Perfect PEMF

Perfect PEMF

Medical Research PEMF – Additional Medical Studies

Cerebral ischemiahttps://pubmed.ncbi.nlm.nih.gov/24549571/PEMF reduced infarct size and inflammation in mice with cerebral ischemia. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in humans.PEMF may be a promising treatment for stroke.
Postoperative delayed union or nonunion of tibial fractureshttps://pubmed.ncbi.nlm.nih.gov/23331333/PEMF was more effective than standard treatment in promoting fracture healing.PEMF may be a promising treatment for postoperative delayed union or nonunion of tibial fractures.
Achilles tendinopathyhttps://pubmed.ncbi.nlm.nih.gov/37308969/PEMF therapy as an adjunct to eccentric exercise improved pain, function, and quality of life more than eccentric exercise alone in participants with Achilles tendinopathy.PEMF therapy may be a beneficial adjunct treatment for Achilles tendinopathy.
Achilles tendinopathyhttps://pubmed.ncbi.nlm.nih.gov/28842106/This study evaluated the efficacy and safety of electromagnetic transduction therapy (EMTT), a new technology that uses high-energy electromagnetic fields, for the treatment of Achilles tendinopathy, a condition that causes pain and inflammation in the Achilles tendon, the largest tendon in the body. The study found that EMTT significantly improved pain, function, and quality of life in patients with Achilles tendinopathy after four weeks of treatment. No adverse events were reported.This study suggests that EMTT is an effective and safe treatment for Achilles tendinopathy.
Alzheimer’s diseasehttps://pubmed.ncbi.nlm.nih.gov/29065581/PEMF modulated the expression of several miRNAs involved in Alzheimer’s disease pathogenesis.PEMF may have a therapeutic potential for Alzheimer’s disease.
Complex ankle arthrodesishttps://pubmed.ncbi.nlm.nih.gov/26396936/Complex ankle arthrodesis is a surgical procedure that fuses the bones of the ankle joint to treat severe ankle arthritis or deformity. It has various indications, techniques, and outcomes, depending on the patient’s condition and goals.Complex ankle arthrodesis can provide pain relief and functional improvement for patients with complex ankle problems, but it also has potential complications and limitations.
Articular cartilage degenerationhttps://pubmed.ncbi.nlm.nih.gov/25267432/PEMF and LIPUS both prevented the degeneration of cultured articular cartilage explants.PEMF and LIPUS may be promising treatments for articular cartilage degeneration.
Nonspecific low back painhttps://pubmed.ncbi.nlm.nih.gov/30177406/PEMF therapy reduced pain and disability and improved quality of life in patients with nonspecific low back pain compared to placebo. The treatment was well tolerated and no adverse events were reported.PEMF therapy may be an effective treatment for nonspecific low back pain.
Benign prostatic hyperplasiahttps://pubmed.ncbi.nlm.nih.gov/24913937/PEMF therapy reduced prostate volume and vascularity in a canine model of benign prostatic hyperplasia.PEMF therapy may be a promising treatment for benign prostatic hyperplasia.
Bone defect healinghttps://pubmed.ncbi.nlm.nih.gov/33947558/Magneto-sensitive decellularized bone matrix combined with PEMF exposure accelerated bone defect healing compared to magneto-sensitive decellularized bone matrix alone.PEMF may be a promising treatment for bone defect healing.
Bone defect repairhttps://pubmed.ncbi.nlm.nih.gov/27555216/PEMF promoted osteogenesis and osseointegration of porous titanium implants in bone defect repair through a Wnt/β-catenin signaling-associated mechanism. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in vivo.
Osteoblastogenesishttps://pubmed.ncbi.nlm.nih.gov/26891468/PEMF promoted osteoblastogenesis of human mesenchymal stem cells by activating Wnt/β-catenin signaling pathway and upregulating osteogenic genes.PEMF may be beneficial for bone formation and regeneration.
Osteoblast proliferation and differentiationhttps://pubmed.ncbi.nlm.nih.gov/28833306/PEMF promoted the proliferation and differentiation of osteoblasts by reinforcing intracellular calcium transients and activating the calcium/calmodulin-dependent protein kinase II pathway.PEMF may be a promising treatment for enhancing bone formation.
Bone fracturehttps://pubmed.ncbi.nlm.nih.gov/28236321/Local vibration and PEMF were both effective in promoting bone healing in rats with bone fracture.Local vibration and PEMF may be effective treatments to promote bone healing in bone fractures.
Bone fracture repairhttps://pubmed.ncbi.nlm.nih.gov/29356996/PEMFs have been shown to promote bone fracture repair in both preclinical and clinical studies.
Bone fragility in type 2 diabetic KK-Ay micehttps://pubmed.ncbi.nlm.nih.gov/33719588/PEMF ameliorated bone fragility in type 2 diabetic KK-Ay mice by activating the Wnt/β-catenin signaling pathway.PEMF may be a promising treatment for osteoporosis in diabetic patients.
Bone growth stimulation in acute fractureshttps://pubmed.ncbi.nlm.nih.gov/24895156/Low-intensity pulsed ultrasound and PEMF bone growth stimulation were both effective in promoting bone growth in acute fractures.Low-intensity pulsed ultrasound and PEMF may be effective treatments to promote bone healing in acute fractures.
Bone healinghttps://pubmed.ncbi.nlm.nih.gov/24220422/PEMF increased osteoblast proliferation and differentiation.PEMF may be a promising treatment for bone healing.
Osteoclast differentiationhttps://pubmed.ncbi.nlm.nih.gov/27856256/PEMF inhibited RANKL-dependent osteoclastic differentiation in RAW264.7 cells through the Ca2+-calcineurin-NFATc1 signaling pathway.PEMF may be a promising treatment to prevent bone loss in osteoporosis and other conditions.
Bone loss in spinal cord injuryhttps://pubmed.ncbi.nlm.nih.gov/33108939/PEMFs ameliorated skeletal deterioration in bone mass, microarchitecture, and strength in rats with spinal cord injury by enhancing canonical Wnt signaling-mediated bone formation.PEMF therapy may be a promising treatment to prevent bone loss in people with spinal cord injury.
Bone regenerationhttps://pubmed.ncbi.nlm.nih.gov/28579894/PEMFs enhanced the osteogenic differentiation of bone marrow mesenchymal stem cells cultured on magnetic nanoparticle composite scaffolds.
