, Yang QH, He SY, Wang XQ. Effects of High-Definition Transcranial Direct Current Stimulation Targeting the Anterior Cingulate Cortex on the Pain Thresholds: A Randomized Controlled Trial. PAIN MEDICINE 2023; 24:89-98. [PMID: 36066447 DOI: 10.1093/pm/pnac135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND The majority of existing clinical studies used active transcranial direct current stimulation (tDCS) over superficial areas of the pain neuromatrix to regulate pain, with conflicting results. Few studies have investigated the effect of tDCS on pain thresholds by focusing on targets in deep parts of the pain neuromatrix. METHODS This study applied a single session of high-definition tDCS (HD-tDCS) targeting the anterior cingulate cortex (ACC) and used a parallel and sham-controlled design to compare the antinociceptive effects in healthy individuals by assessing changes in pain thresholds. Sixty-six female individuals (mean age, 20.5 ± 2.4 years) were randomly allocated into the anodal, cathodal, or sham HD-tDCS groups. The primary outcome of the study was pain thresholds (pressure pain threshold, heat pain threshold, and cold pain threshold), which were evaluated before and after stimulation through the use of quantitative sensory tests. RESULTS Only cathodal HD-tDCS targeting the ACC significantly increased heat pain threshold (P < 0.05) and pressure pain threshold (P < 0.01) in healthy individuals compared with sham stimulation. Neither anodal nor cathodal HD-tDCS showed significant analgesic effects on cold pain threshold. Furthermore, no statistically significant difference was found in pain thresholds between anodal and sham HD-tDCS (P > 0.38). Independent of HD-tDCS protocols, the positive and negative affective schedule scores were decreased immediately after stimulation compared with baseline. CONCLUSIONS The present study has found that cathodal HD-tDCS targeting the ACC provided a strong antinociceptive effect (increase in pain threshold), demonstrating a positive biological effect of HD-tDCS.
Collapse
Affiliation(s)
- Huan-Yu Xiong
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yin-Quan Cao
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Shu-Hao Du
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Qi-Hao Yang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Si-Yi He
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xue-Qiang Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China.,Department of Rehabilitation Medicine, Shanghai Shangti Orthopaedic Hospital, Shanghai, China
| |
Collapse
|
7
|
Callai EMM, Zin LEF, Catarina LS, Ponzoni D, Gonçalves CAS, Vizuete AFK, Cougo MC, Boff J, Puricelli E, Fernandes EK, da Silva Torres IL, Quevedo AS. Evaluation of the immediate effects of a single transcranial direct current stimulation session on astrocyte activation, inflammatory response, and pain threshold in naïve rats. Behav Brain Res 2022; 428:113880. [PMID: 35390432 DOI: 10.1016/j.bbr.2022.113880] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 02/15/2022] [Accepted: 04/01/2022] [Indexed: 11/18/2022]
Abstract
Transcranial direct current stimulation (tDCS) has demonstrated clinical benefits such as analgesia, anti-inflammatory, and neuroprotective effects. However, the mechanisms of action of a single tDCS session are poorly characterized. The present study aimed to evaluate the effects of a single tDCS session on pain sensitivity, inflammatory parameters, and astrocyte activity in naive rats. In the first experiment, sixty-day-old male Wistar rats (n=95) were tested for mechanical pain threshold (von Frey test). Afterward, animals were submitted to a single bimodal tDCS (0.5mA, 20minutes) or sham-tDCS session. According to the group, animals were re-tested at different time intervals (30, 60, 120minutes, or 24hours) after the intervention, euthanized, and the cerebral cortex collected for biochemical analysis. A second experiment (n=16) was performed using a similar protocol to test the hypotheses that S100B levels in the cerebrospinal fluid (CSF) are altered by tDCS. Elisa assay quantified the levels of tumor necrosis factor-alfa (TNF-α), interleukin-10 (IL10), S100 calcium-binding protein B (S100B), and Glial fibrillary acidic protein (GFAP). Data were analyzed using ANOVA and independent t-test (P<0.05). Results showed that tDCS decreased pain sensitivity (30 and 60min), cerebral TNF-α and S100B levels (30min). CSF S100B levels increased 30minutes after intervention. There were no differences in IL10 and GFAP levels. TCDS showed analgesic, anti-inflammatory, and neuroprotective effects in naive animals. Therefore, this non-invasive and inexpensive therapy may potentially be a preemptive alternative to reduce pain, inflammation, and neurodegeneration in situations where patients will undergo medical procedures (e.g., surgery).
Collapse
Affiliation(s)
- Etiane Micheli Meyer Callai
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | | | - Luciana Santa Catarina
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | - Deise Ponzoni
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | | | | | - Milton Cristian Cougo
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | - Jamile Boff
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | - Edela Puricelli
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil
| | | | | | - Alexandre Silva Quevedo
- Postgraduate Program in Dentistry, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, RS, Brazil; Neuroscience Graduate Program, UFRGS, Porto Alegre, RS, Brazil; Pharmacology Graduate Program, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
8
|
Li X, Zhou W, Wang L, Ye Y, Li T. Transcranial Direct Current Stimulation Alleviates the Chronic Pain of Osteoarthritis by Modulating NMDA Receptors in Midbrain Periaqueductal Gray in Rats. J Pain Res 2022; 15:203-214. [PMID: 35115824 PMCID: PMC8801364 DOI: 10.2147/jpr.s333454] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/18/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose Osteoarthritis (OA) is the most common cause to lead to chronic pain. Transcranial direct current stimulation (tDCS) has been widely used to treat nerve disorders and chronic pain. The benefits of tDCS for chronic pain are apparent, but its analgesic mechanism is still unclear. This study observed the analgesic effects of tDCS on OA-induced chronic pain and the changes of NMDA receptor levels in PAG after tDCS treatment in rats to explore the analgesic mechanism of tDCS. Methods After establishing chronic pain by injecting monosodium iodoacetate (MIA) into the rat ankle joint, the rats received tDCS for 14 consecutive days (20 min/day). Before tDCS treatment, Ifenprodil (the selective antagonist of NMDAR2B) was given to rats in different ways: intracerebroventricular (i.c.v.) injection or intraperitoneal (i.p.) injection. The Von Frey and hot plate tests were applied to assess the pain-related behaviors at different time points. The expression level of NMDAR2B was evaluated in midbrain periaqueductal gray (PAG) by Western blot. In addition, NMDAR2B and c-Fos were observed by the Immunohistochemistry staining after tDCS treatment. Results The mechanical allodynia and thermal hyperalgesia were produced after MIA injection. However, tDCS treatment reverted the mechanical allodynia and thermal hyperalgesia. Moreover, tDCS treatment significantly increased the expression of NMDAR2B and the proportion of positive stained cells of NMDAR2B. Besides that, the tDCS treatment also decreased the proportion of positive stained cells of c-Fos in PAG. However, these changes did not occur in the rats given the Ifenprodil (i.c.v.). Conclusion These results indicate that tDCS may increase the expression of NMDA receptors in PAG and strengthen the NMDA receptors-mediated antinociception to alleviate OA-induced chronic pain in rats.
Collapse
Affiliation(s)
- Xinhe Li
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Wenwen Zhou
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Lin Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Yinshuang Ye
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Tieshan Li
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Correspondence: Tieshan Li, Email
| |
Collapse
|
9
|
Santos DS, Stein DJ, Medeiros HR, Dos Santos Pereira F, de Macedo IC, Fregni F, Caumo W, Torres ILS. Transcranial direct current stimulation alters anxious-like behavior and neural parameters in rats with chronic pain exposed to alcohol. J Psychiatr Res 2021; 144:369-377. [PMID: 34735841 DOI: 10.1016/j.jpsychires.2021.10.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/04/2021] [Accepted: 10/25/2021] [Indexed: 11/27/2022]
Abstract
The aim of this study was to evaluate the effects of transcranial direct current stimulation (tDCS) on anxiety-like behavior and neural parameters in rats with chronic pain exposed to alcohol. Thirty-six adult male Wistar rats were randomly assigned to control (CT), neuropathic pain (NP), NPtDCS, NP + alcohol (NPAL), or NPALtDCS groups, subjected to sciatic nerve chronic constriction injury (CCI) and exposed to alcohol (20% v/v solution, 4 g/kg) or vehicle by gavage for 15 days. Afterward, rats were treated using bimodal tDCS (0.5 mA/20 min/8 days) and tested in the open field. Rats were killed 24 h after the last behavioral assessment, and brain and spinal cord tissue samples were collected and processed for NPY immunohistochemistry, expression of Il1a and Il1b in the spinal cord, cerebellum, and hippocampus, and levels of IL-1α and IL-1β in the same brain structures and the striatum. tDCS reverted the anxiety-like behavior induced by CCI and alcohol, and the increased expression of Il1a in the spinal cord induced by alcohol, which increased the expression of Il1b in the cerebellum. In addition, tDCS modulated the hypothalamic NPY-immunoreactivity, increased the levels of IL-1α in the hippocampus (like NP and AL), and increased the expression of Il1b in the spinal cord (like AL). Thus, this study shows that tDCS changes NP and alcohol-induced anxiety-like behavior, possibly through its central modulatory effect of NPY and spinal cord expression of Il1a and Il1b, being considered a treatment option for alcohol and NP-induced anxiety symptoms.
