Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Orthop Traumatol ; 25(1): 17, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38622334

RESUMEN

BACKGROUND: Capacitively coupling electric fields (CCEF) is a method of non-invasive biophysical stimulation that enhances fracture repair and spinal fusion. This multicentre randomized controlled trial aimed to further examine the roles of CCEF in (1) the resolution of vertebral bone marrow oedema (VBME) using a follow-up MRI study and (2) pain relief, analgesic drug consumption and quality of life improvement in stimulated patients who were referred with acute vertebral fragility fractures (VFFs) compared to non-stimulated patients. METHODS: Between September 2016 and December 2019, patients who were referred to the spine centres that participated in this multicentre randomized clinical study with acute VFFs of type OF1 or OF2 were included in the present study. All the VFFs were conservatively managed according to Good Clinical Practice. Moreover, the patients were randomized into two groups: the CCEF group received, as an adjunct to the clinical study protocol, biophysical stimulation with a CCEF device (Osteospine, IGEA) for 8 h per day for 60 days, whereas the control group was treated according to the clinical study protocol. At baseline (T0), the 30-day follow-up (T1), the 60-day follow-up (T2), and the 6-month follow-up (T3), each patient underwent clinical evaluation using the Visual Analogue Scale (VAS) for Pain and the Oswestry Disability Index (ODI). Analgesic therapy with paracetamol 1000 mg tablets for 7 days-or longer, depending on the pain intensity-was performed; patients were required to report their paracetamol consumption on a specific sheet between study day 8 to 180 days of follow-up. MRI studies of the thoracolumbar spine were performed at 0 (T0), 30 (T1) and 60 days of follow-up (T2) using a 1.5-T MRI system in all of the centres that took part in the study. For each VBME area examined via MRI, the vertebral body geometry (i.e. anterior wall height/posterior wall height and vertebral kyphosis) were assessed. RESULTS: A total of 66 patients (male: 9, 13.63%; mean age: 73.15 years old) with 69 VFFs were included in the present study and randomized as follows: 33 patients were included in the control group and the remaining 33 patients were randomized into the CCEF group. In the CCEF group, good compliance with CCEF therapy was observed (adherence = 94%), and no adverse effects were recorded. In the stimulated patients, faster VBME resolution and significantly less vertebral body collapse during follow-up were observed compared to the control patients. Moreover, in the active group, faster pain reduction and improvement in the ODI mean score were observed. Stimulated patients also reported a significantly lower paracetamol consumption rate from the third follow-up after treatment until the 6-month follow-up. In terms of sex-related differences, in the CCEF group, VBME showed a faster resolution in male patients compared with females. CONCLUSION: Biophysical stimulation with CCEF, as an adjunct to traditional conservative treatment, is a useful tool to hasten the VBME resolution process and prevent vertebral body deformation. These MRI findings also correlate with faster back pain resolution and quality of life improvement. From the third follow-up after treatment until the 6-month follow-up, stimulated patients reported a significantly lower paracetamol consumption than control patients, even though back pain and quality of life showed no significant differences between the two groups. LEVEL OF EVIDENCE: II. Trial Registration Register: ClinicalTrials.gov, number: NCT05803681.


Asunto(s)
Fracturas por Compresión , Fracturas de la Columna Vertebral , Femenino , Humanos , Masculino , Anciano , Acetaminofén , Calidad de Vida , Estudios Prospectivos , Dolor de Espalda , Fracturas de la Columna Vertebral/diagnóstico por imagen , Fracturas de la Columna Vertebral/terapia , Analgésicos , Fracturas por Compresión/terapia , Resultado del Tratamiento
2.
Medicina (Kaunas) ; 57(11)2021 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-34833424

RESUMEN

Osteoarthritis (OA) is the most prevalent degenerative joint disease and the main cause of pain and disability in elderly people. OA currently represents a significant social health problem, since it affects 250 million individuals worldwide, mainly adults aged over 65. Although OA is a multifactorial disease, depending on both genetic and environmental factors, it is reported that joint degeneration has a higher prevalence in former athletes. Repetitive impact and loading, joint overuse and recurrent injuries followed by a rapid return to the sport might explain athletes' predisposition to joint articular degeneration. In recent years, however, big efforts have been made to improve the prevention and management of sports injuries and to speed up the athletes' return-to-sport. Biophysics is the study of biological processes and systems using physics-based methods or based on physical principles. Clinical biophysics has recently evolved as a medical branch that investigates the relationship between the human body and non-ionizing physical energy. A physical stimulus triggers a biological response by regulating specific intracellular pathways, thus acting as a drug. Preclinical and clinical trials have shown positive effects of biophysical stimulation on articular cartilage, subchondral bone and synovia. This review aims to assess the role of pulsed electromagnetic fields (PEMFs) and extracorporeal shockwave therapy (ESWT) in the prevention and treatment of joint degeneration in athletes.


Asunto(s)
Cartílago Articular , Anciano , Atletas , Biofisica , Campos Electromagnéticos , Humanos
3.
Cytokine ; 125: 154777, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31400640

RESUMEN

Literature studies suggest important protective effects of low-frequency, low-energy pulsed electromagnetic fields (PEMFs) on inflammatory pathways affecting joint and cerebral diseases. However, it is not clear on which bases they affect neuroprotection and the mechanism responsible is yet unknown. Therefore the aim of this study was to identify the molecular targets of PEMFs anti-neuroinflammatory action. The effects of PEMF exposure in cytokine production by lipopolysaccharide (LPS)-activated N9 microglial cells as well as the pathways involved, including adenylyl cyclase (AC), phospholipase C (PLC), protein kinase C epsilon (PKC-ε) and delta (PKC-δ), p38, ERK1/2, JNK1/2 mitogen activated protein kinases (MAPK), Akt and caspase 1, were investigated. In addition, the ability of PEMFs to modulate ROS generation, cell invasion and phagocytosis, was addressed. PEMFs reduced the LPS-increased production of TNF-α and IL-1ß in N9 cells, through a pathway involving JNK1/2. Furthermore, they decreased the LPS-induced release of IL-6, by a mechanism not dependent on AC, PLC, PKC-ε, PKC-δ, p38, ERK1/2, JNK1/2, Akt and caspase 1. Importantly, a significant effect of PEMFs in the reduction of crucial cell functions specific of microglia like ROS generation, cell invasion and phagocytosis was found. PEMFs inhibit neuroinflammation in N9 cells through a mechanism involving, at least in part, the activation of JNK MAPK signalling pathway and may be relevant to treat a variety of diseases characterized by neuroinflammation.


Asunto(s)
Inflamación/metabolismo , Interleucina-1beta/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Microglía/efectos de la radiación , Factor de Necrosis Tumoral alfa/metabolismo , Inhibidores de Adenilato Ciclasa/farmacología , Adenilil Ciclasas/metabolismo , Animales , Caspasa 1/metabolismo , Línea Celular , Citocinas/metabolismo , Campos Electromagnéticos , Interleucina-6/metabolismo , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Lipopolisacáridos/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Microglía/efectos de los fármacos , Microglía/enzimología , Microglía/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fagocitosis/efectos de los fármacos , Fagocitosis/efectos de la radiación , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Biotechnol Bioeng ; 117(5): 1584-1596, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31985051

RESUMEN

Articular cartilage injuries are a common source of joint pain and dysfunction. We hypothesized that pulsed electromagnetic fields (PEMFs) would improve growth and healing of tissue-engineered cartilage grafts in a direction-dependent manner. PEMF stimulation of engineered cartilage constructs was first evaluated in vitro using passaged adult canine chondrocytes embedded in an agarose hydrogel scaffold. PEMF coils oriented parallel to the articular surface induced superior repair stiffness compared to both perpendicular PEMF (p = .026) and control (p = .012). This was correlated with increased glycosaminoglycan deposition in both parallel and perpendicular PEMF orientations compared to control (p = .010 and .028, respectively). Following in vitro optimization, the potential clinical translation of PEMF was evaluated in a preliminary in vivo preclinical adult canine model. Engineered osteochondral constructs (∅ 6 mm × 6 mm thick, devitalized bone base) were cultured to maturity and implanted into focal defects created in the stifle (knee) joint. To assess expedited early repair, animals were assessed after a 3-month recovery period, with microfracture repairs serving as an additional clinical control. In vivo, PEMF led to a greater likelihood of normal chondrocyte (odds ratio [OR]: 2.5, p = .051) and proteoglycan (OR: 5.0, p = .013) histological scores in engineered constructs. Interestingly, engineered constructs outperformed microfracture in clinical scoring, regardless of PEMF treatment (p < .05). Overall, the studies provided evidence that PEMF stimulation enhanced engineered cartilage growth and repair, demonstrating a potential low-cost, low-risk, noninvasive treatment modality for expediting early cartilage repair.


Asunto(s)
Cartílago Articular/efectos de la radiación , Campos Electromagnéticos , Ingeniería de Tejidos/métodos , Cicatrización de Heridas/efectos de los fármacos , Animales , Cartílago Articular/lesiones , Células Cultivadas , Condrocitos/efectos de la radiación , Perros , Masculino , Rodilla de Cuadrúpedos/lesiones
5.
Int J Mol Sci ; 21(21)2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33126773

RESUMEN

Pulsed electromagnetic fields (PEMFs) are emerging as an innovative, non-invasive therapeutic option in different pathological conditions of the central nervous system, including cerebral ischemia. This study aimed to investigate the mechanism of action of PEMFs in an in vitro model of human astrocytes, which play a key role in the events that occur following ischemia. 1321N1 cells were exposed to PEMFs or hypoxic conditions and the release of relevant neurotrophic and angiogenic factors, such as VEGF, EPO, and TGF-ß1, was evaluated by means of ELISA or AlphaLISA assays. The involvement of the transcription factor HIF-1α was studied by using the specific inhibitor chetomin and its expression was measured by flow cytometry. PEMF exposure induced a time-dependent, HIF-1α-independent release of VEGF from 1321N1 cells. Astrocyte conditioned medium derived from PEMF-exposed astrocytes significantly reduced the oxygen-glucose deprivation-induced cell proliferation and viability decrease in the neuron-like cells SH-SY5Y. These findings contribute to our understanding of PEMFs action in neuropathological conditions and further corroborate their therapeutic potential in cerebral ischemia.


Asunto(s)
Astrocitos/citología , Campos Electromagnéticos , Glucosa/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neuroblastoma/prevención & control , Oxígeno/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Astrocitos/metabolismo , Astrocitos/efectos de la radiación , Hipoxia de la Célula , Supervivencia Celular , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neuroblastoma/etiología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Sustancias Protectoras , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética
6.
J Cell Physiol ; 234(9): 15089-15097, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30656694

RESUMEN

Low-energy low-frequency pulsed electromagnetic fields (PEMFs) exert several protective effects, such as the regulation of kinases, transcription factors as well as cell viability in both central and peripheral biological systems. However, it is not clear on which bases they affect neuroprotection and the mechanism responsible is yet unknown. In this study, we have characterized in nerve growth factor-differentiated pheochromocytoma PC12 cells injured with hypoxia: (i) the effects of PEMF exposure on cell vitality; (ii) the protective pathways activated by PEMFs to relief neuronal cell death, including adenylyl cyclase, phospholipase C, protein kinase C epsilon and delta, p38, ERK1/2, JNK1/2 mitogen-activated protein kinases, Akt and caspase-3; (iii) the regulation by PEMFs of prosurvival heat-shock proteins of 70 (HSP70), cAMP response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF), and Bcl-2 family proteins. The results obtained in this study show a protective effect of PEMFs that are able to reduce neuronal cell death induced by hypoxia by modulating p38, HSP70, CREB, BDNF, and Bcl-2 family proteins. Specifically, we found a rapid activation (30 min) of p38 kinase cascade, which in turns enrolles HSP70 survival chaperone molecule, resulting in a significant CREB phosphorylation increase (24 hr). In this cascade, later (48 hr), BDNF and the antiapoptotic pathway regulated by the Bcl-2 family of proteins are recruited by PEMFs to enhance neuronal survival. This study paves the way to elucidate the mechanisms triggered by PEMFs to act as a new neuroprotective approach to treat cerebral ischemia by reducing neuronal cell death.

7.
Int Orthop ; 43(3): 539-551, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30645684

RESUMEN

INTRODUCTION: Biophysical stimulation is a non-invasive therapy used in orthopaedic practice to increase and enhance reparative and anabolic activities of tissue. METHODS: A sistematic web-based search for papers was conducted using the following titles: (1) pulsed electromagnetic field (PEMF), capacitively coupled electrical field (CCEF), low intensity pulsed ultrasound system (LIPUS) and biophysical stimulation; (2) bone cells, bone tissue, fracture, non-union, prosthesis and vertebral fracture; and (3) chondrocyte, synoviocytes, joint chondroprotection, arthroscopy and knee arthroplasty. RESULTS: Pre-clinical studies have shown that the site of interaction of biophysical stimuli is the cell membrane. Its effect on bone tissue is to increase proliferation, synthesis and release of growth factors. On articular cells, it creates a strong A2A and A3 adenosine-agonist effect inducing an anti-inflammatory and chondroprotective result. In treated animals, it has been shown that the mineralisation rate of newly formed bone is almost doubled, the progression of the osteoarthritic cartilage degeneration is inhibited and quality of cartilage is preserved. Biophysical stimulation has been used in the clinical setting to promote the healing of fractures and non-unions. It has been successfully used on joint pathologies for its beneficial effect on improving function in early OA and after knee surgery to limit the inflammation of periarticular tissues. DISCUSSION: The pooled result of the studies in this review revealed the efficacy of biophysical stimulation for bone healing and joint chondroprotection based on proven methodological quality. CONCLUSION: The orthopaedic community has played a central role in the development and understanding of the importance of the physical stimuli. Biophysical stimulation requires care and precision in use if it is to ensure the success expected of it by physicians and patients.


Asunto(s)
Enfermedades Óseas/terapia , Enfermedades de los Cartílagos/terapia , Terapia por Estimulación Eléctrica/métodos , Fracturas Óseas/terapia , Magnetoterapia/métodos , Animales , Enfermedades Óseas/metabolismo , Enfermedades Óseas/patología , Regeneración Ósea/fisiología , Regeneración Ósea/efectos de la radiación , Huesos/metabolismo , Huesos/patología , Huesos/efectos de la radiación , Cartílago/metabolismo , Cartílago/patología , Cartílago/efectos de la radiación , Enfermedades de los Cartílagos/metabolismo , Enfermedades de los Cartílagos/patología , Condrocitos/metabolismo , Condrocitos/patología , Condrocitos/efectos de la radiación , Terapia por Estimulación Eléctrica/tendencias , Fracturas Óseas/metabolismo , Fracturas Óseas/patología , Humanos , Magnetoterapia/tendencias
8.
Int J Mol Sci ; 20(7)2019 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-30925808

RESUMEN

The roles of low-intensity pulsed ultrasound (LIPUS) and microRNAs (miRNAs) on hMSCs commitments have already been investigated; however, the effects of the application of their co-treatments in an in vitro cell model are still unknown. Our previous studies demonstrated that (i) LIPUS modulated hMSCs cytoskeletal organization and (ii) miRNA-675-5p have a role in HIF-1α signaling modulation during hMSCs osteoblast commitment. We investigated for the first time the role of LIPUS as promoter tool for miRNA expression. Thanks to bioinformatic analysis, we identified miR-31-5p as a LIPUS-induced miRNA and investigated its role through in vitro studies of gain and loss of function. Results highlighted that LIPUS stimulation induced a hypoxia adaptive cell response, which determines a reorganization of cell membrane and cytoskeleton proteins. MiR-31-5p gain and loss of function studies, demonstrated as miR-31-5p overexpression, were able to induce hypoxic and cytoskeletal responses. Moreover, the co-treatments LIPUS and miR-31-5p inhibitor abolished the hypoxic responses including angiogenesis and the expression of Rho family proteins. MiR-31-5p was identified as a LIPUS-mechanosensitive miRNAs and may be considered a new therapeutic option to promote or abolish hypoxic response and cytoskeletal organization on hMSCs during the bone regeneration process.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Madre Mesenquimatosas/efectos de la radiación , MicroARNs/genética , Ondas Ultrasónicas , Regulación hacia Arriba/efectos de la radiación , Diferenciación Celular , Línea Celular , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología
9.
J Cell Physiol ; 233(2): 1558-1573, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28621452

RESUMEN

Low-intensity pulsed ultrasound (LIPUS) as an adjuvant therapy in in vitro and in vivo bone engineering has proven to be extremely useful. The present study aimed at investigating the effect of 30 mW/cm2 LIPUS stimulation on commercially available human mesenchymal stem cells (hMSCs) cultured in basal or osteogenic medium at different experimental time points (7, 14, 21 days). The hypothesis was that LIPUS would improve the osteogenic differentiation of hMSC and guarantying the maintenance of osteogenic committed fraction, as demonstrated by cell vitality and proteomic analysis. LIPUS stimulation (a) regulated the balance between osteoblast commitment and differentiation by specific networks (activations of RhoA/ROCK signaling and upregulation of Ribosome constituent/Protein metabolic process, Glycolysis/Gluconeogenesis, RNA metabolic process/Splicing and Tubulins); (b) allowed the maintenance of a few percentage of osteoblast precursors (21 days CD73+/CD90+: 6%; OCT-3/4+/NANOG+/SOX2+: 10%); (c) induced the activation of osteogenic specific pathways shown by gene expression (early: ALPL, COL1A1, late: RUNX2, BGLAP, MAPK1/6) and related protein release (COL1a1, OPN, OC), in particular in the presence of osteogenic soluble factors able to mimic bone microenvironment. To summarize, LIPUS might be able to improve the osteogenic commitment of hMSCs in vitro, and, at the same time, enhance their osteogenic differentiation.


Asunto(s)
Diferenciación Celular/efectos de la radiación , Células Madre Mesenquimatosas/efectos de la radiación , Osteogénesis/efectos de la radiación , Ondas Ultrasónicas , Linaje de la Célula , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Fenotipo , Mapas de Interacción de Proteínas , Proteómica/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de la radiación , Nicho de Células Madre , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
J Cell Physiol ; 232(5): 1200-1208, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27639248

RESUMEN

In the present study, the effect of low-frequency, low-energy pulsed electromagnetic fields (PEMFs) has been investigated by using different cell lines derived from neuron-like cells and microglial cells. In particular, the primary aim was to evaluate the effect of PEMF exposure in inflammation- and hypoxia-induced injury in two different neuronal cell models, the human neuroblastoma-derived SH-SY5Y cells and rat pheochromocytoma PC12 cells and in N9 microglial cells. In neuron-like cells, live/dead and apoptosis assays were performed in hypoxia conditions from 2 to 48 h. Interestingly, PEMF exposure counteracted hypoxia damage significantly reducing cell death and apoptosis. In the same cell lines, PEMFs inhibited the activation of the hypoxia-inducible factor 1α (HIF-1α), the master transcriptional regulator of cellular response to hypoxia. The effect of PEMF exposure on reactive oxygen species (ROS) production in both neuron-like and microglial cells was investigated considering their key role in ischemic injury. PEMFs significantly decreased hypoxia-induced ROS generation in PC12, SH-SY5Y, and N9 cells after 24 or 48 h of incubation. Moreover, PEMFs were able to reduce some of the most well-known pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, IL-6, and IL-8 release in N9 microglial cells stimulated with different concentrations of LPS for 24 or 48 h of incubation time. These results show a protective effect of PEMFs on hypoxia damage in neuron-like cells and an anti-inflammatory effect in microglial cells suggesting that PEMFs could represent a potential therapeutic approach in cerebral ischemic conditions. J. Cell. Physiol. 232: 1200-1208, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Campos Electromagnéticos , Inflamación/patología , Microglía/patología , Neuronas/patología , Animales , Muerte Celular , Hipoxia de la Célula , Citocinas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos , Ratones , Microglía/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Mediators Inflamm ; 2017: 2740963, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28255202

RESUMEN

Several studies explored the biological effects of low frequency low energy pulsed electromagnetic fields (PEMFs) on human body reporting different functional changes. Much research activity has focused on the mechanisms of interaction between PEMFs and membrane receptors such as the involvement of adenosine receptors (ARs). In particular, PEMF exposure mediates a significant upregulation of A2A and A3ARs expressed in various cells or tissues involving a reduction in most of the proinflammatory cytokines. Of particular interest is the observation that PEMFs, acting as modulators of adenosine, are able to increase the functionality of the endogenous agonist. By reviewing the scientific literature on joint cells, a double role for PEMFs could be hypothesized in vitro by stimulating cell proliferation, colonization of the scaffold, and production of tissue matrix. Another effect could be obtained in vivo after surgical implantation of the construct by favoring the anabolic activities of the implanted cells and surrounding tissues and protecting the construct from the catabolic effects of the inflammatory status. Moreover, a protective involvement of PEMFs on hypoxia damage in neuron-like cells and an anti-inflammatory effect in microglial cells have suggested the hypothesis of a positive impact of this noninvasive biophysical stimulus.


Asunto(s)
Campos Electromagnéticos , Receptores Purinérgicos P1/metabolismo , Animales , Antiinflamatorios/metabolismo , Sistema Nervioso Central/metabolismo , Humanos , Transducción de Señal
12.
BMC Musculoskelet Disord ; 16: 308, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26480822

RESUMEN

BACKGROUND: Osteoarthritis (OA) is the final result of progressive alterations to articular cartilage structure, composition and cellularity, followed by an increase in the concentration of pro-inflammatory cytokines in joint synovial fluid. Even though the effect of pulsed electromagnetic field (PEMF) stimulation in counteracting OA progression and inflammation is of increasing interest, because of its anabolic and anti-inflammatory properties, the present study aimed to improve the knowledge on cartilage extracellular matrix (ECM) and chondrocyte changes related to the exposure of PEMF, from a histological and histomorphometric point of view. METHODS: An in vitro OA model was realized, culturing bovine cartilage explants with a high dose of interleukin 1ß (IL1ß, 50 ng/ml) at different experimental times (24 h, and 7 and 21 days). The effects of PEMFs (75 Hz, 1.5 mT) were evaluated in cartilage explants treated with IL1ß or not (control), in terms of cartilage structure, cellularity and proteoglycans, glycosaminoglycans, collagen II and transforming growth factor ß1 synthesis by using histology, histomorphometry and immunohistochemistry. RESULTS: Making a comparison with control cartilage, IL1ß-treated explants showed a decrease in cartilage matrix, structure and cellularity parameters. PEMFs were able to counteract the progression of OA acting on both cartilage cellularity and ECM in cartilage previously treated with IL1ß. Normal distribution (Kolmogroc-Smirnov test) and homoscedasticity (Levene test) of data were verified, then, the non-parametric Kruskal Wallis test followed by Mann-Whiteny U test for pairwise comparisons were performed. The p-value was adjusted according to the Dunn-Sidak correction. CONCLUSIONS: These results, obtained by culturing and treating cartilage explants from two different joints, confirmed that PEMF stimulation can be used as adjuvant therapy to preserve cartilage from detrimental effects of high inflammatory cytokine levels during OA.


Asunto(s)
Magnetoterapia , Osteoartritis/terapia , Animales , Bovinos , Técnicas In Vitro , Interleucina-1beta
13.
BMC Musculoskelet Disord ; 16: 233, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26328626

RESUMEN

BACKGROUND: The study aimed to evaluate the combined effect of Pulsed Electromagnetic Field (PEMF) biophysical stimulation and bone marrow concentrate (BMC) in osteochondral defect healing in comparison to the treatment with scaffold alone. METHODS: An osteochondral lesion of both knees was performed in ten rabbits. One was treated with a collagen scaffold alone and the other with scaffold seeded with BMC. Half of the animals were stimulated by PEMFs (75 Hz, 1.5 mT, 4 h/day) and at 40 d, macroscopic, histological and histomorphometric analyses were performed to evaluate osteochondral defect regeneration. RESULTS: Regarding cartilage, the addition of BMC to the scaffold improved cell parameters and the PEMF stimulation improved both cell and matrix parameters compared with scaffold alone. The combination of BMC and PEMFs further improved osteochondral regeneration: there was an improvement in macroscopic, cartilage cellularity and matrix parameters and a reduction in the percentage of cartilage under the tidemark. Epiphyseal bone healing improved in all the osteochondral defects regardless of treatment, although PEMFs alone did not significantly improve the reconstruction of subchondral bone in comparison to treatment with scaffold alone. CONCLUSIONS: Results show that BMC and PEMFs might have a separate effect on osteochondral regeneration, but it seems that they have a greater effect when used together. Biophysical stimulation is a non-invasive therapy, free from side effects and should be started soon after BMC transplantation to increase the quality of the regenerated tissue. However, because this is the first explorative study on the combination of a biological and a biophysical treatment for osteochondral regeneration, future preclinical and clinical research should be focused on this topic to explore mechanisms of action and the correct clinical translation.


Asunto(s)
Trasplante de Médula Ósea/métodos , Regeneración Ósea/fisiología , Cartílago Articular/patología , Colágeno/administración & dosificación , Magnetoterapia/métodos , Andamios del Tejido , Animales , Células de la Médula Ósea/fisiología , Regeneración Ósea/efectos de los fármacos , Campos Electromagnéticos , Masculino , Conejos
14.
Bioelectromagnetics ; 35(6): 426-36, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25099126

RESUMEN

Pulsed electromagnetic fields (PEMFs) play a regulatory role on osteoblast activity and are clinically beneficial during fracture healing. Human mesenchymal stem cells (MSCs) derived from different sources have been extensively used in bone tissue engineering. Compared with MSCs isolated from bone marrow (BMSCs), those derived from adipose tissue (ASCs) are easier to obtain and available in larger amounts, although they show a less osteogenic differentiation potential than BMSCs. The hypothesis tested in this study was to evaluate whether PEMFs favor osteogenic differentiation both in BMSCs and in ASCs and to compare the role of PEMFs alone and in combination with the biochemical osteogenic stimulus bone morphogenetic protein (BMP)-2. Early and later osteogenic markers, such as alkaline phosphatase (ALP) activity, osteocalcin levels, and matrix mineralization, were analyzed at different times during osteogenic differentiation. Results showed that PEMFs induced osteogenic differentiation by increasing ALP activity, osteocalcin, and matrix mineralization in both BMSCs and ASCs, suggesting that PEMF activity is maintained during the whole differentiation period. The addition of BMP-2 in PEMF exposed cultures further increased all the osteogenic markers in BMSCs, while in ASCs, the stimulatory role of PEMFs was independent of BMP-2. Our results indicate that PEMFs may stimulate an early osteogenic induction in both BMSCs and ASCs and they suggest PEMFs as a bioactive factor to enhance the osteogenesis of ASCs, which are an attractive cell source for clinical applications. In conclusion, PEMFs may be considered a possible tool to improve autologous cell-based regeneration of bone defects in orthopedics.


Asunto(s)
Tejido Adiposo/citología , Células de la Médula Ósea , Diferenciación Celular , Campos Electromagnéticos , Células Madre Mesenquimatosas/fisiología , Osteogénesis , Adulto , Fosfatasa Alcalina/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Calcificación Fisiológica , Células Cultivadas , Femenino , Humanos , Masculino , Osteocalcina/metabolismo , Periodicidad , Adulto Joven
15.
Int Orthop ; 38(2): 397-403, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24352823

RESUMEN

PURPOSE: It has been reported that even one year after total knee arthroplasty (TKA), a relevant percentage of patients does not attain complete recovery and indicate unfavourable long-term pain outcome. We compared the clinical outcome of 33 patients undergoing TKA randomly assigned to the control or the pulsed electromagnetic field group (I-ONE therapy). METHODS: I-ONE therapy was administered postoperatively four hours per day for 60 days. Patients were assessed before surgery and then at one, two and six months postoperatively using international scores. RESULTS: One month after TKA, pain, knee swelling and functional score were significantly better in the treated compared with the control group. Pain was still significantly lower in the treated group at the six month follow-up. Three years after surgery, severe pain and occasional walking limitations were reported in a significantly lower number of patients in the treated group. CONCLUSIONS: Advantages deriving from early control of joint inflammation may explain the maintenance of results at follow-up. I-ONE therapy should be considered an effective completion of the TKA procedure.


Asunto(s)
Artralgia/etiología , Artralgia/terapia , Artroplastia de Reemplazo de Rodilla/efectos adversos , Campos Electromagnéticos , Osteoartritis de la Rodilla/cirugía , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Encuestas Epidemiológicas , Humanos , Articulación de la Rodilla/diagnóstico por imagen , Articulación de la Rodilla/fisiología , Articulación de la Rodilla/cirugía , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Periodo Posoperatorio , Radiografía , Recuperación de la Función/fisiología , Resultado del Tratamiento
16.
J Orthop Traumatol ; 14(1): 51-7, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23179083

RESUMEN

BACKGROUND: A multicenter retrospective analysis of patients treated for leg fractures was conducted to develop a score that correlates with fracture healing time and to identify the risk gradient for delayed healing. METHODS: Fifty-three patients were analyzed and considered healed when full weight bearing was possible. Patients were divided into those who healed within 180 days and those who took longer to heal. Risk factors associated with delayed healing, fracture morphology, and orthopedic treatments were recorded. The available literature was used to weight the relative risk associated with each factor; values were combined into a score evaluating the risk of delayed healing: L-ARRCO (a literature-based score where the risk of delayed bone healing is calculated using a specific algorithm). Other risk factors associated with delayed healing were then considered in order to calculate a new score, ARRCO. Continuous variables were compared between groups using Student's heteroschedastic two-tail t test. Receiver operating characteristic (ROC) curves and the areas under the curves were calculated to determine the ability of this score to discriminate subjects with delayed healing. RESULTS: The mean L-ARRCO scores of the patients who healed within and after 180 days were significantly different (5.78 ± 1.59 and 7.05 ± 2.46, respectively). The mean ARRCO scores of the patients who healed within and after 180 days were also significantly different (5.92 ± 1.78 and 9.03 ± 2.79, respectively). However, the area under the ROC curve was significantly smaller for L-ARRCO than for ARRCO (0.62 ± 0.09 versus 0.82 ± 0.07). CONCLUSIONS: The ARRCO score is significantly associated with fracture healing time and could be used to identify "fractures at risk," allowing early intervention to stimulate osteogenesis.


Asunto(s)
Fracturas del Fémur/fisiopatología , Curación de Fractura , Fracturas de la Tibia/fisiopatología , Adulto , Fracturas del Fémur/epidemiología , Humanos , Persona de Mediana Edad , Curva ROC , Estudios Retrospectivos , Factores de Riesgo , Fracturas de la Tibia/epidemiología , Factores de Tiempo , Resultado del Tratamiento
17.
Bioelectromagnetics ; 33(4): 279-87, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22012856

RESUMEN

Different effects of pulsed electromagnetic field (PEMF) exposure on brain tissue have been described in pre-clinical models and in clinical settings. Nevertheless, the mechanism of action and the possible interaction with membrane receptors such as adenosine receptors (ARs) has not been investigated. The present study focused on the effect of PEMFs on A1 and A2A ARs in the rat cerebral cortex and cortical neurons. Affinity and density of ARs were evaluated by means of saturation binding experiments while mRNA expression was investigated through retro-transcription polymerase chain reaction (RT-PCR). PEMF treatment of the intact rat cerebral cortex or cortical neurons at 1.5 mT mediated a transient and significant increase in A2A ARs after 4 h (2.0-fold increase) and 6 h (1.4- and 1.8-fold increase, respectively) of exposure. In addition, PEMF treatment of the rat cerebral cortex and rat cortical neurons at 3 mT upregulated A2A ARs after 2 h (2.0- and 2.2-fold increase, respectively) and 4 h (1.6- and 1.9-fold increase, respectively). The treatment of rat cortex membranes with PEMFs at 1.5 and 3 mT induced an increase in A2A AR density after 2 h (1.9- and 2.2-fold increase, respectively) and was constant at all incubation times investigated. In rat cortical neurons, mRNA levels of A1 and A2A ARs were not affected by PEMF exposure for the times and intensities used. These results suggest that PEMF treatment has different biological effects in whole organs or cells in comparison with isolated membranes.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/efectos de la radiación , Campos Electromagnéticos/efectos adversos , Receptor de Adenosina A1/metabolismo , Receptores de Adenosina A2/metabolismo , Animales , Encéfalo/citología , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Regulación de la Expresión Génica/efectos de la radiación , Masculino , Neuronas/citología , Neuronas/metabolismo , Neuronas/efectos de la radiación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A1/genética , Receptores de Adenosina A2/genética
18.
BMC Musculoskelet Disord ; 13: 88, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22672794

RESUMEN

BACKGROUND: Total knee arthroplasty (TKA) is often associated with a severe local inflammatory reaction which, unless controlled, leads to persistent pain up to one year after surgery. Standard and accelerated rehabilitation protocols are currently being implemented after TKA, but no consensus exists regarding the long-term effects. Biophysical stimulation with pulsed electromagnetic fields (PEMFs) has been demonstrated to exert an anti-inflammatory effect, to promote early functional recovery and to maintain a positive long-term effect in patients undergoing joint arthroscopy. The aim of this study was to evaluate whether PEMFs can be used to limit the pain and enhance patient recovery after TKA. METHODS: A prospective, randomized, controlled study in 30 patients undergoing TKA was conducted. Patients were randomized into experimental PEMFs or a control group. Patients in the experimental group were instructed to use I-ONE stimulator 4hours/day for 60days. Postoperatively, all patients received the same rehabilitation program. Treatment outcome was assessed using the Knee Society Score, SF-36 Health-Survey and VAS. Patients were evaluated pre-operatively and one, two, six and 12 months after TKA. Joint swelling and Non Steroidal Anti Inflammatory Drug (NSAID) consumption were recorded. Comparisons between the two groups were carried out using a two-tail heteroschedastic Student's t-test. Analysis of variance for each individual subject during the study was performed using ANOVA for multiple comparisons, applied on each group, and a Dunnet post hoc test. A p value < 0.05 was considered statistically significant. RESULTS: Pre-operatively, no differences were observed between groups in terms of age, sex, weight, height, Knee-Score, VAS, SF-36 and joint swelling, with the exception of the Functional Score. The Knee-Score, SF-36 and VAS demonstrated significantly positive outcomes in the I-ONE stimulated group compared with the controls at follow-ups. In the I-ONE group, NSAID use was reduced and joint swelling resolution was more rapid than in controls. The effect of I-ONE therapy was maintained after use of the device was discontinued. CONCLUSIONS: The results of the study show early functional recovery in the I-ONE group. I-ONE therapy should be considered after TKA to prevent the inflammatory reaction elicited by surgery, for pain relief and to speed functional recovery.


Asunto(s)
Artroplastia de Reemplazo de Rodilla/efectos adversos , Inflamación/prevención & control , Articulación de la Rodilla/cirugía , Magnetoterapia , Dolor Postoperatorio/prevención & control , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Antiinflamatorios no Esteroideos/uso terapéutico , Distribución de Chi-Cuadrado , Evaluación de la Discapacidad , Diseño de Equipo , Femenino , Humanos , Inflamación/etiología , Inflamación/fisiopatología , Italia , Articulación de la Rodilla/fisiopatología , Magnetoterapia/instrumentación , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Dolor Postoperatorio/etiología , Dolor Postoperatorio/fisiopatología , Estudios Prospectivos , Recuperación de la Función , Encuestas y Cuestionarios , Factores de Tiempo , Resultado del Tratamiento
19.
J ISAKOS ; 7(5): 105-112, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35623611

RESUMEN

BACKGROUND: To assess pain relief and clinical outcomes in patients undergoing unicompartmental knee arthroplasty (UKA) stimulated with pulsed electromagnetic fields (PEMFs) compared to a control group. METHODS: A prospective randomised controlled trial (RCT) was performed in which 72 patients undergoing medial UKA were randomised into a control group or an experimental PEMFs group. The patients allocated to the experimental group were instructed to use PEMFs for 4 h per day for 60 days. They were evaluated before a surgery and then during the time points corresponding to 1 month, 2 months, 6 months, 12 months, and 36 months after the surgery. No placebo group was included in the RCT. Clinical assessment included the Visual Analogue Scale (VAS) for pain, Oxford Knee Score (OKS), the Short Form 36 (SF-36) health survey questionnaire, and joint swelling. During each follow-up visit, the consumption of Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) was recorded. RESULTS: The VAS decreased on follow-up visits in both the groups; a statistically significant difference between the groups was observed during the 6 (p = 0.0297), 12 (p = 0.0003), and 36 months (p = 0.0333) follow-ups in favour of the PEMFs group. One month after UKA, the percentages of patients using NSAIDs in the PEMFs and control group were 71% and 92%, respectively (p = 0.0320). At the 2 months point, 15% of the patients in the PEMFs group used NSAIDs compared to 39% in the control group (p = 0.0317). The objective knee girth evaluation showed a statistically significant difference at 6 (p = 0.0204), 12 (p = 0.0005), and 36 (p = 0.0005) months with improved values observed in the PEMFs group. The subjective assessment of the swelling demonstrated a statistically significant difference at 2 (p = 0.0073), 6 (p = 0.0006), 12 (p = 0.0001), and 36 (p = 0.0011) months with better values noted in the PEMFs group. Last, the OKS result was significant higher in the experimental group during all the follow-ups (1mth: p = 0.0295; 2mths: p = 0.0012; 6mths: p = 0.0001; 12mths: p < 0.0001; 36mths: p = 0.0061). CONCLUSIONS: The use of PEMFs leads to significant pain relief, better clinical improvement, and lower NSAIDs consumption after medial UKA when compared to the control group. LEVEL OF EVIDENCE: II.


Asunto(s)
Artroplastia de Reemplazo de Rodilla , Humanos , Manejo del Dolor , Campos Electromagnéticos , Antiinflamatorios no Esteroideos/uso terapéutico , Dolor
20.
Appl Sci (Basel) ; 12(23)2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36970107

RESUMEN

Articular cartilage injuries are a common source of joint pain and dysfunction. As articular cartilage is avascular, it exhibits a poor intrinsic healing capacity for self-repair. Clinically, osteochondral grafts are used to surgically restore the articular surface following injury. A significant challenge remains with the repair properties at the graft-host tissue interface as proper integration is critical toward restoring normal load distribution across the joint. A key to addressing poor tissue integration may involve optimizing mobilization of fibroblast-like synoviocytes (FLS) that exhibit chondrogenic potential and are derived from the adjacent synovium, the specialized connective tissue membrane that envelops the diarthrodial joint. Synovium-derived cells have been directly implicated in the native repair response of articular cartilage. Electrotherapeutics hold potential as low-cost, low-risk, non-invasive adjunctive therapies for promoting cartilage healing via cell-mediated repair. Pulsed electromagnetic fields (PEMFs) and applied direct current (DC) electric fields (EFs) via galvanotaxis are two potential therapeutic strategies to promote cartilage repair by stimulating the migration of FLS within a wound or defect site. PEMF chambers were calibrated to recapitulate clinical standards (1.5 ± 0.2 mT, 75 Hz, 1.3 ms duration). PEMF stimulation promoted bovine FLS migration using a 2D in vitro scratch assay to assess the rate of wound closure following cruciform injury. Galvanotaxis DC EF stimulation assisted FLS migration within a collagen hydrogel matrix in order to promote cartilage repair. A novel tissue-scale bioreactor capable of applying DC EFs in sterile culture conditions to 3D constructs was designed in order to track the increased recruitment of synovial repair cells via galvanotaxis from intact bovine synovium explants to the site of a cartilage wound injury. PEMF stimulation further modulated FLS migration into the bovine cartilage defect region. Biochemical composition, histological analysis, and gene expression revealed elevated GAG and collagen levels following PEMF treatment, indicative of its pro-anabolic effect. Together, PEMF and galvanotaxis DC EF modulation are electrotherapeutic strategies with complementary repair properties. Both procedures may enable direct migration or selective homing of target cells to defect sites, thus augmenting natural repair processes for improving cartilage repair and healing.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA