Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Integr Neurosci ; 23(3): 57, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38538226

RESUMO

Recently, novel non-pharmacological interventions, such as photobiomodulation (PBM) therapy, have shown promise for the treatment of Alzheimer's disease (AD). This article outlines the translation from the preclinical to clinical stages of an innovative brain-gut PBM therapy in a mouse model of AD, a pilot clinical trial involving mild-to-moderate AD patients, and a continuing pivotal clinical trial with a similar patient population. In a mouse model of AD (Aß25-35), daily application of brain-gut PBM therapy to both the head and the abdomen produced a neuroprotective effect against the neurotoxic effects of an Aß25-35 peptide injection by normalizing all the modified behavioral and biochemical parameters. The pilot clinical trial to evaluate brain-gut PBM therapy demonstrated the tolerability and feasibility of the novel PBM-based treatment for mild-to-moderate AD patients. Compared to the sham patients, the PBM-treated patients had lower Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog) comprehension sub-scores, higher forward verbal spans, and lower Trail Making Test (TMT) Part B (TMT-B) execution times, which suggest an improvement in cognitive functions. This pilot study provided important information for the design of a novel pivotal clinical trial, currently in progress, to assess the efficacy of brain-gut PBM therapy in a larger sample of AD patients. This pivotal clinical trial could demonstrate that brain-gut PBM therapy is a safe, well-tolerated, and efficient disease-modifying treatment for mild-to-moderate AD patients and that it has medical and economic benefits.


Assuntos
Doença de Alzheimer , Terapia com Luz de Baixa Intensidade , Animais , Camundongos , Humanos , Doença de Alzheimer/radioterapia , Doença de Alzheimer/tratamento farmacológico , Projetos Piloto , Encéfalo , Cognição
2.
J Alzheimers Dis ; 90(2): 811-822, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36189591

RESUMO

BACKGROUND: Recent innovative non-pharmacological interventions and neurostimulation devices have shown potential for application in the treatment of Alzheimer's disease (AD). These include photobiomodulation (PBM) therapy. OBJECTIVE: This pilot study assesses the safety, compliance with, and efficacy of a brain-gut PBM therapy for mild-to-moderate AD patients. METHODS: This double-blind, randomized, monocentric sham-controlled study started in 2018 and ended prematurely in 2020 due to the COVID-19 pandemic. Fifty-three mild-to-moderate AD patients were randomized, 27 in the PBM group and 26 in the sham group. All patients had 40 treatment sessions lasting 25 min each over 8 weeks and were followed for 4 weeks afterwards. Compliance with the treatment was recorded. Safety was assessed by recording adverse events (AEs), and efficacy was evaluated using neuropsychological tests. RESULTS: The PBM therapy proved to be safe in regard to the number of recorded AEs (44% of the patients), which were balanced between the PBM and sham groups. AEs were mainly mild, and no serious AEs were reported. The majority of the patients (92.5%) were highly compliant, which confirms the feasibility of the PBM treatment. Compared to the sham patients, the PBM patients showed lower ADAS-Cog comprehension subscores, higher forward verbal spans, and lower TMT-B execution times, which suggests an improvement in cognitive functions. CONCLUSION: This study demonstrates the tolerability of and patient compliance with a PBM-based treatment for mild-to-moderate AD patients. It highlights encouraging efficacy trends and provides insights for the design of the next phase trial in a larger AD patient sample.


Assuntos
Doença de Alzheimer , COVID-19 , Terapia com Luz de Baixa Intensidade , Humanos , Projetos Piloto , Pandemias , Resultado do Tratamento , Doença de Alzheimer/radioterapia , Doença de Alzheimer/tratamento farmacológico , Encéfalo , Método Duplo-Cego , Cooperação do Paciente
3.
J Alzheimers Dis ; 83(4): 1453-1469, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34487045

RESUMO

BACKGROUND: Major depressive disorder (MDD) is prevalent and has significant impact on individuals and society. Cognitive symptoms are frequent in MDD and insufficiently treated by antidepressant medications. Transcranial photobiomodulation (t-PBM) is a novel device therapy which shows promise as an antidepressant and pro-cognitive treatment. To date, despite the encouraging results, the optimal stimulation parameters of t-PBM to treat MDD are not established, and clinical studies are very heterogeneous in terms of these parameters. While the literature provides guidance on the appropriate fluence to achieve therapeutic results, little is known on the other parameters. OBJECTIVE: To evaluate the relationship between different parameters and the antidepressant effect of t-PBM. METHODS: We reviewed clinical studies on MDD and on depressive symptoms comorbid with other diseases. We calculated the standardized effect size of the change in symptoms severity before and after t-PBM and we performed a descriptive analysis of the reviewed papers. RESULTS: The greatest effect sizes for the antidepressant effect were found in studies using pulse-wave t-PBM with high peak irradiance (but low average irradiance) over large skin surface. One well-designed and sufficiently powered, double-blind, sham-controlled trial indicated that t-PBM with low irradiance over a small skin surface is ineffective to treat depression. CONCLUSION: The use of t-PBM for Alzheimer's disease and for dementia is still at its inception; these dosimetry lessons from the use of t-PBM for depression might serve as guidance.