Collapse
Affiliation(s)
- Daniela Silva Santos
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Dirson João Stein
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Helouise Richardt Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Dos Santos Pereira
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil
| | - Isabel Cristina de Macedo
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil; Universidade Federal do Pampa, Uruguaiana, RS, Brazil
| | - Felipe Fregni
- Laboratory of Neuromodulation, Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital and Massachusetts General Hospital, Harvard University, Boston, MA, USA
| | - Wolnei Caumo
- Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
10
|
Suchting R, Teixeira AL, Ahn B, Colpo GD, Park J, Ahn H. Changes in Brain-derived Neurotrophic Factor From Active and Sham Transcranial Direct Current Stimulation in Older Adults With Knee Osteoarthritis. Clin J Pain 2021; 37:898-903. [PMID: 34757341 PMCID: PMC8589111 DOI: 10.1097/ajp.0000000000000987] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/01/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Previous work has shown effects of transcranial direct current stimulation (tDCS) on clinical pain measures, qualitative sensory testing measures, and peripheral inflammation. The present report extends this research to investigate the effect of tDCS on brain-derived neurotrophic factor (BDNF) levels. MATERIALS AND METHODS This secondary analysis examined a sample of 40 older adults (50 to 70 y old) with symptomatic knee osteoarthritis randomly assigned in a 1:1 fashion to active (n=20) or sham (n=20) tDCS for 20 minutes on 5 consecutive days. BDNF was measured before the first session and after the final treatment session. Generalized linear modeling evaluated BDNF plasma levels as a function of tDCS group, adjusted for baseline. Bayesian statistical inference was used to quantify the probability that effects of the treatment exist. RESULTS Generalized linear modeling indicated a 90.4% posterior probability that the sham condition had 49.9% higher BDNF at the end of treatment, controlling for baseline. Follow-up analyses within the active TDCS group supported an association between change in BDNF and change in clinical pain, and exploratory analyses found an effect of tDCS on irisin. DISCUSSION Results indicated that tDCS could be a potential nonpharmacological treatment to decrease BDNF levels, which may in turn decrease pain. This study adds to a growing literature suggesting that tDCS affects cortical excitability, and consequentially, the neural circuits implicated in pain modulation. In addition to a direct connection to analgesia, BDNF changes may reflect tDCS-induced changes in different cortical areas and/or neural circuits.
Collapse
Affiliation(s)
- Robert Suchting
- UTHealth McGovern Medical School, Faillace Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Antonio L. Teixeira
- UTHealth McGovern Medical School, Faillace Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Brian Ahn
- UTHealth McGovern Medical School, Faillace Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Gabriela D. Colpo
- UTHealth McGovern Medical School, Faillace Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Juyoung Park
- College of Social Work & Criminal Justice, Phyllis and Harvey Sandler School of Social Work, Florida Atlantic University, Boca Raton, FL, USA
| | - Hyochol Ahn
- College of Nursing, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
11
|
Caumo W, Alves RL, Vicuña P, Alves CFDS, Ramalho L, Sanches PRS, Silva DP, Torres ILDS, Fregni F. Impact of bifrontal home-based transcranial direct current stimulation in pain catastrophizing and disability due to pain in fibromyalgia: a randomized, double-blind sham-controlled study. THE JOURNAL OF PAIN 2021; 23:641-656. [PMID: 34785366 DOI: 10.1016/j.jpain.2021.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 09/05/2021] [Accepted: 11/08/2021] [Indexed: 11/15/2022]
Abstract
This randomized, double-blind trial tested the hypothesis that twenty sessions of home-based anodal(a)-transcranial direct current stimulation (tDCS) (2mA for 20 min) bifrontal, with anodal on the left dorsolateral prefrontal cortex (l-DLPFC) would be better than sham-(s)-tDCS to reduce scores on Pain Catastrophizing Scale (PCS) and disability-related to pain (DRP) assessed by the Profile of Chronic Pain: Screen (PCP:S) (primary outcomes). Secondary outcomes were depressive symptoms, sleep quality, heat pain threshold (HPT), heat pain tolerance (HPTo), and serum brain-derived-neurotrophic-factor (BDNF). Forty-eight women with fibromyalgia, 30-65 years-old were randomized into 2:1 groups [a-tDCS (n=32) or s-tDCS (n=16)]. Post hoc analysis revealed that a-tDCS reduced the PCS total scores by 51.38% compared to 26.96% in s-tDCS, and a-tDCS reduced PCP:S total scores by 31.43% compared to 19.15% in s-tDCS. The a-tDCS improved depressive symptoms, sleep quality and increased the HPTo. The delta-value in the serum BDNF (mean post treatment end minus pre-treatment) was conversely correlated with the a-tDCS effect in pain catastrophizing. In contrast, the a-tDCS impact on reducing the DRP at the treatment end was positively associated with a reduction in the serum BDNF and improvement of depressive symptoms, sleep quality and pain catastrophizing symptoms. PERSPECTIVE: Home-based bifrontal tDCS with a-tDCS on the l-DLPFC are associated with a moderate effect size (ES) in the following outcomes: (i) Decreased rumination and magnification of pain catastrophizing. (ii) Improved the disability for daily activities due to fibromyalgia symptoms. Overall, these findings support the feasibility of self-applied home-based tDCS on DLPFC to improve fibromyalgia symptoms.
Collapse
Affiliation(s)
- Wolnei Caumo
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Laboratory of Pain and Neuromodulation at Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil; Laboratory of Neuromodulation and Center for Clinical Research Learning, Physics and Rehabilitation Department, Spaulding Rehabilitation Hospital, Boston, MA, USA; Pain and Palliative Care Service at HCPA, Brazil; Department of Surgery, School of Medicine, UFRGS, Brazil.
| | - Rael Lopes Alves
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Laboratory of Pain and Neuromodulation at Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.
| | - Paul Vicuña
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Laboratory of Pain and Neuromodulation at Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.
| | - Camila Fernanda da Silveira Alves
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Laboratory of Pain and Neuromodulation at Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.
| | - Leticia Ramalho
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Laboratory of Pain and Neuromodulation at Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.
| | | | | | | | - Felipe Fregni
- Laboratory of Neuromodulation and Center for Clinical Research Learning, Physics and Rehabilitation Department, Spaulding Rehabilitation Hospital, Boston, MA, USA; School of Medicine, UFRGS; Laboratorio de Farmacologia da Dor e Neuromodulação: Investigacoes Pre-clinicas, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.
| |
Collapse
|
12
|
Sherwood MS, McIntire L, Madaris AT, Kim K, Ranganath C, McKinley RA. Intensity-Dependent Changes in Quantified Resting Cerebral Perfusion With Multiple Sessions of Transcranial DC Stimulation. Front Hum Neurosci 2021; 15:679977. [PMID: 34456695 PMCID: PMC8397582 DOI: 10.3389/fnhum.2021.679977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/15/2021] [Indexed: 11/13/2022] Open
Abstract
Transcranial direct current stimulation (tDCS) to the left prefrontal cortex has been shown to produce broad behavioral effects including enhanced learning and vigilance. Still, the neural mechanisms underlying such effects are not fully understood. Furthermore, the neural underpinnings of repeated stimulation remain understudied. In this work, we evaluated the effects of the repetition and intensity of tDCS on cerebral perfusion [cerebral blood flow (CBF)]. A cohort of 47 subjects was randomly assigned to one of the three groups. tDCS of 1- or 2-mA was applied to the left prefrontal cortex on three consecutive days, and resting CBF was quantified before and after stimulation using the arterial spin labeling MRI and then compared with a group that received sham stimulation. A widespread decreased CBF was found in a group receiving sham stimulation across the three post-stimulation measures when compared with baseline. In contrast, only slight decreases were observed in the group receiving 2-mA stimulation in the second and third post-stimulation measurements, but more prominent increased CBF was observed across several brain regions including the locus coeruleus (LC). The LC is an integral region in the production of norepinephrine and the noradrenergic system, and an increased norepinephrine/noradrenergic activity could explain the various behavioral findings from the anodal prefrontal tDCS. A decreased CBF was observed in the 1-mA group across the first two post-stimulation measurements, similar to the sham group. This decreased CBF was apparent in only a few small clusters in the third post-stimulation scan but was accompanied by an increased CBF, indicating that the neural effects of stimulation may persist for at least 24 h and that the repeated stimulation may produce cumulative effects.
Collapse
Affiliation(s)
| | | | - Aaron T. Madaris
- Infoscitex, Inc., Beavercreek, OH, United States
- Department of Biomedical, Industrial and Human Factors Engineering, Wright State University, Dayton, OH, United States
| | - Kamin Kim
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Charan Ranganath
- Department of Psychology, University of California, Davis, Davis, CA, United States
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - R. Andy McKinley
- Air Force Research Laboratory, Wright-Patterson AFB, Dayton, OH, United States
| |
Collapse
|
13
|
Santos DS, Medeiros LF, Stein DJ, De Macedo IC, Da Silva Rios DE, De Oliveira C, Toledo RS, Fregni F, Caumo W, Torres ILS. Bimodal transcranial direct current stimulation reduces alcohol consumption and induces long-term neurochemical changes in rats with neuropathic pain. Neurosci Lett 2021; 759:136014. [PMID: 34111512 DOI: 10.1016/j.neulet.2021.136014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/17/2021] [Accepted: 06/03/2021] [Indexed: 10/21/2022]
Abstract
This study aimed to evaluate the effects of repeated bimodal transcranial direct current stimulation (tDCS) on alcohol consumption and immunohistological and neurochemical parameters in nerve-injured rats. Forty-eight adult male Wistar rats were distributed into six groups: control, neuropathic pain (NP) + sham-tDCS, NP + alcohol + sham-tDCS, alcohol + sham-tDCS, alcohol + tDCS, and NP + alcohol + tDCS. NP is induced by chronic sciatic nerve constriction (CCI). The rats were exposed to a 10% alcohol solution by voluntary consumption for 14 days. From the 16th day after surgery, bimodal tDCS was applied for 20 min/day for 8 days. Brain structures were collected to evaluate the number of neuropeptide Y (NPY)-positive neurons, neurites, and argyrophilic grains by immunohistochemistry, and brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), interleukin (IL)-6, and IL-10 by ELISA. Nerve-injured rats showed a progressive increase in alcohol consumption compared to the non-injured rats. In addition, there was a reduction in voluntary alcohol consumption over time induced by tDCS. Alcohol exposure, chronic pain, and tDCS treatment modulated the central NPY immunoreactivity. tDCS increased the cerebellar levels of IL-6 and IL-10, and CCI and/or tDCS reduced striatal BDNF levels. The current data suggest that tDCS could be a promising non-pharmacological adjuvant to treat patients with chronic pain who use alcohol to relieve their symptoms.
Collapse
Affiliation(s)
- Daniela Silva Santos
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Liciane Fernandes Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Postgraduate Program in Health and Human Development, Universidade La Salle, Canoas, RS, Brazil
| | - Dirson João Stein
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Isabel Cristina De Macedo
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Diego Evandro Da Silva Rios
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carla De Oliveira
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Roberta Ströher Toledo
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Postgraduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Felipe Fregni
- Laboratoryof Neuromodulation, Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital and Massachusetts General Hospital, Harvard University, Boston, MA, USA
| | - Wolnei Caumo
- Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Investigations - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Postgraduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratoryof Neuromodulation, Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital and Massachusetts General Hospital, Harvard University, Boston, MA, USA.
| |
Collapse
|
14
|
Soldatelli MD, Siepmann T, Illigens BMW, Souza dos Santos V, Lucena da S Torres I, Fregni F, Caumo W. Mapping of predictors of the disengagement of the descending inhibitory pain modulation system in fibromyalgia: an exploratory study. Br J Pain 2021; 15:221-233. [PMID: 34055343 PMCID: PMC8138619 DOI: 10.1177/2049463720920760] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The main symptoms of fibromyalgia comprise diffuse pain, disability, depressive symptoms, catastrophizing, sleep disruption and fatigue, associated with dysfunction of the descending pain-modulating system (DPMS). OBJECTIVES We aimed to identify patterns of main symptoms of fibromyalgia and neuroplasticity biomarkers (i.e. brain-derived neurotrophic factor (BDNF) and S100B protein) in non-responders to the conditioned pain modulation task (CPM-task) induced by immersion of hand in cold water (0-1°C). Furthermore, we evaluated if these patterns predict responsiveness to CPM-task. METHODS This cross-sectional study included 117 women with fibromyalgia ((n = 60) non-responders and (n = 57) responders), with age ranging from 30 to 65 years old. We analysed changes in numerical pain scale (NPS-10) during the CPM-task using a standardized protocol. RESULTS A hierarchical multivariate logistic regression analysis was used to construct a propensity score-adjusted index to identify non-responders compared to responders to CPM-task. The following variables were retained in the models: analgesic use four or more times per week, heat pain threshold (HPT), poor sleep quality, pain catastrophizing, serum levels of BDNF, number of psychiatric diagnoses and the impact of symptoms of fibromyalgia on quality of life. Receiver operator characteristics (ROC) analysis showed non-responders can be discriminated from responders by a composite index of more frequent symptoms of fibromyalgia and neuroplasticity markers (area under the curve (AUC) = 0.83, sensitivity = 100% and specificity = 98%). CONCLUSION Patterns of fibromyalgia symptoms and neuroplasticity markers may be helpful to predict responsiveness to the CPM-task which might help personalize treatment and thereby contribute to the care of patients with fibromyalgia.
Collapse
Affiliation(s)
- Matheus Dorigatti Soldatelli
- Graduate Program in Medical Science,
School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre,
Brazil
- Center for Clinical Research and
Management Education, Division of Health Care Sciences, Dresden International
University, Dresden, Germany
- Laboratory of Pain and Neuromodulation,
School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre,
Brazil
| | - Timo Siepmann
- Center for Clinical Research and
Management Education, Division of Health Care Sciences, Dresden International
University, Dresden, Germany
- Department of Neurology, University
Hospital Carl Gustav Carus Technische Universitat, Dresden, Germany
| | - Ben Min-Woo Illigens
- Center for Clinical Research and
Management Education, Division of Health Care Sciences, Dresden International
University, Dresden, Germany
- Department of Neurology, Beth Israel
Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vinicius Souza dos Santos
- Laboratory of Pain and Neuromodulation,
School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre,
Brazil
| | - Iraci Lucena da S Torres
- Graduate Program in Medical Science,
School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre,
Brazil
- Pain and Palliative Care Service at
Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Felipe Fregni
- Department of Neurology, Beth Israel
Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Wolnei Caumo
- Graduate Program in Medical Science,
School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre,
Brazil
- Laboratory of Pain and Neuromodulation,
School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre,
Brazil
- Pain and Palliative Care Service at
Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Surgery Department, School of Medicine,
Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
15
|
Lopes BC, Medeiros LF, Stein DJ, Cioato SG, de Souza VS, Medeiros HR, Sanches PRS, Fregni F, Caumo W, Torres ILS. tDCS and exercise improve anxiety-like behavior and locomotion in chronic pain rats via modulation of neurotrophins and inflammatory mediators. Behav Brain Res 2021; 404:113173. [PMID: 33577881 DOI: 10.1016/j.bbr.2021.113173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/16/2022]
Abstract
Anxiety disorders cause distress and are commonly found to be comorbid with chronic pain. Both are difficult-to-treat conditions for which alternative treatment options are being pursued. This study aimed to evaluate the effects of transcranial direct current stimulation (tDCS), treadmill exercise, or both, on anxiety-like behavior and associated growth factors and inflammatory markers in the hippocampus and sciatic nerve of rats with neuropathic pain. Male Wistar rats (n = 216) were subjected to sham-surgery or sciatic nerve constriction for pain induction. Fourteen days following neuropathic pain establishment, either bimodal tDCS, treadmill exercise, or a combination of both was used for 20 min a day for 8 consecutive days. The elevated plus-maze test was used to assess anxiety-like behavior and locomotor activity during the early (24 h) or late (7 days) phase after the end of treatment. BDNF, TNF-ɑ, and IL-10 levels in the hippocampus, and BDNF, NGF, and IL-10 levels in the sciatic nerve were assessed 48 h or 7 days after the end of treatment. Rats from the pain groups developed an anxiety-like state. Both tDCS and treadmill exercise provided ethological and neurochemical alterations induced by pain in the early and/or late phase, and a modest synergic effect between tDCS and exercise was observed. These results indicate that non-invasive neuromodulatory approaches can attenuate both anxiety-like status and locomotor activity and alter the biochemical profile in the hippocampus and sciatic nerve of rats with neuropathic pain and that combined interventions may be considered as a treatment option.
Collapse
Affiliation(s)
- Bettega Costa Lopes
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, 90035-007 Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), 90050-170 Porto Alegre, Brazil
| | - Liciane Fernandes Medeiros
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, 90035-007 Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal Rio Grande do Sul, 90050-170 Porto Alegre, Brazil; Programa de Pós-Graduação em Saúde e Desenvolvimento Humano, Universidade La Salle, 92010-000 Canoas, Brazil.
| | - Dirson João Stein
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, 90035-007 Porto Alegre, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, Brazil
| | - Stefania Giotti Cioato
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, 90035-007 Porto Alegre, Brazil
| | - Vanessa Silva de Souza
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, 90035-007 Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal Rio Grande do Sul, 90050-170 Porto Alegre, Brazil
| | - Helouise Richardt Medeiros
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, 90035-007 Porto Alegre, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, Brazil
| | - Paulo Roberto Stefani Sanches
- Laboratório de Engenharia Biomédica, Grupo de Pesquisa e Pós-Graduação, Hospital de Clínicas de Porto Alegre, 90035-003 Porto Alegre, Brazil
| | - Felipe Fregni
- Laboratory of Neuromodulation, Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital & Massachusetts General Hospital. Harvard Medical School and Center for Non-invasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, 02215 Boston, USA
| | - Wolnei Caumo
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, Brazil
| | - Iraci L S Torres
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-Clínicas, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, 90035-007 Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), 90050-170 Porto Alegre, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal Rio Grande do Sul, 90050-170 Porto Alegre, Brazil.
| |
Collapse
|
16
|
Regner GG, Torres ILS, de Oliveira C, Pflüger P, da Silva LS, Scarabelot VL, Ströher R, de Souza A, Fregni F, Pereira P. Transcranial direct current stimulation (tDCS) affects neuroinflammation parameters and behavioral seizure activity in pentylenetetrazole-induced kindling in rats. Neurosci Lett 2020; 735:135162. [PMID: 32569808 DOI: 10.1016/j.neulet.2020.135162] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 06/07/2020] [Accepted: 06/14/2020] [Indexed: 10/24/2022]
Abstract
Despite the introduction of new antiepileptic drugs, about 30 % of patients with epilepsy are refractory to drug therapy. Thus, the search for non-pharmacological interventions such as transcranial direct current stimulation (tDCS) may be an alternative, either alone or in combination with low doses of anticonvulsants. This study evaluated the effect of anodal (a-tDCS) and cathodal tDCS (c-tDCS) on seizure behavior and neuroinflammation parameters. Rats were submitted to the kindling model induced by pentylenetetrazole (PTZ) using diazepam (DZP) as anticonvulsant standard. tDCS groups were submitted to 10 sessions of a-tDCS or c-tDCS or SHAM-tDCS. Every 3 days they received saline (SAL), low dose of DZP (alone or in combination with tDCS) or effective dose of DZP 30 min before administration of PTZ, totaling 16 days of protocol. Neither a-tDCS nor c-tDCS reduced the occurrence of clonic forelimb seizures (convulsive motor seizures - stage 3 by the adapted Racine scale we based on). Associated with DZP, c-tDCS (c-tDCS/DZP0.15) increased the latency to first clonic forelimb seizure on the 10th and 16th days. Hippocampal IL-1β levels were reduced by c-tDCS and c-tDCS/DZP0.15. In contrast, these treatments induced an increase in cortical IL-1β levels. Hippocampal TNF-α levels were not altered by c-tDCS or a-tDCS, but c-tDCS and c-tDCS/DZP0.15 increased those levels in cerebral cortex. Cortical NGF levels were increased by c-tDCS and c-tDCS/DZP0.15. a-tDCS/DZP0.15 reduced hippocampal BDNF levels and c-tDCS/DZP0.15 increased these levels in cerebral cortex. In conclusion, c-tDCS alone or in combination with a low dose of DZP showed to affect neuroinflammation, improving central neurotrophin levels and decreasing hippocampal IL-1β levels after PTZ-induced kindling without statistically significant effect on seizure behavior.
Collapse
Affiliation(s)
- Gabriela Gregory Regner
- Laboratory of Neuropharmacology and Preclinical Toxicology Laboratory, Postgraduate Program in Biological Sciences: Pharmacology and Therapeutics, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil; Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Studies - Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil
| | - Iraci L S Torres
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Studies - Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Carla de Oliveira
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Studies - Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil; Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pricila Pflüger
- Laboratory of Neuropharmacology and Preclinical Toxicology Laboratory, Postgraduate Program in Biological Sciences: Pharmacology and Therapeutics, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
| | - Lisiane Santos da Silva
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Studies - Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil
| | - Vanessa Leal Scarabelot
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Studies - Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil
| | - Roberta Ströher
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Studies - Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil
| | - Andressa de Souza
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Studies - Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS 90035-003, Brazil
| | - Felipe Fregni
- Laboratory of Neuromodulation, Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital and Massachusetts General Hospital, Harvard University, Boston, United States
| | - Patrícia Pereira
- Laboratory of Neuropharmacology and Preclinical Toxicology Laboratory, Postgraduate Program in Biological Sciences: Pharmacology and Therapeutics, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil
| |
Collapse
|
17
|
Pedron S, Dumontoy S, Dimauro J, Haffen E, Andrieu P, Van Waes V. Open-tES: An open-source stimulator for transcranial electrical stimulation designed for rodent research. PLoS One 2020; 15:e0236061. [PMID: 32663223 PMCID: PMC7360043 DOI: 10.1371/journal.pone.0236061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Non-invasive neuromodulatory techniques, including transcranial direct current stimulation (tDCS), have been shown to modulate neuronal function and are used both in cognitive neuroscience and to treat neuropsychiatric conditions. In this context, animal models provide a powerful tool to identify the neurobiological mechanisms of action of tDCS. However, finding a current generator that is easily usable and which allows a wide range of stimulation parameters can be difficult and/or expensive. Here, we introduce the Open-tES device, a project under a Creative Commons License (CC BY, SA 4.0) shared on the collaborative platform Git-Hub. This current generator allows tDCS (and other kinds of stimulations) to be realized, is suitable for rodents, is easy to use, and is low-cost. Characterization has been performed to measure the precision and accuracy of the current delivered. We also aimed to compare its effects with a commercial stimulator used in clinical trials (DC-Stimulator Plus, NeuroConn, Germany). To achieve this, a behavioral study was conducted to evaluate its efficacy for decreasing depression related-behavior in mice. The stimulator precision and accuracy were better than 250 nA and 25 nA, respectively. The behavioral evaluation performed in mice in the present study did not reveal any significant differences between the commercial stimulator used in clinical trials and the Open-tES device. Accuracy and precision of the stimulator ensure high repeatability of the stimulations. This current generator constitutes a reliable and inexpensive tool that is useful for preclinical studies in the field of non-invasive electrical brain stimulation.
Collapse
Affiliation(s)
- Solène Pedron
- Laboratory of Integrative and Clinical Neuroscience EA481, Université Bourgogne Franche-Comté, Besançon, France
| | - Stéphanie Dumontoy
- Laboratory of Integrative and Clinical Neuroscience EA481, Université Bourgogne Franche-Comté, Besançon, France
| | - Julien Dimauro
- Laboratory of Integrative and Clinical Neuroscience EA481, Université Bourgogne Franche-Comté, Besançon, France
| | - Emmanuel Haffen
- Laboratory of Integrative and Clinical Neuroscience EA481, Université Bourgogne Franche-Comté, Besançon, France
| | - Patrice Andrieu
- Laboratory of Integrative and Clinical Neuroscience EA481, Université Bourgogne Franche-Comté, Besançon, France
| | - Vincent Van Waes
- Laboratory of Integrative and Clinical Neuroscience EA481, Université Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
18
|
LOW-DOSE NALTREXONE REVERSES FACIAL MECHANICAL ALLODYNIA IN A RAT MODEL OF TRIGEMINAL NEURALGIA. Neurosci Lett 2020; 736:135248. [PMID: 32673692 DOI: 10.1016/j.neulet.2020.135248] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
Abstract
Trigeminal neuralgia (TN) is a type of neuropathic pain characterized by intense pain; although anticonvulsants are used as an option to relieve pain, adverse side effects can decrease patient adherence. In this context, a low dose of naltrexone is effective in relieving pain in other pain conditions. Thus, the objective of the present study was to evaluate the analgesic effect of low-dose naltrexone on facial mechanical allodynia in a rat model of TN, as well as its effect(s) on biomarkers in the central nervous system (tumor necrosis factor-alpha, brain-derived neurotrophic factor [BDNF], interleukin [IL]-10, and toll-like receptor-4). Fifty-nine adult male Wistar rats (CEUA-HCPA#2017-0575) were allocated to following groups: control; sham-pain + vehicle; sham-pain + carbamazepine (100 mg/kg); sham-pain + naltrexone (0.5 mg/kg); pain + vehicle; pain + carbamazepine; and pain + naltrexone. TN was induced using chronic constriction of the infraorbital nerve. Facial allodynia was assessed using von Frey test. Drugs were administered by gavage 14 days after surgery for 10 days. At baseline, the mechanical threshold was similar between groups (P > 0.05; generalized estimating equation). Seven days after surgery, facial allodynia was observed in sham-TN and pain-TN groups (P < 0.05). Fourteen days after surgery, only pain-TN groups exhibited facial allodynia. The first dose of low-dose naltrexone or carbamazepine partially reversed facial allodynia. After 10 days of treatment, both drugs completely reversed it. Spinal cord levels of BDNF and IL-10 were modulated by low-dose naltrexone. Thus, low-dose naltrexone may be suitable to relieve TN; however, the exact mechanisms need to be clarified.
Collapse
|
19
|
Lopes BC, Medeiros LF, Silva de Souza V, Cioato SG, Medeiros HR, Regner GG, Lino de Oliveira C, Fregni F, Caumo W, Torres IL. Transcranial direct current stimulation combined with exercise modulates the inflammatory profile and hyperalgesic response in rats subjected to a neuropathic pain model: Long-term effects. Brain Stimul 2020; 13:774-782. [DOI: 10.1016/j.brs.2020.02.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/06/2020] [Accepted: 02/19/2020] [Indexed: 12/12/2022] Open
|
20
|
Memory and Cognition-Related Neuroplasticity Enhancement by Transcranial Direct Current Stimulation in Rodents: A Systematic Review. Neural Plast 2020; 2020:4795267. [PMID: 32211039 PMCID: PMC7061127 DOI: 10.1155/2020/4795267] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/27/2020] [Accepted: 02/06/2020] [Indexed: 12/19/2022] Open
Abstract
Brain stimulation techniques, including transcranial direct current stimulation (tDCS), were identified as promising therapeutic tools to modulate synaptic plasticity abnormalities and minimize memory and learning deficits in many neuropsychiatric diseases. Here, we revised the effect of tDCS on the modulation of neuroplasticity and cognition in several animal disease models of brain diseases affecting plasticity and cognition. Studies included in this review were searched following the terms (“transcranial direct current stimulation”) AND (mice OR mouse OR animal) and according to the PRISMA statement requirements. Overall, the studies collected suggest that tDCS was able to modulate brain plasticity due to synaptic modifications within the stimulated area. Changes in plasticity-related mechanisms were achieved through induction of long-term potentiation (LTP) and upregulation of neuroplasticity-related proteins, such as c-fos, brain-derived neurotrophic factor (BDNF), or N-methyl-D-aspartate receptors (NMDARs). Taken into account all revised studies, tDCS is a safe, easy, and noninvasive brain stimulation technique, therapeutically reliable, and with promising potential to promote cognitive enhancement and neuroplasticity. Since the use of tDCS has increased as a novel therapeutic approach in humans, animal studies are important to better understand its mechanisms as well as to help improve the stimulation protocols and their potential role in different neuropathologies.
Collapse
|
21
|
Callai EMM, Scarabelot VL, Fernandes Medeiros L, Oliveira C, Souza A, Macedo IC, Cioato SG, Finamor F, Caumo W, Quevedo ADS, Torres ILS. Transcranial direct current stimulation (tDCS) and trigeminal pain: A preclinical study. Oral Dis 2019; 25:888-897. [DOI: 10.1111/odi.13038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 12/04/2018] [Accepted: 12/21/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Etiane Micheli Meyer Callai
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
- Graduate Program in Biological Sciences: Physiology ICBS, UFRGS Porto Alegre Brazil
| | - Vanessa Leal Scarabelot
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
| | - Liciane Fernandes Medeiros
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
- Postgraduate Program in Medical Sciences, School of Medicine Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Carla Oliveira
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
- Postgraduate Program in Medical Sciences, School of Medicine Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Andressa Souza
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
- Post‐Graduate Program in Health and Human Development, Centro Universitário Unilasalle Canoas Brazil
| | - Isabel Cristina Macedo
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
| | - Stefania Giotti Cioato
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics ICBS, UFRGS Porto Alegre Brazil
| | - Fabrício Finamor
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics ICBS, UFRGS Porto Alegre Brazil
| | - Wolnei Caumo
- Postgraduate Program in Medical Sciences, School of Medicine Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Alexandre da Silva Quevedo
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics ICBS, UFRGS Porto Alegre Brazil
| | - Iraci L. S. Torres
- Pharmacology of Pain and Neuromodulation Laboratory: Preclinical Researches, Department of Pharmacology, Institute of Basic Health Sciences (ICBS) Universidade Federal do Rio Grande do Sul (UFRGS) Porto Alegre Brazil
- Animal Experimentation Unit and Graduate Research Group Hospital de Clínicas de Porto Alegre Porto Alegre Brazil
- Graduate Program in Biological Sciences: Physiology ICBS, UFRGS Porto Alegre Brazil
- Postgraduate Program in Medical Sciences, School of Medicine Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics ICBS, UFRGS Porto Alegre Brazil
| |
Collapse
|
22
|
Scarabelot VL, de Oliveira C, Medeiros LF, de Macedo IC, Cioato SG, Adachi LNS, Paz AH, de Souza A, Caumo W, Torres ILS. Transcranial direct-current stimulation reduces nociceptive behaviour in an orofacial pain model. J Oral Rehabil 2018; 46:40-50. [PMID: 30281821 DOI: 10.1111/joor.12726] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 09/17/2018] [Accepted: 09/26/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Transcranial direct-current stimulation (tDCS) is a noninvasive method of brain stimulation suggested as a therapeutic tool for pain and is related to the reversal of maladaptive plasticity associated with chronic pain. OBJECTIVES This study investigated the effect of tDCS, a non-pharmacological therapy, on local mechanical hyperalgesia, and remote thermal hyperalgesia in rats submitted to orofacial inflammatory pain model, by facial von Frey and hot plate tests, respectively. In addition, we evaluated levels of BDNF, NGF, IL-10 and IL-6 in the brainstem and blood serum of these animals at 24 hours and 7 days after the end of tDCS treatment. METHODS Rats were subjected to temporomandibular joint pain and treated with tDCS. The animals were divided into control, pain and pain + treatment groups. Mechanical and thermal hyperalgesia were evaluated at baseline, 7 days after administration of complete Freund's adjuvant, and immediately, 24 hours, and 7 days after the tDCS treatment. Neuroimmunomodulators levels were determined by ELISA. Statistical analyses were performed by (GEE)/Bonferroni (behavioural tests), three-way ANOVA/SNK (neurochemical tests) and Kruskal-Wallis (histological analysis). RESULTS Transcranial direct-current stimulation reduced mechanical and thermal hyperalgesia (P < 0.01). We observed interaction between factors (pain and treatment) increasing brainstem BDNF (P < 0.01) and NGF (P < 0.05) levels. Furthermore, we found an increase in IL-6 and IL-10 levels in the brainstem at 24 hours and 7 days after tDCS, respectively. CONCLUSION We showed that tDCS reduces thermal and mechanical hyperalgesia induced by orofacial pain until 7 days after treatment. These findings demonstrate that tDCS was effective in the control of orofacial inflammatory pain.
Collapse
Affiliation(s)
- Vanessa L Scarabelot
- Post-Graduate Program in Biological Sciences: Physiology, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carla de Oliveira
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Liciane F Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Isabel C de Macedo
- Post-Graduate Program in Biological Sciences: Physiology, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Stefania G Cioato
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lauren Naomi S Adachi
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ana Helena Paz
- Morphological Sciences Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Andressa de Souza
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Wolnei Caumo
- Department of Surgery in Medical School, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Iraci L S Torres
- Post-Graduate Program in Biological Sciences: Physiology, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
23
|
Shahaf G, Kuperman P, Bloch Y, Yariv S, Granovsky Y. Monitoring Migraine Cycle Dynamics with an Easy-to-Use Electrophysiological Marker-A Pilot Study. SENSORS 2018; 18:s18113918. [PMID: 30441751 PMCID: PMC6263618 DOI: 10.3390/s18113918] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/01/2018] [Accepted: 11/11/2018] [Indexed: 11/22/2022]
Abstract
Migraine attacks can cause significant discomfort and reduced functioning for days at a time, including the pre-ictal and post-ictal periods. During the inter-ictsal period, however, migraineurs seem to function normally. It is puzzling, therefore, that event-related potentials of migraine patients often differ in the asymptomatic and inter-ictal period. Part of the electrophysiological dynamics demonstrated in the migraine cycle are attention related. In this pilot study we evaluated an easy-to-use new marker, the Brain Engagement Index (BEI), for attention monitoring during the migraine cycle. We sampled 12 migraine patients for 20 days within one calendar month. Each session consisted of subjects’ reports of stress level and migraine-related symptoms, and a 5 min EEG recording, with a 2-electrode EEG device, during an auditory oddball task. The first minute of the EEG sample was analyzed. Repetitive samples were also obtained from 10 healthy controls. The brain engagement index increased significantly during the pre-ictal (p ≈ 0.001) and the ictal (p ≈ 0.020) periods compared with the inter-ictal period. No difference was observed between the pre-ictal and ictal periods. Control subjects demonstrated intermediate Brain Engagement Index values, that is, higher than inter-ictal, yet lower than pre-ictal. Our preliminary results demonstrate the potential advantage of the use of a simple EEG system for improved prediction of migraine attacks. Further study is required to evaluate the efficacy of the Brain Engagement Index in monitoring the migraine cycle and the possible effects of interventions.
Collapse
Affiliation(s)
| | - Pora Kuperman
- The Laboratory of Clinical Neurophysiology, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| | - Yuval Bloch
- The Emotion-Cognition Research Center, Shalvata Mental Health Care Center, Hod-Hasharon 45100, Israel.
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel.
| | - Shahak Yariv
- Department of Psychiatry, Emek Medical Center, Afula 1834111, Israel.
| | - Yelena Granovsky
- The Laboratory of Clinical Neurophysiology, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
- Department of Neurology, Rambam Medical Center, Haifa 3655306, Israel.
| |
Collapse
|
24
|
Naegel S, Biermann J, Theysohn N, Kleinschnitz C, Diener HC, Katsarava Z, Obermann M, Holle D. Polarity-specific modulation of pain processing by transcranial direct current stimulation - a blinded longitudinal fMRI study. J Headache Pain 2018; 19:99. [PMID: 30355321 PMCID: PMC6755563 DOI: 10.1186/s10194-018-0924-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/24/2018] [Indexed: 12/21/2022] Open
Abstract
Background To enrich the hitherto insufficient understanding regarding the mechanisms of action of transcranial direct current stimulation (tDCS) in pain disorders, we investigated its modulating effects on cerebral pain processing using functional magnetic resonance imaging (fMRI). Methods Thirteen right-handed healthy participants received 20 min of 1.5 mA tDCS applied over the primary motor cortex thrice and under three different stimulation pattern (1.anodal-tDCS, 2.cathodal-tDCS, and 3.sham-tDCS) in a blinded cross-over design. After tDCS neural response to electric trigeminal-nociceptive stimulation was investigated using a block designed fMRI. Results Pain stimulation showed a distinct activation pattern within well-established brain regions associated with pain processing. Following anodal tDCS increased activation was detected in the thalamus, basal ganglia, amygdala, cingulate, precentral, postcentral, and dorsolateral prefrontal cortex, while cathodal t-DCS showed decreased response in these areas (pFWE < 0.05). Interestingly the observed effect was reversed in both control conditions (visual- and motor-stimulation). Behavioral data remained unchanged irrespective of the tDCS stimulation mode. Conclusions This study demonstrates polarity-specific modulation of cerebral pain processing, in reconfirmation of previous electrophysiological data. Anodal tDCS leads to an activation of the central pain-network while cathodal tDCS does not. Results contribute to a network-based understanding of tDCS’s impact on cerebral pain-processing.
Collapse
Affiliation(s)
- Steffen Naegel
- Department of Neurology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany.
| | - Josephine Biermann
- Department of Neurology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Nina Theysohn
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Hans-Christoph Diener
- Department of Neurology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Zaza Katsarava
- Department of Neurology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany.,Department of Neurology, Evangelical Hospital Unna, Holbeinstr. 10, 59423, Unna, Germany.,EVEX Medical Corporation, 40 Vazha-Pshavela Avenue, Tbilisi, 0177, Georgia.,Sechenov University Moscow, 8-2 Trubetskaya str., Moscow, 119991, Russian Federation
| | - Mark Obermann
- Department of Neurology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany.,Center for Neurology, Asklepios Hospitals Schildautal, Karl-Herold-Straße 1, 38723, Seesen, Germany
| | - Dagny Holle
- Department of Neurology, University of Duisburg-Essen, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
| |
Collapse
|
25
|
Souza A, Martins DF, Medeiros LF, Nucci-Martins C, Martins TC, Siteneski A, Caumo W, dos Santos ARS, Torres IL. Neurobiological mechanisms of antiallodynic effect of transcranial direct current stimulation (tDCS) in a mice model of neuropathic pain. Brain Res 2018; 1682:14-23. [DOI: 10.1016/j.brainres.2017.12.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 09/14/2017] [Accepted: 12/08/2017] [Indexed: 01/07/2023]
|
26
|
Ziomber A, Surowka AD, Antkiewicz-Michaluk L, Romanska I, Wrobel P, Szczerbowska-Boruchowska M. Combined brain Fe, Cu, Zn and neurometabolite analysis - a new methodology for unraveling the efficacy of transcranial direct current stimulation (tDCS) in appetite control. Metallomics 2018; 10:397-405. [PMID: 29384550 DOI: 10.1039/c7mt00329c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Obesity is a chronic, multifactorial origin disease that has recently become one of the most frequent lifestyle disorders. Unfortunately, current obesity treatments seem to be ineffective. At present, transcranial direct current brain stimulation (tDCS) represents a promising novel treatment methodology that seems to be efficient, well-tolerated and safe for a patient. Unfortunately, the biochemical action of tDCS remains unknown, which prevents its widespread use in the clinical arena, although neurobiochemical changes in brain signaling and metal metabolism are frequently reported. Therefore, our research aimed at exploring the biochemical response to tDCS in situ, in the brain areas triggering feeding behavior in obese animals. The objective was to propose a novel neurochemical (serotoninergic and dopaminergic signaling) and trace metal analysis of Fe, Cu and Zn. In doing so, we used energy-dispersive X-ray fluorescence (EDXRF) and high-performance liquid chromatography (HPLC). Anodal-type stimulation (atDCS) of the right frontal cortex was utilized to down-regulate food intake and body weight gain in obese rats. EDXRF was coupled with the external standard method in order to quantify the chemical elements within appetite-triggering brain areas. Major dopamine metabolites were assessed in the brains, based on the HPLC assay utilizing the external standard assay. Our study confirms that elemental analysis by EDXRF and brain metabolite assay by HPLC can be considered as a useful tool for the in situ investigation of the interplay between neurochemical and Fe/Cu/Zn metabolism in the brain upon atDCS. With this methodology, an increase in both Cu and Zn in the satiety center of the stimulated group could be reported. In turn, the most significant neurochemical changes involved dopaminergic and serotoninergic signaling in the brain reward system.
Collapse
Affiliation(s)
- Agata Ziomber
- Jagiellonian University, Chair of Pathophysiology, Faculty of Medicine, Krakow, Poland
| | - Artur Dawid Surowka
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, al. A. Mickiewicza 30, 30-059 Krakow, Poland.
| | - Lucyna Antkiewicz-Michaluk
- Department of Neurochemistry, Institute of Pharmacology Polish Academy of Sciences, ul. Smetna 12, 31-343 Kraków, Poland
| | - Irena Romanska
- Department of Neurochemistry, Institute of Pharmacology Polish Academy of Sciences, ul. Smetna 12, 31-343 Kraków, Poland
| | - Pawel Wrobel
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, al. A. Mickiewicza 30, 30-059 Krakow, Poland.
| | - Magdalena Szczerbowska-Boruchowska
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, al. A. Mickiewicza 30, 30-059 Krakow, Poland.
| |
Collapse
|
27
|
Lopes TDS, Silva WDS, Ribeiro SB, Figueiredo CA, Campbell FQ, Daltro GDC, Valenzuela A, Montoya P, Lucena RDCS, Baptista AF. Does Transcranial Direct Current Stimulation Combined with Peripheral Electrical Stimulation Have an Additive Effect in the Control of Hip Joint Osteonecrosis Pain Associated with Sickle Cell Disease? A Protocol for a One-Session Double Blind, Block-Randomized Clinical Trial. Front Hum Neurosci 2017; 11:633. [PMID: 29326577 PMCID: PMC5742338 DOI: 10.3389/fnhum.2017.00633] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/11/2017] [Indexed: 12/29/2022] Open
Abstract
Chronic pain in Sickle Cell Disease (SCD) is probably related to maladaptive plasticity of brain areas involved in nociceptive processing. Transcranial Direct Current Stimulation (tDCS) and Peripheral Electrical Stimulation (PES) can modulate cortical excitability and help to control chronic pain. Studies have shown that combined use of tDCS and PES has additive effects. However, to date, no study investigated additive effects of these neuromodulatory techniques on chronic pain in patients with SCD. This protocol describes a study aiming to assess whether combined use of tDCS and PES more effectively alleviate pain in patients with SCD compared to single use of each technique. The study consists of a one-session double blind, block-randomized clinical trial (NCT02813629) in which 128 participants with SCD and femoral osteonecrosis will be enrolled. Stepwise procedures will occur on two independent days. On day 1, participants will be screened for eligibility criteria. On day 2, data collection will occur in four stages: sample characterization, baseline assessment, intervention, and post-intervention assessment. These procedures will last ~5 h. Participants will be divided into two groups according to homozygous for S allele (HbSS) (n = 64) and heterozygous for S and C alleles (HbSC) (n = 64) genotypes. Participants in each group will be randomly assigned, equally, to one of the following interventions: (1) active tDCS + active PES; (2) active tDCS + sham PES; (3) sham tDCS + active PES; and (4) sham tDCS + sham PES. Active tDCS intervention will consist of 20 min 2 mA anodic stimulation over the primary motor cortex contralateral to the most painful hip. Active PES intervention will consist of 30 min sensory electrical stimulation at 100 Hz over the most painful hip. The main study outcome will be pain intensity, measured by a Visual Analogue Scale. In addition, electroencephalographic power density, cortical maps of the gluteus maximus muscle elicited by Transcranial Magnetic Stimulation (TMS), serum levels of Brain-derived Neurotrophic Factor (BDNF), and Tumor Necrosis Factor (TNF) will be assessed as secondary outcomes. Data will be analyzed using ANOVA of repeated measures, controlling for confounding variables.
Collapse
Affiliation(s)
- Tiago da Silva Lopes
- Health and Functionality Study Group, Federal University of Bahia, Salvador, Brazil.,Graduate Program in Medicine and Health, Federal University of Bahia, Salvador, Brazil
| | - Wellington Dos Santos Silva
- Health and Functionality Study Group, Federal University of Bahia, Salvador, Brazil.,Graduate Program in Medicine and Health, Federal University of Bahia, Salvador, Brazil.,Health Section, Adventist Faculty of Bahia, Cachoeira, Brazil
| | - Sânzia B Ribeiro
- Health and Functionality Study Group, Federal University of Bahia, Salvador, Brazil.,Health Section, Adventist Faculty of Bahia, Cachoeira, Brazil
| | | | - Fernanda Q Campbell
- Health and Functionality Study Group, Federal University of Bahia, Salvador, Brazil
| | | | | | - Pedro Montoya
- Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
| | - Rita de C S Lucena
- Health and Functionality Study Group, Federal University of Bahia, Salvador, Brazil.,Graduate Program in Medicine and Health, Federal University of Bahia, Salvador, Brazil
| | - Abrahão F Baptista
- Health and Functionality Study Group, Federal University of Bahia, Salvador, Brazil.,Graduate Program in Medicine and Health, Federal University of Bahia, Salvador, Brazil.,Center for Mathematics, Computation and Cognition, Federal University of ABC, São Bernardo do Campo, Brazil
| |
Collapse
|
28
|
Abstract
Transcranial electrical brain stimulation can modulate cortical excitability and plasticity in humans and rodents. The most common form of stimulation in humans is transcranial direct current stimulation (tDCS). Less frequently, transcranial alternating current stimulation (tACS) or transcranial random noise stimulation (tRNS), a specific form of tACS using an electrical current applied randomly within a pre-defined frequency range, is used. The increase of noninvasive electrical brain stimulation research in humans, both for experimental and clinical purposes, has yielded an increased need for basic, mechanistic, safety studies in animals. This article describes a model for transcranial electrical brain stimulation (tES) through the intact skull targeting the motor system in alert rodents. The protocol provides step-by-step instructions for the surgical set-up of a permanent epicranial electrode socket combined with an implanted counter electrode on the chest. By placing a stimulation electrode into the epicranial socket, different electrical stimulation types, comparable to tDCS, tACS, and tRNS in humans, can be delivered. Moreover, the practical steps for tES in alert rodents are introduced. The applied current density, stimulation duration, and stimulation type may be chosen depending on the experimental needs. The caveats, advantages, and disadvantages of this set-up are discussed, as well as safety and tolerability aspects.
Collapse
Affiliation(s)
- Brita Fritsch
- Department of Neurology, Albert-Ludwigs-University Freiburg;
| | | | - Janine Reis
- Department of Neurology, Albert-Ludwigs-University Freiburg
| |
Collapse
|
29
|
Schunck RVA, Macedo IC, Laste G, de Souza A, Valle MTC, Salomón JLO, Nunes EA, Campos ACW, Gnoatto SCB, Bergold AM, Konrath EL, Dallegrave E, Arbo MD, Torres ILS, Leal MB. Standardized Passiflora incarnata L. Extract Reverts the Analgesia Induced by Alcohol Withdrawal in Rats. Phytother Res 2017; 31:1199-1208. [PMID: 28568647 DOI: 10.1002/ptr.5839] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 04/14/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022]
Abstract
Passiflora incarnata L. (Passifloraceae) has been traditionally used for treatment of anxiety, insomnia, drug addiction, mild infections, and pain. The aim of this study was to investigate the effect of a commercial extract of P. incarnata in the analgesia induced by alcohol withdrawal syndrome in rats. In addition, brain-derived neurotrophic factor and interleukin-10 levels were evaluated in prefrontal cortex, brainstem, and hippocampus. Male adult rats received by oral gavage: (1: water group) water for 19 days, 1 day interval and water (8 days); (2: P. incarnata group) water for 19 days, 1 day interval and P. incarnata 200 mg/kg (8 days); (3: alcohol withdrawal group) alcohol for 19 days, 1 day interval and water (8 days); and (4: P. incarnata in alcohol withdrawal) alcohol for 19 days, 1 day interval and P. incarnata 200 mg/kg (8 days). The tail-flick and hot plate tests were used as nociceptive response measures. Confirming previous study of our group, it was showed that alcohol-treated groups presented an increase in the nociceptive thresholds after alcohol withdrawal, which was reverted by P. incarnata, measured by the hot plate test. Besides, alcohol treatment increased brain-derived neurotrophic factor and interleukin-10 levels in prefrontal cortex, which was not reverted by P. incarnata. Considering these results, the P. incarnata treatment might be a potential therapy in the alcohol withdrawal syndrome. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Rebeca Vargas Antunes Schunck
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/107, 90050-170, Porto Alegre, RS, Brazil.,Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/202, 90050-170, Porto Alegre, RS, Brazil
| | - Isabel Cristina Macedo
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Gabriela Laste
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Andressa de Souza
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Marina Tuerlinckx Costa Valle
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/107, 90050-170, Porto Alegre, RS, Brazil.,Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/202, 90050-170, Porto Alegre, RS, Brazil
| | - Janaína L O Salomón
- Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/202, 90050-170, Porto Alegre, RS, Brazil
| | - Ellen Almeida Nunes
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Andreia Cristina Wildner Campos
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Simone Cristina Baggio Gnoatto
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Ana Maria Bergold
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Eduardo L Konrath
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Eliane Dallegrave
- Departamento de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245, 90050-170, Porto Alegre, RS, Brazil
| | - Marcelo Dutra Arbo
- Laboratório de Toxicologia (LATOX), Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Av. Ipiranga 2752, 90050-000, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/107, 90050-170, Porto Alegre, RS, Brazil.,Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, 90050-170, Porto Alegre, RS, Brazil
| | - Mirna Bainy Leal
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/107, 90050-170, Porto Alegre, RS, Brazil.,Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500/202, 90050-170, Porto Alegre, RS, Brazil
| |
Collapse
|
30
|
Scarabelot VL, Medeiros LF, de Oliveira C, Adachi LNS, de Macedo IC, Cioato SG, de Freitas JS, de Souza A, Quevedo A, Caumo W, Torres ILDS. Melatonin Alters the Mechanical and Thermal Hyperalgesia Induced by Orofacial Pain Model in Rats. Inflammation 2017; 39:1649-59. [PMID: 27378529 DOI: 10.1007/s10753-016-0399-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Melatonin is a neuroendocrine hormone that presents a wide range of physiological functions including regulating circadian rhythms and sleep, enhancing immune function, sleep improvement, and antioxidant effects. In addition, melatonin has received special attention in pain treatment since it is effective and presents few adverse effects. In this study, we evaluated the effect of acute dose of melatonin upon hyperalgesia induced by complete Freund's adjuvant in a chronic orofacial pain model in Sprague-Dawley rats. Nociceptive behavior was assessed by facial Von Frey and the hot plate tests at baseline and thereafter 30, 60, and 120 min, 24 h, and 7 days after melatonin treatment. We demonstrated that acute melatonin administration alters mechanical and thermal hyperalgesia induced by an orofacial pain model (TMD), highlighting that the melatonin effect upon mechanical hyperalgesia remained until 7 days after its administration. Besides, we observed specific tissue profiles of neuroimmunomodulators linked to pain conditions and/or melatonin effect (brain-derived neurotrophic factor, nerve growth factor, and interleukins 6 and 10) in the brainstem levels, and its effects were state-dependent of the baseline of these animals.
Collapse
Affiliation(s)
- Vanessa Leal Scarabelot
- Postgraduate Program in Biological Sciences: Physiology, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90050-170, Brazil
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
- Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-003, RS, Brazil
| | - Liciane Fernandes Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
- Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-003, RS, Brazil
| | - Carla de Oliveira
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
- Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-003, RS, Brazil
- Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lauren Naomi Spezia Adachi
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
- Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-003, RS, Brazil
- Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Isabel Cristina de Macedo
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
- Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-003, RS, Brazil
| | - Stefania Giotti Cioato
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
- Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-003, RS, Brazil
| | - Joice S de Freitas
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
- Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-003, RS, Brazil
| | - Andressa de Souza
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
| | - Alexandre Quevedo
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil
| | - Wolnei Caumo
- Department of Surgery in Medical School, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90035-003, RS, Brazil
| | - Iraci Lucena da Silva Torres
- Postgraduate Program in Biological Sciences: Physiology, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90050-170, Brazil.
- Laboratory of Pain Pharmacology and Neuromodulation: Preclinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500 sala 305, Porto Alegre, 90050-170, RS, Brazil.
- Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-003, RS, Brazil.
- Postgraduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
31
|
Caumo W, Deitos A, Carvalho S, Leite J, Carvalho F, Dussán-Sarria JA, Lopes Tarragó MDG, Souza A, Torres ILDS, Fregni F. Motor Cortex Excitability and BDNF Levels in Chronic Musculoskeletal Pain According to Structural Pathology. Front Hum Neurosci 2016; 10:357. [PMID: 27471458 PMCID: PMC4946131 DOI: 10.3389/fnhum.2016.00357] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/30/2016] [Indexed: 12/26/2022] Open
Abstract
The central sensitization syndrome (CSS) encompasses disorders with overlapping symptoms in a structural pathology spectrum ranging from persistent nociception [e.g., osteoarthritis (OA)] to an absence of tissue injuries such as the one presented in fibromyalgia (FM) and myofascial pain syndrome (MPS). First, we hypothesized that these syndromes present differences in their cortical excitability parameters assessed by transcranial magnetic stimulation (TMS), namely motor evoked potential (MEP), cortical silent period (CSP), short intracortical inhibition (SICI) and short intracortical facilitation (SICF). Second, considering that the presence of tissue injury could be detected by serum neurotrophins, we hypothesized that the spectrum of structural pathology (i.e., from persistent nociception like in OA, to the absence of tissue injury like in FM and MPS), could be detected by differential efficiency of their descending pain inhibitory system, as assessed by the conditioned pain modulation (CPM) paradigm. Third, we explored whether brain-derived neurotrophic factor (BDNF) had an influence on the relationship between motor cortex excitability and structural pathology. This cross-sectional study pooled baseline data from three randomized clinical trials. We included females (n = 114), aged 19-65 years old with disability by chronic pain syndromes (CPS): FM (n = 19), MPS (n = 54), OA (n = 27) and healthy subjects (n = 14). We assessed the serum BDNF, the motor cortex excitability by parameters the TMS measures and the change on numerical pain scale [NPS (0-10)] during CPM-task. The adjusted mean (SD) on the SICI observed in the absence of tissue injury was 56.36% lower than with persistent nociceptive input [0.31(0.18) vs. 0.55 (0.32)], respectively. The BDNF was inversely correlated with the SICI and with the change on NPS (0-10)during CPM-task. These findings suggest greater disinhibition in the motor cortex and the descending pain inhibitory system in FM and MPS than in OA and healthy subjects. Likewise, the inter-hemispheric disinhibition as well as the dysfunction in the descending pain modulatory system is higher in chronic pain without tissue injury compared to a structural lesion. In addition, they suggest that a greater level of serum BDNF may be involved in the processes that mediate the disinhibition of motor cortex excitability, as well as the function of descending inhibitory pain modulation system, independently of the physiopathology mechanism of musculoskeletal pain syndromes.
Collapse
Affiliation(s)
- Wolnei Caumo
- Post-graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS)Porto Alegre, Brazil; Laboratory of Pain and Neuromodulation at UFRGSPorto Alegre, Brazil; Anesthesiologist, Pain and Palliative Care Service at Hospital de Clínicas de Porto Alegre (HCPA)Porto Alegre, Brazil; Pain and Anesthesia in Surgery Department, School of Medicine, UFRGSPorto Alegre, Brazil
| | - Alícia Deitos
- Post-graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS)Porto Alegre, Brazil; Laboratory of Pain and Neuromodulation at UFRGSPorto Alegre, Brazil
| | - Sandra Carvalho
- Neuropsychophysiology Laboratory, CIPsi, School of Psychology (EPsi), University of Minho, Campus de Gualtar Braga, Portugal
| | - Jorge Leite
- Neuropsychophysiology Laboratory, CIPsi, School of Psychology (EPsi), University of Minho, Campus de Gualtar Braga, Portugal
| | - Fabiana Carvalho
- Post-graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS)Porto Alegre, Brazil; Laboratory of Pain and Neuromodulation at UFRGSPorto Alegre, Brazil
| | - Jairo Alberto Dussán-Sarria
- Post-graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS)Porto Alegre, Brazil; Laboratory of Pain and Neuromodulation at UFRGSPorto Alegre, Brazil
| | - Maria da Graça Lopes Tarragó
- Post-graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS)Porto Alegre, Brazil; Laboratory of Pain and Neuromodulation at UFRGSPorto Alegre, Brazil
| | - Andressa Souza
- Post-graduate Program in Health and Human Development, La Salle University Center Canoas, Brazil
| | - Iraci Lucena da Silva Torres
- Post-graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS)Porto Alegre, Brazil; Department of Pharmacology, Instituto de Ciências Básicas da Saúde, UFRGSPorto Alegre, Brazil
| | - Felipe Fregni
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School Boston, MA, USA
| |
Collapse
|
32
|
Botelho LM, Morales-Quezada L, Rozisky JR, Brietzke AP, Torres ILS, Deitos A, Fregni F, Caumo W. A Framework for Understanding the Relationship between Descending Pain Modulation, Motor Corticospinal, and Neuroplasticity Regulation Systems in Chronic Myofascial Pain. Front Hum Neurosci 2016; 10:308. [PMID: 27445748 PMCID: PMC4921500 DOI: 10.3389/fnhum.2016.00308] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/07/2016] [Indexed: 12/30/2022] Open
Abstract
Myofascial pain syndrome (MPS) is a leading cause of chronic musculoskeletal pain. However, its neurobiological mechanisms are not entirely elucidated. Given the complex interaction between the networks involved in pain process, our approach, to providing insights into the neural mechanisms of pain, was to investigate the relationship between neurophysiological, neurochemical and clinical outcomes such as corticospinal excitability. Recent evidence has demonstrated that three neural systems are affected in chronic pain: (i) motor corticospinal system; (ii) internal descending pain modulation system; and (iii) the system regulating neuroplasticity. In this cross-sectional study, we aimed to examine the relationship between these three central systems in patients with chronic MPS of whom do/do not respond to the Conditioned Pain Modulation Task (CPM-task). The CPM-task was to immerse her non-dominant hand in cold water (0-1°C) to produce a heterotopic nociceptive stimulus. Corticospinal excitability was the primary outcome; specifically, the motor evoked potential (MEP) and intracortical facilitation (ICF) as assessed by transcranial magnetic stimulation (TMS). Secondary outcomes were the cortical excitability parameters [current silent period (CSP) and short intracortical inhibition (SICI)], serum brain-derived neurotrophic factor (BDNF), heat pain threshold (HPT), and the disability related to pain (DRP). We included 33 women, (18-65 years old). The MANCOVA model using Bonferroni's Multiple Comparison Test revealed that non-responders (n = 10) compared to responders (n = 23) presented increased intracortical facilitation (ICF; mean ± SD) 1.43 (0.3) vs. 1.11 (0.12), greater motor-evoked potential amplitude (μV) 1.93 (0.54) vs. 1.40 (0.27), as well a higher serum BDNF (pg/Ml) 32.56 (9.95) vs. 25.59 (10.24), (P < 0.05 for all). Also, non-responders presented a higher level of DRP and decreased HPT (P < 0.05 for all). These findings suggest that the loss of net descending pain inhibition was associated with an increase in ICF, serum BDNF levels, and DRP. We propose a framework to explain the relationship and potential directionality of these factors. In this framework we hypothesize that increased central sensitization leads to a loss of descending pain inhibition that triggers compensatory mechanisms as shown by increased motor cortical excitability.
Collapse
Affiliation(s)
- Leonardo M Botelho
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Pain and Palliative Care Service at Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Laboratory of Pain and Neuromodulation, Hospital de Clínicas de Porto AlegrePorto Alegre, Brazil
| | - Leon Morales-Quezada
- Laboratory of Neuromodulation of Spaulding Rehabilitation of Harvard Medical School Boston, MA, USA
| | - Joanna R Rozisky
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Laboratory of Pain and Neuromodulation, Hospital de Clínicas de Porto AlegrePorto Alegre, Brazil
| | - Aline P Brietzke
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Laboratory of Pain and Neuromodulation, Hospital de Clínicas de Porto AlegrePorto Alegre, Brazil
| | - Iraci L S Torres
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Pharmacology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil
| | - Alicia Deitos
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Laboratory of Pain and Neuromodulation, Hospital de Clínicas de Porto AlegrePorto Alegre, Brazil
| | - Felipe Fregni
- Laboratory of Neuromodulation of Spaulding Rehabilitation of Harvard Medical SchoolBoston, MA, USA; Department of Physical Medicine and Rehabilitation, Harvard Medical SchoolBoston, MA, USA
| | - Wolnei Caumo
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Pain and Palliative Care Service at Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Laboratory of Pain and Neuromodulation, Hospital de Clínicas de Porto AlegrePorto Alegre, Brazil; Surgery Department, School of Medicine, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil
| |
Collapse
|
33
|
Shahaf G. Migraine as dysfunctional drive reduction: Insight from electrophysiology. Med Hypotheses 2016; 91:62-66. [DOI: 10.1016/j.mehy.2016.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/11/2016] [Indexed: 11/24/2022]
|
34
|
Dimov LF, Franciosi AC, Campos ACP, Brunoni AR, Pagano RL. Top-Down Effect of Direct Current Stimulation on the Nociceptive Response of Rats. PLoS One 2016; 11:e0153506. [PMID: 27071073 PMCID: PMC4829148 DOI: 10.1371/journal.pone.0153506] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/30/2016] [Indexed: 01/06/2023] Open
Abstract
Transcranial direct current stimulation (tDCS) is an emerging, noninvasive technique of neurostimulation for treating pain. However, the mechanisms and pathways involved in its analgesic effects are poorly understood. Therefore, we investigated the effects of direct current stimulation (DCS) on thermal and mechanical nociceptive thresholds and on the activation of the midbrain periaqueductal gray (PAG) and the dorsal horn of the spinal cord (DHSC) in rats; these central nervous system areas are associated with pain processing. Male Wistar rats underwent cathodal DCS of the motor cortex and, while still under stimulation, were evaluated using tail-flick and paw pressure nociceptive tests. Sham stimulation and naive rats were used as controls. We used a randomized design; the assays were not blinded to the experimenter. Immunoreactivity of the early growth response gene 1 (Egr-1), which is a marker of neuronal activation, was evaluated in the PAG and DHSC, and enkephalin immunoreactivity was evaluated in the DHSC. DCS did not change the thermal nociceptive threshold; however, it increased the mechanical nociceptive threshold of both hind paws compared with that of controls, characterizing a topographical effect. DCS decreased the Egr-1 labeling in the PAG and DHSC as well as the immunoreactivity of spinal enkephalin. Altogether, the data suggest that DCS disinhibits the midbrain descending analgesic pathway, consequently inhibiting spinal nociceptive neurons and causing an increase in the nociceptive threshold. This study reinforces the idea that the motor cortex participates in the neurocircuitry that is involved in analgesia and further clarifies the mechanisms of action of tDCS in pain treatment.
Collapse
Affiliation(s)
- Luiz Fabio Dimov
- Laboratory of Neuromodulation and Experimental Pain, Hospital Sírio Libanês, Rua Prof Daher Cutait, 69, Sao Paulo, SP, 01308-060, Brazil
| | - Adriano Cardozo Franciosi
- Laboratory of Neuromodulation and Experimental Pain, Hospital Sírio Libanês, Rua Prof Daher Cutait, 69, Sao Paulo, SP, 01308-060, Brazil
| | - Ana Carolina Pinheiro Campos
- Laboratory of Neuromodulation and Experimental Pain, Hospital Sírio Libanês, Rua Prof Daher Cutait, 69, Sao Paulo, SP, 01308-060, Brazil
| | - André Russowsky Brunoni
- Service of Interdisciplinary Neuromodulation (SIN), Department and Institute of Psychiatry, Faculty of Medicine of University of São Paulo, Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, University of São Paulo, Rua Doutor Ovidio Pires de Campos, 785, Sao Paulo, SP, 05403-000, Brazil.,Center for Clinical and Epidemiological Research & Interdisciplinary Center for Applied Neuromodulation (CINA), University Hospital, University of São Paulo, São Paulo, Avenida Professor Lineu Prestes 2565, ext. 3, Sao Paulo, SP, 05508-000, Brazil
| | - Rosana Lima Pagano
- Laboratory of Neuromodulation and Experimental Pain, Hospital Sírio Libanês, Rua Prof Daher Cutait, 69, Sao Paulo, SP, 01308-060, Brazil
| |
Collapse
|
35
|
Cioato SG, Medeiros LF, Marques Filho PR, Vercelino R, de Souza A, Scarabelot VL, de Oliveira C, Adachi LNS, Fregni F, Caumo W, Torres IL. Long-Lasting Effect of Transcranial Direct Current Stimulation in the Reversal of Hyperalgesia and Cytokine Alterations Induced by the Neuropathic Pain Model. Brain Stimul 2016; 9:209-17. [DOI: 10.1016/j.brs.2015.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 11/05/2015] [Accepted: 12/06/2015] [Indexed: 12/27/2022] Open
|
36
|
Filho PRM, Vercelino R, Cioato SG, Medeiros LF, de Oliveira C, Scarabelot VL, Souza A, Rozisky JR, Quevedo ADS, Adachi LNS, Sanches PRS, Fregni F, Caumo W, Torres ILS. Transcranial direct current stimulation (tDCS) reverts behavioral alterations and brainstem BDNF level increase induced by neuropathic pain model: Long-lasting effect. Prog Neuropsychopharmacol Biol Psychiatry 2016; 64:44-51. [PMID: 26160698 DOI: 10.1016/j.pnpbp.2015.06.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/24/2015] [Accepted: 06/30/2015] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Neuropathic pain (NP) is a chronic pain modality that usually results of damage in the somatosensory system. NP often shows insufficient response to classic analgesics and remains a challenge to medical treatment. The transcranial direct current stimulation (tDCS) is a non-invasive technique, which induces neuroplastic changes in central nervous system of animals and humans. The brain derived neurotrophic factor plays an important role in synaptic plasticity process. Behavior changes such as decreased locomotor and exploratory activities and anxiety disorders are common comorbidities associated with NP. OBJECTIVE Evaluate the effect of tDCS treatment on locomotor and exploratory activities, and anxiety-like behavior, and peripheral and central BDNF levels in rats submitted to neuropathic pain model. METHODS Rats were randomly divided: Ss, SsS, SsT, NP, NpS, and NpT. The neuropathic pain model was induced by partial sciatic nerve compression at 14 days after surgery; the tDCS treatment was initiated. The animals of treated groups were subjected to a 20 minute session of tDCS, for eight days. The Open Field and Elevated Pluz Maze tests were applied 24 h (phase I) and 7 days (phase II) after the end of tDCS treatment. The serum, spinal cord, brainstem and cerebral cortex BDNF levels were determined 48 h (phase I) and 8 days (phase II) after tDCS treatment by ELISA. RESULTS The chronic constriction injury (CCI) induces decrease in locomotor and exploratory activities, increases in the behavior-like anxiety, and increases in the brainstem BDNF levels, the last, in phase II (one-way ANOVA/SNK, P<0.05 for all). The tDCS treatment already reverted all these effects induced by CCI (one-way ANOVA/SNK, P<0.05 for all). Furthermore, the tDCS treatment decreased serum and cerebral cortex BDNF levels and it increased these levels in the spinal cord in phase II (one-way ANOVA/SNK, P<0.05). CONCLUSION tDCS reverts behavioral alterations associated to neuropathic pain, indicating possible analgesic and anxiolytic tDCS effects. tDCS treatment induces changes in the BDNF levels in different regions of the central nervous system (CNS), and this effect can be attributed to different cellular signaling activations.
Collapse
Affiliation(s)
- Paulo Ricardo Marques Filho
- Post-Graduate Program in Medicine: Medical Sciences - Medicine School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Rafael Vercelino
- Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Post-Graduate Program in Biological Sciences - Physiology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil
| | - Stefania Giotti Cioato
- Post-Graduate Program in Medicine: Medical Sciences - Medicine School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Liciane Fernandes Medeiros
- Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Post-Graduate Program in Biological Sciences: Pharmacology and Experimental Therapeutic, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil
| | - Carla de Oliveira
- Post-Graduate Program in Medicine: Medical Sciences - Medicine School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Vanessa Leal Scarabelot
- Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Post-Graduate Program in Biological Sciences - Physiology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil
| | - Andressa Souza
- Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil
| | - Joanna Ripoll Rozisky
- Post-Graduate Program in Medicine: Medical Sciences - Medicine School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Alexandre da Silva Quevedo
- Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil
| | - Lauren Naomi Spezia Adachi
- Post-Graduate Program in Medicine: Medical Sciences - Medicine School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Paulo Roberto S Sanches
- Biomedical Engineering of Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Felipe Fregni
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Wolnei Caumo
- Post-Graduate Program in Medicine: Medical Sciences - Medicine School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Iraci L S Torres
- Post-Graduate Program in Medicine: Medical Sciences - Medicine School, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Pharmacology of Pain and Neuromodulation Laboratory: Pre-clinical Researches Department of Pharmacology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Post-Graduate Program in Biological Sciences - Physiology, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil; Post-Graduate Program in Biological Sciences: Pharmacology and Experimental Therapeutic, Universidade Federal do Rio Grande do Sul, ICBS, Porto Alegre, RS 90050-170, Brazil.
| |
Collapse
|
37
|
da Graca-Tarragó M, Deitos A, Patrícia Brietzke A, Torres ILS, Cadore Stefani L, Fregni F, Caumo W. Electrical Intramuscular Stimulation in Osteoarthritis Enhances the Inhibitory Systems in Pain Processing at Cortical and Cortical Spinal System. PAIN MEDICINE 2015; 17:877-891. [PMID: 26398594 DOI: 10.1111/pme.12930] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/13/2015] [Accepted: 08/16/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To determine if in knee osteoarthritis (KOA), one session of active electrical intramuscular stimulation (a-EIMS) compared with sham causes an effect on the motor cortex excitability parameters [motor evoked potential (MEP; the primary outcome), short intracortical inhibition (SICI), intracortical facilitation (ICF) and cortical silent period (CSP)] and pain measurements [pain pressure threshold (PPT); visual analog scale (VAS) and change in numerical pain scale (NPS0-10 ) during the conditioned pain modulation (CPM)-task]. This study also set out to determine if serum brain-derived neurotrophic factor (BDNF) mediates the effect of treatment on the cortical spinal system as assessed by MEP and PPT. DESIGN Randomized clinical trial. SUBJECTS AND METHODS Women with KOA, 50-75-years old received a 30-min session of either sham (n = 13) or a-EIMS (n = 13) with 2 Hz. The pain measures and excitability parameters were measured before and immediately after a-EIMS or sham. RESULTS The a-EIMS group compared with sham decreased the MEP by 31,67% [confidence interval (CI) 95%, 2.34-60.98]. For the secondary outcomes, the a-EIMS reduced the ICF and increased the CSP but not changed the SICI. The a-EIMS improved the pain reported on VAS, the PPT, and the score of the NPS (0-10) during the CPM-task The BDNF was negatively correlated with the PPT (r = -0.56). CONCLUSIONS The serum BDNF revealed an inverse relationship with PPT independent of the treatment group. These results suggest that a-EIMS enhanced the corticospinal inhibitory systems in cortical and infracortical pain processing sites most likely by bottom-up regulation mechanisms.
Collapse
Affiliation(s)
- Maria da Graca-Tarragó
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain and Neuromodulation at HCPA, Porto Alegre, Brazil
| | - Alícia Deitos
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain and Neuromodulation at HCPA, Porto Alegre, Brazil
| | - Aline Patrícia Brietzke
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain and Neuromodulation at HCPA, Porto Alegre, Brazil
| | - Iraci L S Torres
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Pharmacology Department, Instituto De Ciências Básicas Da Saúde, UFRGS, Porto Alegre, Brazil
| | - Luciana Cadore Stefani
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain and Neuromodulation at HCPA, Porto Alegre, Brazil.,Surgery Department, School of Medicine at UFRGS, Porto Alegre, Brazil.,Anesthesia and Perioperative Pain Medicine at Hospital De Clínicas De Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Felipe Fregni
- Department of Physical Medicine and Rehabilitation Boston, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolnei Caumo
- Post-Graduate Program in Medical Sciences, School of Medicine, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain and Neuromodulation at HCPA, Porto Alegre, Brazil.,Surgery Department, School of Medicine at UFRGS, Porto Alegre, Brazil.,Pain and Palliative Care Service at Hospital De Clínicas De Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|