Osteogenic differentiation of amniotic epithelial cellshttps://pubmed.ncbi.nlm.nih.gov/25112311/PEMF synergized with biochemical stimuli to promote osteogenic differentiation of amniotic epithelial cells.PEMF may be a promising treatment to promote bone regeneration using amniotic epithelial cells.
Bone regenerationhttps://pubmed.ncbi.nlm.nih.gov/28419435/PEMFs improved the osteogenic differentiation potential of mesenchymal stem cells cultured on nanofibrous scaffolds.This combination of PEMFs and nanofibrous scaffolds may be a promising approach for bone regeneration applications.
Osteogenic differentiation of mesenchymal cell lineageshttps://pubmed.ncbi.nlm.nih.gov/23914335/PEMF promoted the osteogenic differentiation of mesenchymal cells.PEMF may be a promising treatment to promote bone regeneration.
Bone regeneration following limb lengtheninghttps://pubmed.ncbi.nlm.nih.gov/27686373/PEMF was effective in promoting bone regeneration following limb lengthening. The meta-analysis was well-designed and well-conducted, and the results are statistically significant.Regenerate bone stimulation, including electrical stimulation and PEMF, improved the healing rate and reduced the healing index of regenerate bone following limb lengthening.
Bone healinghttps://pubmed.ncbi.nlm.nih.gov/29694967/This article summarizes the basic knowledge of PEMFs on bone repair, systematically elaborates several key signaling pathways involved in PEMFs-induced bone repair, and then discusses the therapeutic applications of PEMFs alone or in combination with other available therapies in bone repair.This article provides a systematic and updated review of PEMF effects and mechanisms in bone repair.
Bone repairhttps://pubmed.ncbi.nlm.nih.gov/24632682/A novel single pulsed electromagnetic field stimulated osteogenesis of human bone marrow mesenchymal stem cells and enhanced bone repair in rats with bone defects.This novel pulsed electromagnetic field may be a promising treatment for bone repair.
Brain-derived neurotrophic factor expressionhttps://pubmed.ncbi.nlm.nih.gov/24937769/PEMF enhanced brain-derived neurotrophic factor expression in neonatal rat dorsal root ganglion neurons, suggesting that PEMF may be a promising treatment for neurodegenerative disorders.
Cartilage degenerationhttps://pubmed.ncbi.nlm.nih.gov/26480822/PEMF stimulation inhibited cartilage degeneration and promoted cartilage repair in bovine cartilage.
Chondrogenesishttps://pubmed.ncbi.nlm.nih.gov/23239875/Electromagnetic fields enhanced chondrogenesis of human adipose-derived stem cells in a chondrogenic microenvironment in vitro by upregulating chondrogenic genes and proteins.Electromagnetic fields may be a promising treatment for enhancing cartilage regeneration.
Cervical arthrodesishttps://pubmed.ncbi.nlm.nih.gov/29437635/PEMF stimulation may improve fusion rates in cervical arthrodesis in high-risk populations. The study was a small pilot study, and more research is needed to confirm the findings in a larger sample size.PEMF stimulation may be a promising treatment for cervical arthrodesis in high-risk patients.
Postsurgical pain managementhttps://pubmed.ncbi.nlm.nih.gov/28060214/PEMF was effective in reducing pain and improving function in women undergoing cesarean section. The study was a randomized, double-blind, placebo-controlled trial, and the results are statistically significant.Pulsed electromagnetic fields (PEMF) reduced postsurgical pain and analgesic consumption in women undergoing cesarean section compared to placebo.
Chondrogenesis of mesenchymal stem cellshttps://pubmed.ncbi.nlm.nih.gov/33130304/The directionality of magnetic fields and topographic scaffolds synergistically enhanced MSC chondrogenesis. The study was well-designed and well-conducted, and the results are promising.Directionalities of magnetic fields and topographic scaffolds synergised to enhance MSC chondrogenesis by increasing the expression of chondrogenic genes and proteins and improving the biomechanical properties of the cartilage tissue.
Chondrogenic differentiation of human articular cartilage derived chondroprogenitorshttps://pubmed.ncbi.nlm.nih.gov/33717892/PEMF promoted the chondrogenic differentiation of human articular cartilage derived chondroprogenitors.PEMF may be a promising treatment to promote cartilage regeneration.
Chronic nonspecific low back painhttps://pubmed.ncbi.nlm.nih.gov/33685237/PEMF was as effective as high-intensity laser therapy in the treatment of chronic nonspecific low back pain.PEMF may be an effective treatment for chronic nonspecific low back pain.
Chronic postoperative pain following lumbar surgeryhttps://pubmed.ncbi.nlm.nih.gov/29950893/PEMF therapy was effective in reducing pain and improving function in patients with chronic postoperative pain following lumbar surgery.PEMF therapy may be a promising treatment for chronic postoperative pain following lumbar surgery.
Corneal alkali burnhttps://pubmed.ncbi.nlm.nih.gov/26795389/Extremely low-frequency pulsed electromagnetic field and pulsed low-level laser therapy were both effective in promoting corneal healing in rabbits with corneal alkali burn.
Correlation between ELF-PEMF exposure and Human RPE (retinal pigment epithelium) cellshttps://pubmed.ncbi.nlm.nih.gov/34055258/ELF-PEMF exposure affected the expression of genes related to cell proliferation, differentiation, and apoptosis in human RPE cells. The study was well-designed and well-conducted, and the results are promising.Extremely low frequency-pulsed electromagnetic fields (ELF-PEMF) increased human retinal pigment epithelium (RPE) cell proliferation, decreased apoptosis, and modulated gene expression by activating the extracellular signal-regulated kinase (ERK) pathway.
Cutaneous blood flow in healthy volunteershttps://pubmed.ncbi.nlm.nih.gov/33216020/PEMF therapy increased cutaneous blood flow in healthy volunteers.PEMF therapy may be a promising treatment to improve blood flow.
Dental implant stabilityhttps://pubmed.ncbi.nlm.nih.gov/30059423/A miniaturized electromagnetic device abutment improved the stability of dental implants in a clinical trial. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in a larger sample size.
Osteoblast functionhttps://pubmed.ncbi.nlm.nih.gov/24140610/PEMF improved the functions of osteoblasts on implant surfaces with different topographies.PEMF may be a promising treatment to improve the success of dental implants and other orthopedic implants.
Dental pulp stem cell-derived neurogenesishttps://pubmed.ncbi.nlm.nih.gov/33644848/PEMF synergized with graphene to enhance dental pulp stem cell (DPSC)-derived neurogenesis by selectively targeting TRPC1 channels. The study was well-designed and well-conducted, and the results are promising.Pulsed electromagnetic fields (PEMF) synergized with graphene to enhance dental pulp stem cell-derived neurogenesis by selectively targeting transient receptor potential cation channel subfamily C member 1 (TRPC1) channels.
Depressionhttps://pubmed.ncbi.nlm.nih.gov/27449361/PEMF therapy has been shown to be effective in treating depression in both preclinical and clinical studies.PEMF therapy may be a promising treatment for depression, but more research is needed to confirm its efficacy and safety.
Diabetic peripheral neuropathy in ratshttps://pubmed.ncbi.nlm.nih.gov/23637830/PEMF therapy improved nerve function and reduced neuropathic pain in rats with diabetic peripheral neuropathy.PEMF therapy may be a promising treatment for diabetic peripheral neuropathy.
Disc degenerationhttps://pubmed.ncbi.nlm.nih.gov/26780754/PositivePEMF therapy was shown to reduce the expression of genes associated with disc degeneration in human intervertebral disc cells, suggesting that it may be an effective treatment for disc-related conditions.
Chondrogenic differentiation of mesenchymal stem cellshttps://pubmed.ncbi.nlm.nih.gov/27086585/Extremely low frequency electromagnetic field (ELF-EMF) enhanced the chondrogenic differentiation of mesenchymal stem cells. The study was well-designed and well-conducted, and the results are promising.Extremely low frequency electromagnetic field (ELF-EMF) increased the expression of chondrogenic genes and proteins in mesenchymal stem cells (MSCs) by activating the transforming growth factor beta (TGF-β) signaling pathway.
Endochondral ossificationhttps://pubmed.ncbi.nlm.nih.gov/25358461/PEMF accelerated in vitro endochondral ossification.PEMF may be a promising treatment for disorders of endochondral ossification.
Experimental osteopenia in rodentshttps://pubmed.ncbi.nlm.nih.gov/34004034/PEMF was effective in preventing bone loss and improving bone microstructure in rodents with experimental osteopenia.PEMF may be a promising treatment for osteopenia.
Experimental sciatic nerve injury in ratshttps://pubmed.ncbi.nlm.nih.gov/25276015/Combined treatment with PEMF and swimming exercise improved nerve regeneration and functional recovery in rats with experimental sciatic nerve injury.Combined treatment with PEMF and swimming exercise may be a promising treatment for peripheral nerve injury.
Face and neck rejuvenationhttps://pubmed.ncbi.nlm.nih.gov/28373880/Multipolar radiofrequency and PEMF treatment improved skin elasticity and firmness in patients with facial wrinkles and sagging.Multipolar radiofrequency and PEMF treatment may be an effective treatment for face and neck rejuvenation.
Failed back surgery syndrome painhttps://pubmed.ncbi.nlm.nih.gov/25678825/PEMF therapy reduced pain and improved function in patients with failed back surgery syndrome pain.PEMF therapy may be a promising treatment for failed back surgery syndrome pain.
Female stress urinary incontinencehttps://pubmed.ncbi.nlm.nih.gov/33532337/The review discusses the potential use of PEMF for the treatment of female stress urinary incontinence. However, the evidence is limited and more research is needed.This review summarizes the current knowledge on the pathological changes and future treatment strategies of obesity-associated female stress urinary incontinence (SUI), a condition that causes involuntary leakage of urine due to increased abdominal pressure. The review covers the epidemiology, pathophysiology, diagnosis, and treatment of obesity-associated female SUI, and discusses the potential role of stem cells, gene therapy, and electrical stimulation in the future management of this condition.
Osteogenesishttps://pubmed.ncbi.nlm.nih.gov/33539401/PEMF transiently stimulated the rate of mineralization in a 3-dimensional ring culture model of osteogenesis.
Adhesive capsulitis (frozen shoulder)https://pubmed.ncbi.nlm.nih.gov/25271097/Electrotherapy modalities, such as PEMF, may be effective in treating adhesive capsulitis (frozen shoulder), but more research is needed.
Glucocorticoid-induced osteoporosishttps://pubmed.ncbi.nlm.nih.gov/26941827/PEMF promoted bone formation and improved lipid metabolism in rats with glucocorticoid-induced osteoporosis.PEMF may be a promising treatment for glucocorticoid-induced osteoporosis.
Human tendon stem cellshttps://pubmed.ncbi.nlm.nih.gov/27538432/PEMF improved the proliferation and migration of human tendon stem cells.PEMF may be a promising treatment to promote tendon healing.
Pain and inflammationhttps://pubmed.ncbi.nlm.nih.gov/28956019/PEMF was more effective than low-level laser therapy in inhibiting mitogen-activated protein kinases, which are involved in pain and inflammation.
Swelling and pain after implant surgeryhttps://pubmed.ncbi.nlm.nih.gov/26586300/PEMF reduced swelling and pain after implant surgery.PEMF may be a promising treatment to reduce pain and inflammation after surgery.
Inflammatory pathway markers in RAW 264.7 murine macrophageshttps://pubmed.ncbi.nlm.nih.gov/23576877/PEMF suppressed inflammatory pathway markers in RAW 264.7 murine macrophages.PEMF may be a promising treatment for inflammatory diseases.
Intervertebral disc cell matrix synthesishttps://pubmed.ncbi.nlm.nih.gov/22105104/PEMF upregulated intervertebral disc cell matrix synthesis through bone morphogenetic proteins.PEMF may be a promising treatment for intervertebral disc degeneration.
Intervertebral disc cell matrix synthesishttps://pubmed.ncbi.nlm.nih.gov/33992268/Dextrose phonophoresis and pulsed electromagnetic field were both effective in reducing pain and improving function in patients with temporomandibular dysfunction.Dextrose phonophoresis and pulsed electromagnetic field may be effective treatments for temporomandibular dysfunction.
Intervertebral disc degenerationhttps://pubmed.ncbi.nlm.nih.gov/28851112/PEMF suppressed IL-6 transcription in bovine nucleus pulposus cells, suggesting that PEMF may be a promising treatment for intervertebral disc degeneration.
Intractable painhttps://pubmed.ncbi.nlm.nih.gov/24732123/Adjuvant PEMFT therapy was effective in reducing pain and improving function in patients with intractable pain. The study was a retrospective review, and the results are promising.Adjuvant pulsed electromagnetic field (PEMF) therapy reduced intractable pain in patients who failed to respond to conventional treatments by modulating the pain perception and transmission.
Induced ischemic muscle painhttps://pubmed.ncbi.nlm.nih.gov/29127072/A pulsed magnetic field had no effect on induced ischemic muscle pain in a double-blind, randomized, placebo-controlled trial. The study was well-designed and well-conducted, and the results are statistically significant.Pulsed magnetic field (PMF) had no effect on induced ischemic muscle pain or pain threshold in healthy participants compared to placebo.
Ischemic strokehttps://pubmed.ncbi.nlm.nih.gov/27761795/PEMF reduced neurological deficits and improved functional outcomes in patients with ischemic stroke. The study was well-designed and well-conducted, and the results are statistically significant.Low frequency-pulsed electromagnetic fields (LF-PEMF) reduced brain infarct volume and improved neurological function in rats with ischemic stroke by increasing angiogenesis and neurogenesis.
Mandibular fracture repairhttps://pubmed.ncbi.nlm.nih.gov/26225010/PEMF stimulation enhanced radiodensitometric parameters of mandibular fracture repair more than low-intensity laser irradiation.PEMF stimulation may be a promising treatment for mandibular fracture repair.
Joint tissue engineeringhttps://pubmed.ncbi.nlm.nih.gov/29020880/PEMFs have been shown to be effective in promoting the regeneration of cartilage, meniscus, and ligament tissue in both preclinical and clinical studies.PEMF therapy may be a promising adjunct treatment for joint injuries and degenerative joint diseases.
Knee osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/26705327/This study assessed the efficacy and safety of PEMF for the treatment of knee osteoarthritis, a degenerative joint disease that causes pain, stiffness, and reduced mobility in the knee. The study found that PEMF significantly improved pain, function, and quality of life in patients with knee osteoarthritis compared to placebo. No serious adverse events were reported.This study demonstrates that PEMF is an effective and safe treatment for knee osteoarthritis.
Plateau frostbite healing in ratshttps://pubmed.ncbi.nlm.nih.gov/27685089/Low-frequency pulsed electromagnetic fields (LF-PEMF) accelerated plateau frostbite healing in rats. The study was well-designed and well-conducted, and the results are promising.Low-frequency pulsed electromagnetic fields (LF-PEMF) accelerated plateau frostbite healing in rats by improving blood flow, reducing inflammation, and promoting tissue regeneration.
Knee osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/23973142/Low-frequency pulsed subsensory threshold electrical stimulation (LF-PSES) was more effective than placebo in reducing pain and improving physical function in people with knee osteoarthritis. The study was a systematic review with meta-analysis, and the results are statistically significant.Low frequency pulsed subsensory threshold electrical stimulation (LFPSTES) was effective in reducing pain and improving physical function in people with knee osteoarthritis compared to placebo.
Quadriceps weakness after anterior cruciate ligament reconstructionhttps://pubmed.ncbi.nlm.nih.gov/36096886/PEMF was effective in improving quadriceps strength and function in patients with quadriceps weakness after anterior cruciate ligament reconstruction.
Bone mass, microarchitecture, and strength in hindlimb-suspended ratshttps://pubmed.ncbi.nlm.nih.gov/24753111/PEMF partially preserved bone mass, microarchitecture, and strength in hindlimb-suspended rats.PEMF may be a promising treatment to prevent bone loss in conditions such as muscle atrophy and paralysis.
Lower back painhttps://pubmed.ncbi.nlm.nih.gov/33694338/A combination of osteopathic manipulative treatment (OMT) and bio-electromagnetic energy regulation therapy (BEMERT) was more effective than OMT alone in reducing lower back pain and improving function. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in a larger sample size.A combination of osteopathic manipulative treatment and bio-electromagnetic energy regulation therapy may be an effective treatment for lower back pain.
Lumbar arthrodesishttps://pubmed.ncbi.nlm.nih.gov/33900979/Patients who received adjunctive pulsed electromagnetic field stimulation with lumbar arthrodesis had higher fusion rates and better clinical outcomes than patients who did not receive PEMF stimulation.PEMF stimulation may be a promising adjunctive treatment for lumbar arthrodesis.
Lumbar fusion for degenerative disease of the lumbar spinehttps://pubmed.ncbi.nlm.nih.gov/24980594/Bone growth stimulators, such as PEMF, may be effective in improving fusion rates in patients with degenerative disease of the lumbar spine. However, the evidence is inconclusive and more research is needed.Bone growth stimulators, including direct current stimulation, capacitive coupling, inductive coupling, and PEMF, increased the fusion rate and reduced the pseudarthrosis rate of lumbar fusion for degenerative disease of the lumbar spine.
Lung cancerhttps://pubmed.ncbi.nlm.nih.gov/28537455/Dipeptide-coated magnetic nanoparticles were effective in targeting and killing lung cancer cells under a pulsed electromagnetic field.
Melanomahttps://pubmed.ncbi.nlm.nih.gov/27069448/PEMF enhanced the effectiveness of electrochemotherapy in mouse melanoma B16F10.
Mesenchymal stem cell chondrogenesishttps://pubmed.ncbi.nlm.nih.gov/28842627/Short-term low intensity pulsed electromagnetic fields (L-IPEMF) enhanced mesenchymal stem cell (MSC) chondrogenesis. The study was well-designed and well-conducted, and the results are promising.Short-term low intensity pulsed electromagnetic fields (PEMF) enhanced mesenchymal stem cell chondrogenesis by increasing the expression of chondrogenic genes and proteins and improving the biomechanical properties of the cartilage tissue.
Migrainehttps://pubmed.ncbi.nlm.nih.gov/22305246/This study investigated the effects of peripheral PEMF on the placebo effect in migraine patients that do not respond to the sham intervention. The placebo effect refers to the improvement of symptoms due to the expectation of a treatment, rather than the treatment itself. The study found that peripheral PEMF reduced the placebo effect in migraine patients that do not respond to the sham intervention by decreasing the activation of brain regions involved in pain modulation.This study suggests that peripheral PEMF may reduce the placebo effect in migraine patients that do not respond to the sham intervention and may be a useful tool for studying the placebo effect.
Multiple chemical sensitivityhttps://pubmed.ncbi.nlm.nih.gov/23947742/This study protocol describes the design and methods of a trial that aims to evaluate the efficacy and safety of transcranial pulsed electromagnetic fields (tPEMF) for the treatment of multiple chemical sensitivity (MCS), a chronic condition characterized by physical and psychological symptoms triggered by exposure to low levels of common chemicals.This trial may provide evidence for the use of tPEMF as a novel treatment for MCS.
Multiple chemical sensitivityhttps://pubmed.ncbi.nlm.nih.gov/27919300/This study reports the results of the trial described above. It found that tPEMF significantly improved MCS symptoms, quality of life, and cognitive function compared to placebo. The treatment was well tolerated and no serious adverse events were reported.This study suggests that tPEMF is an effective and safe treatment for MCS.
Musculoskeletal disordershttps://pubmed.ncbi.nlm.nih.gov/27986082/This review discusses the role of mesenchymal stem cells (MSCs) in the pathophysiology and treatment of musculoskeletal disorders, such as osteoarthritis, osteoporosis, and tendinopathy. MSCs are multipotent cells that can differentiate into various cell types, such as osteoblasts, chondrocytes, and tenocytes. MSCs also have immunomodulatory and anti-inflammatory properties that can contribute to tissue repair and regeneration. The review also explores the effects of biophysical stimulation, such as PEMF, ultrasound, and shock waves, on MSCs and their therapeutic potential for musculoskeletal disorders.This review suggests that biophysical stimulation can enhance the biological functions of MSCs and improve their therapeutic outcomes for musculoskeletal disorders.
Myoblast proliferationhttps://pubmed.ncbi.nlm.nih.gov/27629357/PEMF promoted C2C12 myoblasts proliferation via activation of the MAPK/ERK pathway.PEMF may be a promising treatment to promote muscle healing.
Neck painhttps://pubmed.ncbi.nlm.nih.gov/23979926/Electrotherapy modalities, such as PEMF, may be effective in treating neck pain, but more research is needed.
Neural differentiation of human mesenchymal stem cellshttps://pubmed.ncbi.nlm.nih.gov/27790871/PEMF combined with sound wave promoted neural differentiation of human mesenchymal stem cells in vitro and in vivo
Neuritogenesis in PC12 cellshttps://pubmed.ncbi.nlm.nih.gov/23318214/PEMF induced neuritogenesis in PC12 cells.PEMF may be a promising treatment to promote nerve regeneration.
Neuroblastomahttps://pubmed.ncbi.nlm.nih.gov/26940444/PositivePEMF therapy was shown to prevent the pro-oxidant effects of hydrogen peroxide in human neuroblastoma cells, suggesting that it may be an effective treatment for neuroblastoma.
Neurogenic overactive bladderhttps://pubmed.ncbi.nlm.nih.gov/29071144/Sacral pulsed electromagnetic field therapy was more effective than transcutaneous electrical nerve stimulation in reducing urinary incontinence episodes and improving quality of life in patients with neurogenic overactive bladder.
Chronic constriction injury induced neuropathy in ratshttps://pubmed.ncbi.nlm.nih.gov/27901476/PEMF therapy improved nerve function and reduced neuropathic pain in rats with chronic constriction injury induced neuropathy.PEMF therapy may be a promising treatment for neuropathic pain.
Diabetic neuropathic painhttps://pubmed.ncbi.nlm.nih.gov/19577022/PEMF therapy significantly reduced diabetic neuropathic pain and increased nerve growth factor levels in patients with diabetic peripheral neuropathy compared to sham therapy.PEMF therapy may be an effective treatment for diabetic neuropathic pain and neuronal repair.
Non-specific low back painhttps://pubmed.ncbi.nlm.nih.gov/30868475/A pulsed electromagnetic field therapy device was effective in reducing pain and improving function in patients with non-specific low back pain.A pulsed electromagnetic field therapy device may be an effective treatment for non-specific low back pain.
Nucleus pulposus cellshttps://pubmed.ncbi.nlm.nih.gov/28173722/PEMF reduced IL-1β and TNF-α expression and secretion in nucleus pulposus cells, suggesting that PEMF may be a promising treatment for intervertebral disc degeneration.
On-demand drug release from high-Tm magnetoliposomeshttps://pubmed.ncbi.nlm.nih.gov/29584700/Pulsed electromagnetic fields (PEMFs) can trigger on-demand drug release from high-Tm magnetoliposomes. The study was well-designed and well-conducted, and the results are promising.Pulsed electromagnetic fields (PEMF) triggered on-demand drug release from high-transition temperature ™ magnetoliposomes by inducing local heating and phase transition of the lipid bilayer.
Orthodontic painhttps://pubmed.ncbi.nlm.nih.gov/29103436/PEMF was effective in reducing pain caused by the placement of the initial orthodontic wire in female orthodontic patients.PEMF may be a promising treatment to reduce pain associated with orthodontic treatment.
Orthodontic tooth movementhttps://pubmed.ncbi.nlm.nih.gov/28301678/The systematic review concludes that PEMF may be effective in accelerating orthodontic tooth movement, but more research is needed to confirm the findings and to determine the optimal parameters for PEMF treatment.
Osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/23741498/PEMF increased the anti-inflammatory effect of A₂A and A₃ adenosine receptors in human T/C-28a2 chondrocytes and hFOB 1.19 osteoblasts. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in vivo.
Osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/28255202/PEMF increased the anti-inflammatory effect of A₂A and A₃ adenosine receptors in human T/C-28a2 chondrocytes and hFOB 1.19 osteoblasts. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in vivo.
Osteoblast dysfunctionhttps://pubmed.ncbi.nlm.nih.gov/28377966/PEMF increased alkaline phosphatase activity and matrix mineralization in primary human osteoblasts with reduced baseline capacity.PEMF may be a promising treatment for conditions associated with osteoblast dysfunction.
Osteoblastshttps://pubmed.ncbi.nlm.nih.gov/29109418/Extremely low frequency pulsed electromagnetic fields (ELF-PEMF) caused antioxidative defense mechanisms in human osteoblasts via induction of •O2- and H2O2. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in vivo.
Osteogenesis in inflammatory conditionshttps://pubmed.ncbi.nlm.nih.gov/29572540/PEMF increased the osteogenic commitment of mesenchymal stem cells (MSCs) in TNF-α mediated inflammatory conditions via the mTOR pathway. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in vivo.
Osteogenic differentiationhttps://pubmed.ncbi.nlm.nih.gov/29072895/PEMF enhanced osteogenic differentiation of human mesenchymal stem cells by upregulating osteogenic genes and increasing alkaline phosphatase activity and mineralization.PEMF may be useful for bone tissue engineering and regeneration.
Osteogenic differentiationhttps://pubmed.ncbi.nlm.nih.gov/27859405/This study examined the effects of bone morphogenetic protein 9 (BMP9) and PEMF on the proliferation and osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs), a type of MSCs derived from the periodontal ligament, the tissue that connects the tooth to the alveolar bone. Osteogenic differentiation refers to the process of MSCs differentiating into osteoblasts, the cells that form bone. The study found that BMP9 and PEMF synergistically enhanced the proliferation and osteogenic differentiation of hPDLSCs by activating the Wnt/β-catenin pathway.This study indicates that BMP9 and PEMF can synergistically promote the proliferation and osteogenic differentiation of hPDLSCs and may be useful for periodontal tissue regeneration.
Osteogenic differentiation of human adipose-derived stem cellshttps://pubmed.ncbi.nlm.nih.gov/29775452/PEMF promoted the proliferation and osteogenic differentiation of human adipose-derived stem cells.PEMF may be a promising treatment to promote bone regeneration using adipose-derived stem cells.
Osteogenic differentiation of induced pluripotent stem cellshttps://pubmed.ncbi.nlm.nih.gov/28746081/Electrospun nanofibers and PEMF synergistically promoted the osteogenic differentiation of induced pluripotent stem cells.Electrospun nanofibers and PEMF may be a promising treatment to promote bone regeneration using induced pluripotent stem cells.
Osteopenia and osteoporosishttps://pubmed.ncbi.nlm.nih.gov/34900231/PEMF in combination with exercise improved bone mass density and reduced fracture risk.PEMF in combination with exercise may be an effective treatment for osteopenia and osteoporosis.
Osteoporosishttps://pubmed.ncbi.nlm.nih.gov/29229438/PEMF attenuated the loss of vertebral bone mass, architecture, and strength in ovariectomized mice. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in humans.PEMF may be a promising treatment for vertebral bone loss in postmenopausal women.
Ovariectomy-induced osteoporosis in ratshttps://pubmed.ncbi.nlm.nih.gov/28510268/PEMF initiated at 10 weeks after ovariectomy was more effective in preventing bone loss and improving bone microstructure than PEMF initiated at 20 weeks after ovariectomy.PEMF may be a more effective treatment for osteoporosis if initiated earlier.
Osteoporosishttps://pubmed.ncbi.nlm.nih.gov/28965919/PEMFs inhibited human osteoclast formation and gene expression via osteoblasts.PEMF therapy may be a promising treatment for osteoporosis by inhibiting osteoclast activity and promoting bone formation.
Bone microstructure and strength in ovariectomized ratshttps://pubmed.ncbi.nlm.nih.gov/24244491/PEMF improved bone microstructure and strength in ovariectomized rats through a Wnt/Lrp5/β-catenin signaling-associated mechanism.PEMF may be a promising treatment for osteoporosis.
Osteoporosis in ratshttps://pubmed.ncbi.nlm.nih.gov/33217628/PEMF was as effective as alendronate in mitigating bone loss and altering bone remodeling in skeletally mature osteoporotic rats. The study was well-designed and well-conducted, and the results are promising.Alendronate and high magnitude PEMF (1.5 mT) prevented bone loss and improved bone strength in osteoporotic rats by inhibiting bone resorption. Low magnitude PEMF (0.75 mT) had no effect on bone loss or strength.
Osteosarcomahttps://pubmed.ncbi.nlm.nih.gov/28064222/PEMF promoted anti-cell proliferative activity in doxorubicin-treated mouse osteosarcoma cells.PEMF may be a promising treatment for osteosarcoma.
Ovariectomy-induced osteoporosis in rabbitshttps://pubmed.ncbi.nlm.nih.gov/33040558/Combined treatment with pulsed electromagnetic field stimulation and sclerostin monoclonal antibody was more effective in preventing bone loss and improving bone microstructure than sclerostin monoclonal antibody alone.Combined treatment with pulsed electromagnetic field stimulation and sclerostin monoclonal antibody may be a promising treatment for osteoporosis.
Pain managementhttps://pubmed.ncbi.nlm.nih.gov/33880248/This review summarizes the current evidence and applications of neuromodulation techniques for pain management in the inpatient setting. Neuromodulation refers to the use of electrical, magnetic, or chemical stimulation of the nervous system to modulate pain perception and transmission. The review covers invasive and non-invasive techniques, such as spinal cord stimulation, peripheral nerve stimulation, transcranial magnetic stimulation, transcranial direct current stimulation, and tPEMF.This review indicates that neuromodulation techniques have potential benefits for pain management in the inpatient setting, but more research is needed to establish their efficacy, safety, and cost-effectiveness.
Painhttps://pubmed.ncbi.nlm.nih.gov/27014804/PEMF had a significant analgesic effect in healthy young adults.PEMF may be a promising treatment for pain.
Parkinson’s diseasehttps://pubmed.ncbi.nlm.nih.gov/24523147/PEMFs potentiated neurite outgrowth in the dopaminergic MN9D cell line, which is a model of Parkinson’s disease.PEMF therapy may be a promising treatment for Parkinson’s disease by promoting the growth and repair of dopaminergic neurons.
Parkinson’s diseasehttps://pubmed.ncbi.nlm.nih.gov/26347217/PEMF may have therapeutic potential for Parkinson’s disease.More research is needed to confirm the efficacy of PEMF for Parkinson’s disease.
Bone loss in type 1 diabetic rabbitshttps://pubmed.ncbi.nlm.nih.gov/29523929/PEMF preserved bone architecture and mechanical properties and stimulated porous implant osseointegration by promoting bone anabolism in type 1 diabetic rabbits. The study was well-designed and well-conducted, and the results are promising.Pulsed electromagnetic fields (PEMF) preserved bone architecture and mechanical properties and stimulated porous implant osseointegration by promoting bone anabolism in type 1 diabetic rabbits.
Bone formationhttps://pubmed.ncbi.nlm.nih.gov/30067871/PEMF promoted bone formation by activating the sAC-cAMP-PKA-CREB signaling pathway. The study was well-designed and well-conducted, and the results are promising.Pulsed electromagnetic fields (PEMF) promoted bone formation by activating the soluble adenylyl cyclase-cyclic adenosine monophosphate-protein kinase A-cAMP response element-binding protein (sAC-cAMP-PKA-CREB) signaling pathway in osteoblasts.
Breast cancer cell line MCF 7https://pubmed.ncbi.nlm.nih.gov/28668834/Pulsed electromagnetic fields (PEMFs) affected the absorption spectra of MCF 7 breast cancer cells, indicating that PEMFs may interact with the cells and have biological effects. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings and to elucidate the underlying mechanisms.Pulsed electromagnetic fields (PEMF) induced apoptosis and inhibited proliferation of breast cancer cell line MCF 7 by altering the absorption spectra and the expression of apoptotic genes and proteins.
Postoperative pain and inflammationhttps://pubmed.ncbi.nlm.nih.gov/25919263/PEMF reduced postoperative interleukin-1β, pain, and inflammation in TRAM flap breast reconstruction patients. The study was a double-blind, placebo-controlled trial, and the results are statistically significant.Pulsed electromagnetic fields (PEMF) reduced postoperative interleukin-1β, pain, and inflammation in transverse rectus abdominis myocutaneous (TRAM) flap breast reconstruction patients compared to placebo.
Failed digital arthrodesishttps://pubmed.ncbi.nlm.nih.gov/29547026/Percutaneous Kirschner wire repair of failed digital arthrodesis using pulsed electromagnetic field (PEMF) therapy was successful in a 52-year-old male patient. The patient reported complete pain relief and full range of motion in the affected finger at 6 months follow-up. The study was a case report, and more research is needed to confirm the efficacy of PEMF for this condition.Percutaneous Kirschner wire repair of failed digital arthrodesis using PEMF therapy achieved successful fusion and pain relief in a patient with rheumatoid arthritis and failed digital arthrodesis.
Peripheral nerve regenerationhttps://pubmed.ncbi.nlm.nih.gov/26991921/Schwann-like cells differentiated from human dental pulp stem cells combined with PEMF improved peripheral nerve regeneration in a rat model of peripheral nerve injury.Schwann-like cells differentiated from human dental pulp stem cells combined with PEMF may be a promising treatment for peripheral nerve injury.
Postnatal carpal tunnel syndromehttps://pubmed.ncbi.nlm.nih.gov/27980864/PEMF was as effective as ultrasound in the treatment of postnatal carpal tunnel syndrome.PEMF may be an effective treatment for postnatal carpal tunnel syndrome.
Histomorphometry and osteocalcin in disuse osteoporosis ratshttps://pubmed.ncbi.nlm.nih.gov/28582887/PEMF improved histomorphometry and increased osteocalcin levels in disuse osteoporosis rats.PEMF may be a promising treatment to prevent bone loss in conditions such as bed rest and spaceflight.
Primary dysmenorrheahttps://pubmed.ncbi.nlm.nih.gov/29184281/High-intensity laser therapy and pulsed electromagnetic field therapy were both effective in reducing pain and improving quality of life in patients with primary dysmenorrhea.High-intensity laser therapy and pulsed electromagnetic field therapy may be effective treatments for primary dysmenorrhea.
Proliferation, tissue-specific gene expression, and cytokines release of human tendon cellshttps://pubmed.ncbi.nlm.nih.gov/23345006/PEMF affected the proliferation, tissue-specific gene expression, and cytokines release of human tendon cells.PEMF may be a promising treatment to promote tendon healing.
Osteogenesis of MC3T3-E1 cellshttps://pubmed.ncbi.nlm.nih.gov/24907521/Different pulse frequencies of PEMF had different effects on osteogenesis of MC3T3-E1 cells.More research is needed to optimize PEMF parameters for promoting bone regeneration.
Benign prostatic hyperplasia (BPH)https://pubmed.ncbi.nlm.nih.gov/28878453/Significant improvement in urinary symptoms and reduction in prostate volumePulsed electromagnetic field therapy was shown to be an effective treatment for BPH, improving urinary symptoms and reducing prostate volume in patients.
Early knee osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/36922781/Laser therapy and pulsed electromagnetic field therapy were both effective in reducing pain and improving function in patients with early knee osteoarthritis.Laser therapy and pulsed electromagnetic field therapy may be effective treatments for early knee osteoarthritis.
Pulsed electromagnetic field exposure at 3 GHzhttps://pubmed.ncbi.nlm.nih.gov/27436662/Pulsed electromagnetic field exposure at 3 GHz did not cause any significant adverse effects on human cells.Pulsed electromagnetic field exposure at 3 GHz may be safe for use in medical devices.
Resting blood pressure in aging adultshttps://pubmed.ncbi.nlm.nih.gov/23675619/Pulsed electromagnetic field therapy (PEMFT) reduced resting blood pressure in aging adults. The study was well-designed and well-conducted, and the results are statistically significant.Pulsing electromagnetic field (PEMF) therapy reduced resting blood pressure in aging adults by improving endothelial function and reducing oxidative stress.
Rotator cuff diseasehttps://pubmed.ncbi.nlm.nih.gov/27283591/Electrotherapy modalities, such as transcutaneous electrical nerve stimulation (TENS), pulsed electromagnetic fields (PEMF), and ultrasound, are often used for the treatment of rotator cuff disease, a condition that affects the tendons and muscles of the shoulder joint. The review assesses the evidence of electrotherapy modalities for rotator cuff disease and provides recommendations for clinical practice.Electrotherapy modalities may be effective treatments for rotator cuff disease, but more research is needed to determine the optimal parameters and protocols of electrotherapy modalities.
Rotator cuff tendon-to-bone healinghttps://pubmed.ncbi.nlm.nih.gov/29174271/Pulsed electromagnetic field therapy at different frequencies and durations was effective in promoting rotator cuff tendon-to-bone healing in a rat model.Pulsed electromagnetic field therapy may be a promising treatment for rotator cuff tendon-to-bone injury.
Rotator cuff repairhttps://pubmed.ncbi.nlm.nih.gov/27282093/PEMF therapy improved tendon-to-bone healing in a rat rotator cuff repair model.PEMF therapy may be a promising treatment to improve the outcomes of rotator cuff repair surgery.
Rotator cuff tearshttps://pubmed.ncbi.nlm.nih.gov/27138553/PEMF promoted the proliferation and differentiation of tenocytes and myoblasts in vitro.PEMF may be a promising treatment for rotator cuff tears.
Sciatic nerve regenerationhttps://pubmed.ncbi.nlm.nih.gov/28360885/PEMF promoted sciatic nerve regeneration in rats, and the effect was most pronounced when PEMF was applied at the circadian peak of nerve regeneration activity. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in humans.
Severe haemophilia A with osteoporosishttps://pubmed.ncbi.nlm.nih.gov/24249841/This study evaluated the effects of short-term resistance training and PEMF on bone metabolism and joint function in severe haemophilia A patients with osteoporosis. Haemophilia A is a bleeding disorder caused by a deficiency of factor VIII, a protein involved in blood clotting. Osteoporosis is a condition characterized by low bone mass and increased fracture risk. The study found that resistance training and PEMF improved bone mineral density, bone turnover markers, joint range of motion, and joint pain compared to control.This study demonstrates that resistance training and PEMF are beneficial for bone health and joint function in severe haemophilia A patients with osteoporosis.
Subacromial impingement syndromehttps://pubmed.ncbi.nlm.nih.gov/24217037/Subacromial impingement syndrome (SAIS) is a common shoulder disorder that causes pain and functional impairment. Physiotherapy and manual therapy are commonly used treatments for SAIS, but their effectiveness and optimal protocols are unclear. The review evaluates the evidence of physiotherapy and manual therapy for SAIS and provides recommendations for clinical practice.Physiotherapy and manual therapy may be effective treatments for SAIS, but more research is needed to compare different modalities and protocols.
Spinal fusionhttps://pubmed.ncbi.nlm.nih.gov/25278882/Electrical stimulation enhanced spinal fusion in patients with degenerative disc disease. The study was a systematic review, and the results are statistically significant.Electrical stimulation was effective in enhancing spinal fusion by increasing the fusion rate and reducing the time to fusion. The most effective electrical stimulation modalities were direct current stimulation and PEMF.
Steroid-induced osteonecrosis of the femoral headhttps://pubmed.ncbi.nlm.nih.gov/24421074/PEMF protected the balance between adipogenesis and osteogenesis in rats with steroid-induced osteonecrosis of the femoral head. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in humans.
Survival and neuronal differentiation of human bone marrow mesenchymal stem cellshttps://pubmed.ncbi.nlm.nih.gov/26898125/PEMF promoted the survival and neuronal differentiation of human bone marrow mesenchymal stem cells.PEMF may be a promising treatment to promote nerve regeneration using bone marrow mesenchymal stem cells.
Tendon cell functionhttps://pubmed.ncbi.nlm.nih.gov/24957914/PEMF improved the function of human tendon cells in a dose-dependent manner.PEMF may be a promising treatment for tendon injuries.
Tendon repairhttps://pubmed.ncbi.nlm.nih.gov/30154867/This study investigated the effects of PEMF on the tenogenic commitment of umbilical cord-derived MSCs, a potential source of cells for tendon repair. Tenogenic commitment refers to the process of MSCs differentiating into tenocytes, the cells that form tendons. The study found that PEMF increased the expression of tenogenic genes and proteins, and enhanced the biomechanical properties of MSCs in a tenogenic microenvironment.This study suggests that PEMF can improve the tenogenic commitment of umbilical cord-derived MSCs and may be a potential strategy for tendon repair.
Tendon-to-bone healinghttps://pubmed.ncbi.nlm.nih.gov/33934605/Focused continuous pulsed electromagnetic field therapy promoted early tendon-to-bone healing.Focused continuous pulsed electromagnetic field therapy may be a promising treatment for tendon-to-bone injury.
Orthodontic tooth movementhttps://pubmed.ncbi.nlm.nih.gov/25332914/The review concludes that PEMF may accelerate orthodontic tooth movement, but more research is needed to confirm the findings and to determine the optimal parameters for PEMF treatment.
Total knee arthroplastyhttps://pubmed.ncbi.nlm.nih.gov/24352823/Pulsed electromagnetic field therapy improved pain and function in patients undergoing total knee arthroplasty.Pulsed electromagnetic field therapy may be a promising adjunctive treatment for total knee arthroplasty.
Parkinson’s diseasehttps://pubmed.ncbi.nlm.nih.gov/33909634/Long-term treatment with transcranial pulsed electromagnetic fields (tPEMF) improved movement speed and elevated cerebrospinal erythropoietin in patients with Parkinson’s disease. The study was a small pilot study, and more research is needed to confirm the findings and to elucidate the underlying mechanisms.Long-term treatment with transcranial pulsed electromagnetic fields (T-PEMF) improved movement speed and elevated cerebrospinal erythropoietin in patients with Parkinson’s disease compared to sham stimulation.
Parkinson’s diseasehttps://pubmed.ncbi.nlm.nih.gov/30704504/8 weeks of treatment with tPEMF reduced hand tremor and improved inter-hand coherence in patients with Parkinson’s disease. The study was a small randomized controlled trial, and more research is needed to confirm the findings and to elucidate the underlying mechanisms.Eight weeks of treatment with transcranial pulsed electromagnetic fields (T-PEMF) reduced hand tremor and inter-hand coherence in patients with Parkinson’s disease compared to sham stimulation.
Treatment-resistant depressionhttps://pubmed.ncbi.nlm.nih.gov/20385376/Transcranial low voltage pulsed electromagnetic fields (TL-VPEMF) were effective in improving depressive symptoms in patients with treatment-resistant depression. The study was a small pilot study, and more research is needed to confirm the findings.Transcranial low voltage pulsed electromagnetic fields (TLV-PEMF) improved depressive symptoms and cognitive function in patients with treatment-resistant depression compared to sham stimulation.
Tumor growthhttps://pubmed.ncbi.nlm.nih.gov/23781986/Pulsed electromagnetic and pulsed vector magnetic potential field both inhibited the growth of tumor cells in vitro.PEMF may be a promising treatment for cancer.
Various conditions in equine athleteshttps://pubmed.ncbi.nlm.nih.gov/27012509/PEMF therapy has been shown to be effective in treating a variety of conditions in equine athletes, such as tendon and ligament injuries, bone fractures, and arthritis.PEMF therapy may be a valuable tool for veterinarians treating equine athletes.
Osteogenesis and chondrogenesishttps://pubmed.ncbi.nlm.nih.gov/33467447/The review discusses the signaling pathways through which PEMF stimulates osteogenesis and chondrogenesis. The authors conclude that PEMF is a promising therapeutic option for a variety of bone and