Assuntos
Doença de Alzheimer/radioterapia , Transtorno Depressivo Maior/terapia , Terapia com Luz de Baixa Intensidade , Doença de Alzheimer/diagnóstico , Método Duplo-Cego , Humanos , Resultado do Tratamento
5.
J Alzheimers Dis ; 83(4): 1415-1429, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34219711

RESUMO

BACKGROUND: Anxious-depressive-like behavior has been recognized as an early endophenotype in Alzheimer's disease (AD). Recent studies support early treatment of anxious-depressive-like behavior as a potential target to alleviate memory loss and reduce the risk of developing dementia. We hypothesize that photobiomodulation (PBM) could be an effective method to alleviate depression and anxiety at the early stage of AD pathogenesis. OBJECTIVE: To analyze the effect of PBM treatment on anxious-depressive-like behavior at the early stage of AD. METHODS: Using a novel transgenic AD rat model, animals were divided into wild-type, AD+sham PBM, and AD+PBM groups. Two-minute daily PBM (irradiance: 25 mW/cm2 and fluence: 3 J/cm2 at the cortical level) was applied transcranially to the brain of AD animals from 2 months of age to 10 months of age. After completing PBM treatment at 10 months of age, behavioral tests were performed to measure learning, memory, and anxious-depressive-like behavior. Neuronal apoptosis, neuronal degeneration, neuronal damage, mitochondrial function, neuroinflammation, and oxidative stress were measured to test the effects of PBM on AD animals. RESULTS: Behavioral tests showed that: 1) no spatial memory deficits were detected in TgF344 rats at 10 months of age; 2) PBM alleviated anxious-depressive-like behavior in TgF344 rats; 3) PBM attenuated neuronal damage, degeneration, and apoptosis; and 4) PBM suppresses neuroinflammation and oxidative stress. CONCLUSION: Our findings support our hypothesis that PBM could be an effective method to alleviate depression and anxiety during the early stage of AD development. The mechanism underlying these beneficial effects may be due to the improvement of mitochondria function and integrity and the inhibition of neuroinflammation and oxidative stress.


Assuntos
Ansiedade/prevenção & controle , Depressão/prevenção & controle , Terapia com Luz de Baixa Intensidade , Ratos Transgênicos , Doença de Alzheimer/radioterapia , Animais , Apoptose , Modelos Animais de Doenças , Humanos , Masculino , Aprendizagem em Labirinto , Mitocôndrias/efeitos da radiação , Neurônios/efeitos da radiação , Estresse Oxidativo/efeitos da radiação , Ratos
6.
J Photochem Photobiol B ; 221: 112207, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34119804

RESUMO

Transcranial photobiomodulation (tPBM) is the process of delivering light photons through the skull to benefit from its modifying effect. Brain disorders are important health problems. The aim of this review was to determine the existing evidence of effectiveness, useful parameters, and safety of tPBM in the management of traumatic brain injury, stroke, Parkinson, and Alzheimer's disease as the common brain disorders. Four online databases, including Cochrane, Pub Med, Embase, and Google scholar were searched according to the Preferred Reporting Items for Systematic Reviews and meta-analyses (PRISMA) guidelines. 4728 articles were obtained in the initial search. Only those articles that were published until September 2020 and designed as randomized clinical trials (RCTs) or animal-controlled studies were included. 6 RCTs, 2 related supplementary articles, and 38 controlled animal studies met the inclusion criteria of this study. No RCTs were performed in the fields of Alzheimer's and Parkinson's diseases. The human RCTs and animal studies reported no adverse events resulted from the use of tPBM. Useful parameters of tPBM were identified according to the controlled animal studies. Since the investigated RCTs had no homogenous results, making an evidence-based decision for definite therapeutic application of tPBM is still unattainable. Altogether, these data support the need for large confirmatory well-designed RCTs for using tPBM as a novel, safe, and easy-to-administer treatment of brain disorders. EVIDENCE BEFORE THIS STUDY: High prevalence and complications of brain disorders and also side effects of neuropsychiatric medications have encouraged researchers to find alternative therapeutic techniques which tPBM can be one of them. In present review we tried to determine the existing evidence of effectiveness, useful parameters, and safety of tPBM in the management of traumatic brain injury, stroke, Alzheimer, and Parkinson's disease as common brain disorders. Four online databases, including "Cochrane", "Pub Med", "Embase", and "Google scholar" were searched. Only those articles that were published until September 2020 and designed as RCTs or animal-controlled studies were included. Search keywords were the followings: transcranial photobiomodulation" OR "transcranial low-level laser therapy" AND "stroke" OR "traumatic brain injury" OR "Alzheimer" OR "Parkinson". Several studies have confirmed effectiveness of tPBM in treatment of different brain disorders but the level of evidence of its effectiveness remain to be determined. ADDED VALUE OF THIS STUDY: In this study we systematically reviewed human RCTs to determine the existing evidence of tPBM effectiveness in management of four mentioned brain disorders. Since the outcomes of the reviewed RCTs were not homogeneous, further well-designed RCTs are required to decide more definitively on the evidence of this noninvasive and probably safe therapeutic intervention. We hypothesized that non-homogeneous outcomes could be due to inefficiency of PBM parameters. Controlled animal studies have the advantage of using objective tests to evaluate the results and compare them with the control group. We determined useful tPBM parameters based on these studies. IMPLICATIONS OF ALL THE AVAILABLE EVIDENCE: This research is part of our main project of tinnitus treatment using photobiomodulation (PBM). Evidence of central nervous system involvement in tinnitus led us to believe that treatment protocol of tinnitus should also include transcranial PBM. The determined useful parameters can be helpful in designing more efficient tPBM protocols in the management of brain disorders and tinnitus as a common debilitating symptom that can be associated with these disorders.


Assuntos
Encefalopatias/radioterapia , Terapia com Luz de Baixa Intensidade , Doença de Alzheimer/radioterapia , Animais , Lesões Encefálicas Traumáticas/radioterapia , Modelos Animais de Doenças , Humanos , Doença de Parkinson/radioterapia , Ensaios Clínicos Controlados Aleatórios como Assunto
7.
J Photochem Photobiol B ; 216: 112152, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33610085

RESUMO

BACKGROUND: Emerging evidence suggests that the gut microbiota plays an important role in the pathological progression of Alzheimer's disease (AD). Photobiomodulation (PBM) therapy is believed to have a positive regulatory effect on the imbalance of certain body functions, including inflammation, immunity, wound healing, nerve repair, and pain. Previous studies have found that the intestinal flora of patients with AD is in an unbalanced state. Therefore, we have proposed the use of gut flora-targeted PBM (gf-targeted PBM) as a method to improve AD in an Aß-induced AD mouse model. METHODS: PBM was performed on the abdomen of the mice at the wavelengths of 630 nm, 730 nm, and 850 nm at 100 J/cm2 for 8 weeks. Morris water maze test, immunofluorescence and proteomic of hippocampus, and intestinal flora detection of fecal were used to evaluate the treatment effects of gf-targeted PBM on AD rats. RESULTS: PBM at all three wavelengths (especially 630 nm and 730 nm) significantly improved learning retention as measured by the Morris water maze. In addition, we found reduced amyloidosis and tau phosphorylation in the hippocampus by immunofluorescence in AD mice. By using a quantitative proteomic analysis of the hippocampus, we found that gf-targeted PBM significantly altered the expression levels of 509 proteins (the same differentially expressed proteins in all three wavelengths of PBM), which involved the pathways of hormone synthesis, phagocytosis, and metabolism. The 16 s rRNA gene sequencing of fecal contents showed that PBM significantly altered the diversity and abundance of intestinal flora. Specifically, PBM treatment reversed the typical increase of Helicobacter and uncultured Bacteroidales and the decrease of Rikenella seen in AD mice. CONCLUSIONS: Our data indicate that gf-targeted PBM regulates the diversity of intestinal flora, which may improve damage caused by AD. Gf-targeted PBM has the potential to be a noninvasive microflora regulation method for AD patients.


Assuntos
Doença de Alzheimer , Microbioma Gastrointestinal , Terapia com Luz de Baixa Intensidade , Animais , Humanos , Masculino , Camundongos , Ratos , Doença de Alzheimer/radioterapia , Amiloidose/radioterapia , Modelos Animais de Doenças , Microbioma Gastrointestinal/efeitos da radiação , Hipocampo/efeitos da radiação , Terapia com Luz de Baixa Intensidade/métodos , Teste do Labirinto Aquático de Morris/efeitos da radiação , Fosforilação/efeitos da radiação , Proteômica , Proteínas tau/metabolismo
8.
Photodiagnosis Photodyn Ther ; 30: 101777, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32305654

RESUMO

Alzheimer's Disease (AD) is a neurodegenerative progressive disorder for which there is currently no cure. Recent research demonstrates a robust correlation between type-2 diabetes mellitus (T2DM) and the development of MCI and AD, now referred to as type-3 diabetes. Both AD and T2DM, as metabolic pathologies, can be traced to the level of mitochondrial function. The metabolic hypothesis suggests that the cause of AD might be rooted in mitochondrial dysfunction accompanied by fuel shortage in the brain. Although glucose is known to be the deferred source of fuel for cells, ketone bodies have been observed to provide metabolically compromised brain cells with an alternative fuel source, bypassing deficiencies in GLUT transport due to increased insulin resistance. By keeping glucose and insulin levels low to allow for the production of ketones, there is evidence that mitochondrial function will be restored and cognition/memory improved. Further, visible red or near-infrared (NIR) light has been shown to heal and stimulate damaged tissue by interacting with the mitochondria to restore function. This case study evaluates the effects of a 10-week clinically prescribed ketogenic nutrition protocol combined with transcranial photobiomodulation (PBM) with a 59-year-old male, heterozygous ApoE4 carrier, with a dual diagnosis of mild AD and an 11 year history of insulin dependent type 2 diabetes (T2DM). Statistically significant results reflect an 83% reduction in HOMA-IR; 64% decrease in the triglyceride/HDL ratio; HgA1c reduction from 9.44% to 6.4%; 57% decrease in VLDL and triglycerides; and normalized cognition as measured via the MoCA (Montreal Cognitive Assessment), 26/30 post intervention.


Assuntos
Doença de Alzheimer/terapia , Apolipoproteína E4/metabolismo , Diabetes Mellitus Tipo 2/terapia , Terapia com Luz de Baixa Intensidade/métodos , Doença de Alzheimer/complicações , Doença de Alzheimer/dietoterapia , Doença de Alzheimer/radioterapia , Glicemia , Disfunção Cognitiva , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/dietoterapia , Diabetes Mellitus Tipo 2/radioterapia , Dieta Cetogênica , Hemoglobinas Glicadas , Humanos , Cetose , Lipídeos/sangue , Masculino , Testes de Estado Mental e Demência , Pessoa de Meia-Idade
9.
J Gerontol A Biol Sci Med Sci ; 75(4): 631-639, 2020 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-30346494

RESUMO

Photobiomodulation using low-level light-emitting diode can be rapidly applied in neurological and physiological disorders safely and noninvasively. Photobiomodulation is effective for chronic diseases because of fewer side effects than drugs. Here we investigated the effects of photobiomodulation using light-emitting diode on amyloid plaques, gliosis, and neuronal loss to prevent and/or recover cognitive impairment, and optimal timing of photobiomodulation initiation for recovering cognitive function in a mouse model of Alzheimer's disease. 5XFAD mice were used as an Alzheimer's disease model. Animals receiving photobiomodulation treatment were divided into two groups: an early group starting photobiomodulation at 2 months of age (5XFAD+Early), and a late group starting photobiomodulation at 6 months of age (5XFAD+Delay). Both groups received photobiomodulation 20 minutes per session three times per week for 14 weeks. The Morris water maze, passive avoidance, and elevated plus maze tests were performed at 10 months of age. Immunohistochemistry and Western blot were performed after behavioral evaluation. The results showed that photobiomodulation treatment at early stages reduced amyloid accumulation, neuronal loss, and microgliosis and alleviated the cognitive dysfunction in 5XFAD mice, possibly by increasing insulin degrading enzyme related to amyloid-beta degradation. Photobiomodulation may be an excellent candidate for advanced preclinical Alzheimer's disease research.


Assuntos
Doença de Alzheimer/radioterapia , Terapia com Luz de Baixa Intensidade , Fatores Etários , Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Aprendizagem da Esquiva/efeitos da radiação , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/efeitos da radiação , Cognição/efeitos da radiação , Modelos Animais de Doenças , Gliose/patologia , Gliose/prevenção & controle , Humanos , Lasers Semicondutores/uso terapêutico , Masculino , Aprendizagem em Labirinto/efeitos da radiação , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Microglia/efeitos da radiação , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Proteólise/efeitos da radiação
10.
Photobiomodul Photomed Laser Surg ; 37(3): 159-167, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31050946

RESUMO

Objective: We present a case report of reversal of cognitive impairment, olfactory dysfunction, and quality of life measures in a patient with cognitive decline after multi-modality photobiomodulation (PBM) therapy. Background: Transcranial and intranasal PBM has been introduced as a light-based therapeutic technique in which exposure to low levels of red to near-infrared (NIR) light stimulates neuronal function, leading to beneficial neurological effects. Materials and methods: Patient received twice-daily PBM therapy at home using three different wearable light-emitting diode (LED) devices. For the first week containing a mixture of continuous wave mode red (635 nm) and NIR (810 nm) LEDs, a prototype transcranial light helmet and a body pad were used. The body pad was placed on various areas on the lower back and the helmet was worn while seated. After the first week of treatment, an intranasal LED device, 10-Hz pulsed wave mode NIR (810 nm), was initiated in the left nostril twice daily. All three devices were applied simultaneously for an irradiation time of 25 min per session. Results: The patient showed a significant improvement in the Montreal Cognitive Assessment score from 18 to 24 and in the Working Memory Questionnaire score from 53 to 10. The cognitive enhancement was accompanied by reversal of olfactory dysfunction as measured by the Alberta Smell Test and peanut butter odor detection test. Quality-of-life measures improved and caregiver stress was reduced. No adverse effects were reported. Conclusions: PBM therapy may be a promising noninvasive approach for patients with neurodegenerative diseases.


Assuntos
Doença de Alzheimer/psicologia , Doença de Alzheimer/radioterapia , Disfunção Cognitiva/terapia , Terapia com Luz de Baixa Intensidade , Transtornos do Olfato/terapia , Qualidade de Vida , Doença de Alzheimer/complicações , Disfunção Cognitiva/etiologia , Feminino , Humanos , Pessoa de Meia-Idade , Transtornos do Olfato/etiologia
11.
Sci Rep ; 7(1): 15012, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29118388

RESUMO

Synaptic dysfunction due to the disrupting binding of amyloid beta (Aß) and tau oligomers is one of the earliest impairments in Alzheimer's Disease (AD), driving initial cognitive deficits and clinical manifestation. Consequently, there is ample consensus that preventing early synaptic dysfunction would be an effective therapeutic strategy for AD. With this goal in mind, we investigated the effect of a treatment of mice with near infrared (NIR) light on synaptic vulnerability to Aß oligomers. We found that Aß oligomer binding to CNS synaptosomes isolated from wild type (wt) mice treated with NIR light was significantly reduced and the resulting suppression of long term potentiation (LTP) by Aß oligomers was prevented. Similarly, APP transgenic mice treated with NIR showed a significant reduction of endogenous Aß at CNS synapses. We further found that these phenomena were accompanied by increased synaptic mitochondrial membrane potential in both wt and Tg2576 mice. This study provides evidence that NIR light can effectively reduce synaptic vulnerability to damaging Aß oligomers, thus furthering NIR light therapy as a viable treatment for AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Raios Infravermelhos , Ligação Proteica/efeitos da radiação , Sinapses/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/radioterapia , Peptídeos beta-Amiloides/toxicidade , Animais , Modelos Animais de Doenças , Feminino , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Potenciação de Longa Duração/efeitos da radiação , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sinaptossomos/metabolismo , Sinaptossomos/efeitos da radiação
12.
J Healthc Eng ; 2017: 2530270, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29065581

RESUMO

The aim of the present study was to investigate on the effects of a low-frequency pulsed electromagnetic field (LF-PEMF) in an experimental cell model of Alzheimer's disease (AD) to assess new therapies that counteract neurodegeneration. In recent scientific literature, it is documented that the deep brain stimulation via electromagnetic fields (EMFs) modulates the neurophysiological activity of the pathological circuits and produces clinical benefits in AD patients. EMFs are applied for tissue regeneration because of their ability to stimulate cell proliferation and immune functions via the HSP70 protein family. However, the effects of EMFs are still controversial and further investigations are required. Our results demonstrate the ability of our LF-PEMF to modulate gene expression in cell functions that are dysregulated in AD (i.e., BACE1) and that these effects can be modulated with different treatment conditions. Of relevance, we will focus on miRNAs regulating the pathways involved in brain degenerative disorders.


Assuntos
Doença de Alzheimer/radioterapia , Proliferação de Células/efeitos da radiação , Campos Eletromagnéticos , MicroRNAs/efeitos da radiação , Doença de Alzheimer/sangue , Humanos , Magnetoterapia , Modelos Biológicos
13.
Photomed Laser Surg ; 35(8): 432-441, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28186867

RESUMO

OBJECTIVE: This study investigated whether patients with mild to moderately severe dementia or possible Alzheimer's disease (AD) with Mini-Mental State Exam (MMSE) Baseline scores of 10-24 would improve when treated with near-infrared photobiomodulation (PBM) therapy. BACKGROUND: Animal studies have presented the potential of PBM for AD. Dysregulation of the brain's default mode network (DMN) has been associated with AD, presenting the DMN as an identifiable target for PBM. MATERIALS AND METHODS: The study used 810 nm, 10 Hz pulsed, light-emitting diode devices combining transcranial plus intranasal PBM to treat the cortical nodes of the DMN (bilateral mesial prefrontal cortex, precuneus/posterior cingulate cortex, angular gyrus, and hippocampus). Five patients with mild to moderately severe cognitive impairment were entered into 12 weeks of active treatment as well as a follow-up no-treatment, 4-week period. Patients were assessed with the MMSE and Alzheimer's Disease Assessment Scale (ADAS-cog) tests. The protocol involved weekly, in-clinic use of a transcranial-intranasal PBM device; and daily at-home use of an intranasal-only device. RESULTS: There was significant improvement after 12 weeks of PBM (MMSE, p < 0.003; ADAS-cog, p < 0.023). Increased function, better sleep, fewer angry outbursts, less anxiety, and wandering were reported post-PBM. There were no negative side effects. Precipitous declines were observed during the follow-up no-treatment, 4-week period. This is the first completed PBM case series to report significant, cognitive improvement in mild to moderately severe dementia and possible AD cases. CONCLUSIONS: Results suggest that larger, controlled studies are warranted. PBM shows potential for home treatment of patients with dementia and AD.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/radioterapia , Terapia com Luz de Baixa Intensidade/métodos , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Testes Neuropsicológicos , Prognóstico , Medição de Risco , Estudos de Amostragem , Índice de Gravidade de Doença , Resultado do Tratamento
14.
Neurobiol Aging ; 49: 165-182, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27815990

RESUMO

Beta amyloid (Aß) is well accepted to play a central role in the pathogenesis of Alzheimer's disease (AD). The present work evaluated the therapeutic effects of low-level laser irradiation (LLI) on Aß-induced neurotoxicity in rat hippocampus. Aß 1-42 was injected bilaterally to the hippocampus CA1 region of adult male rats, and 2-minute daily LLI treatment was applied transcranially after Aß injection for 5 consecutive days. LLI treatment suppressed Aß-induced hippocampal neurodegeneration and long-term spatial and recognition memory impairments. Molecular studies revealed that LLI treatment: (1) restored mitochondrial dynamics, by altering fission and fusion protein levels thereby suppressing Aß-induced extensive fragmentation; (2) suppressed Aß-induced collapse of mitochondrial membrane potential; (3) reduced oxidized mitochondrial DNA and excessive mitophagy; (4) facilitated mitochondrial homeostasis via modulation of the Bcl-2-associated X protein/B-cell lymphoma 2 ratio and of mitochondrial antioxidant expression; (5) promoted cytochrome c oxidase activity and adenosine triphosphate synthesis; (6) suppressed Aß-induced glucose-6-phosphate dehydrogenase and nicotinamide adenine dinucleotide phosphate oxidase activity; (7) enhanced the total antioxidant capacity of hippocampal CA1 neurons, whereas reduced the oxidative damage; and (8) suppressed Aß-induced reactive gliosis, inflammation, and tau hyperphosphorylation. Although development of AD treatments has focused on reducing cerebral Aß levels, by the time the clinical diagnosis of AD or mild cognitive impairment is made, the brain is likely to have already been exposed to years of elevated Aß levels with dire consequences for multiple cellular pathways. By alleviating a broad spectrum of Aß-induced pathology that includes mitochondrial dysfunction, oxidative stress, neuroinflammation, neuronal apoptosis, and tau pathology, LLI could represent a new promising therapeutic strategy for AD.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/radioterapia , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Região CA1 Hipocampal/metabolismo , Terapia com Luz de Baixa Intensidade , Dinâmica Mitocondrial , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Doença de Alzheimer/psicologia , Animais , Apoptose , Região CA1 Hipocampal/patologia , Inflamação , Lasers Semicondutores/uso terapêutico , Terapia com Luz de Baixa Intensidade/métodos , Masculino , Neurônios/patologia , Estresse Oxidativo , Ratos Sprague-Dawley , Reconhecimento Psicológico , Memória Espacial , Tauopatias/etiologia , Tauopatias/radioterapia
15.
Photomed Laser Surg ; 34(12): 627-630, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27294393

RESUMO

OBJECTIVE: This communication reviews the ability of low-level laser therapy (LLLT) to stimulate mesenchymal stem cells (MSCs) in autologous bone marrow (BM) to enhance the capacity of MSCs to infiltrate the brain, clear ß-amyloid, and improve cognition. BACKGROUND: We recently reported that LLLT applied to the BM enhanced the proliferation of MSCs and their mobilization toward the ischemic heart region, suggesting a possible application of this approach in regenerative medicine and neurodegenerative diseases. It was also shown that circulating monocytes can infiltrate the brain and reduce brain amyloid load in an Alzheimer's disease (AD) mouse model. METHODS AND RESULTS: MSCs from wild-type mice stimulated with LLLT demonstrated an increased ability to maturate toward a monocyte lineage and to increase phagocytosis of soluble Aß in vitro. Furthermore, weekly LLLT for 2 months to the BM, starting at 4 months of age (progressive stage of the disease in these 5XFAD transgenic male mice), improved memory and spatial learning, compared to a sham-treated AD mouse model. Histology revealed a significant reduction in Aß brain burden in the laser-treated mice compared to the nonlaser-treated ones. CONCLUSIONS: The application of LLLT to the BM is suggested as a therapeutic approach in progressive stages of AD, and its potential role in mediating MSC therapy in brain amyloidogenic disease is implied.


Assuntos
Doença de Alzheimer/radioterapia , Células da Medula Óssea/efeitos da radiação , Terapia com Luz de Baixa Intensidade , Doenças Neurodegenerativas/radioterapia , Animais , Progressão da Doença , Camundongos
16.
Neurotoxicology ; 53: 290-300, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26945731

RESUMO

Although numerous studies have reported the influence of extremely low frequency magnetic field (ELF-MF) exposure on human health, its effects on cognitive deficits in Alzheimer's disease (AD) have remained under debate. Moreover, the influence of ELF-MF on hyperphosphorylated tau, which is one of the most common pathological hallmarks of AD, has not been reported to date. Therefore, transgenic mice (3xTg) were used in the present study. 3xTg mice, which express an APP/PS1 mutation combined with a tau (P301L) mutation and that develop cognitive deficits at 6 months of age, were subjected to ELF-MF (50Hz, 500µT) exposure or sham exposure daily for 3 months. We discovered that ELF-MF exposure ameliorated cognitive deficits and increased synaptic proteins in 3xTg mice. The protective effects of ELF-MF exposure may have also been caused by the inhibition of apoptosis and/or decreased oxidative stress levels that were observed in the hippocampus tissues of treated mice. Furthermore, tau hyperphosphorylation was decreased in vivo because of ELF-MF exposure, and this decrease was induced by the inhibition of GSK3ß and CDK5 activities and activation of PP2Ac. We are the first to report that exposure to ELF-MF can attenuate tau phosphorylation. These findings suggest that ELF-MF exposure could act as a valid therapeutic strategy for ameliorating cognitive deficits and attenuating tau hyperphosphorylation in AD.


Assuntos
Doença de Alzheimer/complicações , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/terapia , Magnetoterapia/métodos , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/radioterapia , Precursor de Proteína beta-Amiloide/genética , Animais , Apoptose/genética , Apoptose/efeitos da radiação , Condicionamento Psicológico/fisiologia , Condicionamento Psicológico/efeitos da radiação , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/efeitos da radiação , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Aprendizagem em Labirinto/fisiologia , Aprendizagem em Labirinto/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Fosforilação/efeitos da radiação , Presenilina-1/genética , Espécies Reativas de Oxigênio/metabolismo , Proteínas tau/genética
17.
PLoS One ; 7(4): e35751, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22558216

RESUMO

Few studies have investigated physiologic and cognitive effects of "long-term" electromagnetic field (EMF) exposure in humans or animals. Our recent studies have provided initial insight into the long-term impact of adulthood EMF exposure (GSM, pulsed/modulated, 918 MHz, 0.25-1.05 W/kg) by showing 6+ months of daily EMF treatment protects against or reverses cognitive impairment in Alzheimer's transgenic (Tg) mice, while even having cognitive benefit to normal mice. Mechanistically, EMF-induced cognitive benefits involve suppression of brain ß-amyloid (Aß) aggregation/deposition in Tg mice and brain mitochondrial enhancement in both Tg and normal mice. The present study extends this work by showing that daily EMF treatment given to very old (21-27 month) Tg mice over a 2-month period reverses their very advanced brain Aß aggregation/deposition. These very old Tg mice and their normal littermates together showed an increase in general memory function in the Y-maze task, although not in more complex tasks. Measurement of both body and brain temperature at intervals during the 2-month EMF treatment, as well as in a separate group of Tg mice during a 12-day treatment period, revealed no appreciable increases in brain temperature (and no/slight increases in body temperature) during EMF "ON" periods. Thus, the neuropathologic/cognitive benefits of EMF treatment occur without brain hyperthermia. Finally, regional cerebral blood flow in cerebral cortex was determined to be reduced in both Tg and normal mice after 2 months of EMF treatment, most probably through cerebrovascular constriction induced by freed/disaggregated Aß (Tg mice) and slight body hyperthermia during "ON" periods. These results demonstrate that long-term EMF treatment can provide general cognitive benefit to very old Alzheimer's Tg mice and normal mice, as well as reversal of advanced Aß neuropathology in Tg mice without brain heating. Results further underscore the potential for EMF treatment against AD.


Assuntos
Doença de Alzheimer/radioterapia , Peptídeos beta-Amiloides/metabolismo , Circulação Cerebrovascular/efeitos da radiação , Magnetoterapia/métodos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Temperatura Corporal/efeitos da radiação , Cognição/efeitos da radiação , Modelos Animais de Doenças , Campos Eletromagnéticos , Humanos , Aprendizagem em Labirinto/efeitos da radiação , Memória/efeitos da radiação , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos da radiação , Desempenho Psicomotor/efeitos da radiação
18.
Photomed Laser Surg ; 30(1): 54-60, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22029866

RESUMO

OBJECTIVE: The aim of the present study is to present the results of in vitro experiments with possible relevance in the treatment of Alzheimer's disease (AD). BACKGROUND DATA: Despite intensive research efforts, there is no treatment for AD. One root cause of AD is the extra- and intracellular deposition of amyloid-beta (Aß) fibrils in the brain. Recently, it was shown that extracellular Aß can enter brain cells, resulting in neurotoxicity. METHODS: After internalization of Aß(42) into human neuroblastoma (SH-EP) cells, they were irradiated with moderately intense 670-nm laser light (1000 Wm(-2)) and/or treated with epigallocatechin gallate (EGCG). RESULTS: In irradiated cells, Aß(42) aggregate amounts were significantly lower than in nonirradiated cells. Likewise, in EGCG-treated cells, Aß(42) aggregate amounts were significantly lower than in non-EGCG-treated cells. Except for the cells simultaneously laden with Aß(42) and EGCG, there was a significant increase in cell numbers in response to laser irradiation. EGCG alone had no effect on cell proliferation. Laser irradiation significantly increased ATP levels in Aß(42)-free cells, when compared to nonirradiated cells. Laser-induced clearance of Aß(42) aggregates occurred at the expense of cellular ATP. CONCLUSIONS: Irradiation with moderate levels of 670-nm light and EGCG supplementation complementarily reduces Aß aggregates in SH-EP cells. Transcranial penetration of moderate levels of red to near-infrared (NIR) light has already been amply exploited in the treatment of patients with acute stroke; the blood-brain barrier (BBB) penetration of EGCG has been demonstrated in animals. We hope that our approach will inspire a practical therapy for AD.


Assuntos
Doença de Alzheimer/radioterapia , Amiloide/ultraestrutura , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/diagnóstico por imagem , Catequina/análogos & derivados , Terapia com Luz de Baixa Intensidade , Neuroblastoma/ultraestrutura , Trifosfato de Adenosina/metabolismo , Amiloide/efeitos dos fármacos , Amiloide/efeitos da radiação , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Catequina/uso terapêutico , Humanos , Microscopia de Fluorescência , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/radioterapia , Células Tumorais Cultivadas , Ultrassonografia
19.
Aging Clin Exp Res ; 21(4-5): 277-81, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19959915

RESUMO

BACKGROUND AND AIMS: Hypovitaminosis D and K due to malnutrition or sunlight deprivation, compensatory hyperparathyroidism, increased bone resorption, low bone mineral density (BMD), and an increased risk of falls may contribute to an increased risk of hip fractures in patients with Alzheimer's disease. The purpose of the present study was to clarify the efficacy of interventions against hip fractures in patients with Alzheimer's disease. METHODS: With respect to randomized controlled trials (RCTs) regarding Alzheimer's disease and hip fractures, the literature was searched with PubMed. RESULTS: Three RCTs were identified, and the relative risk (RR) and 95% confidence interval (CI) were calculated for individual RCTs. Exposure to sunlight with calcium supplementation, menatetrenone (vitamin K2) plus calcium and vitamin D supplementation, and risedronate plus calcium and vitamin D supplementation improved hypovitaminosis D and hyperparathyroidism, contributing to a reduction in bone resorption. Risedronate itself strongly decreased bone resorption. Menatetrenone also decreased the serum level of undercarboxylated osteocalcin. The three interventions increased metacarpal BMD and reduced the incidence of hip fractures. The respective RRs (95% CI) were 0.22 (0.049-0.999), 0.13 (0.031-0.554), and 0.26 (0.100- 0.690). CONCLUSIONS: The present study clarified the efficacy of three interventions, including exposure to sunlight, menatetrenone, and risedronate with calcium and/or vitamin D supplementation against hip fractures in patients with Alzheimer's disease.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/radioterapia , Conservadores da Densidade Óssea/uso terapêutico , Densidade Óssea/efeitos dos fármacos , Ácido Etidrônico/análogos & derivados , Fraturas do Quadril/prevenção & controle , Luz Solar , Idoso , Cálcio/uso terapêutico , Transtornos Cognitivos/etiologia , Suplementos Nutricionais , Ácido Etidrônico/uso terapêutico , Feminino , Hemostáticos/uso terapêutico , Fraturas do Quadril/epidemiologia , Humanos , Masculino , Ensaios Clínicos Controlados Aleatórios como Assunto , Ácido Risedrônico , Vitamina D/uso terapêutico , Vitamina K 2/análogos & derivados , Vitamina K 2/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA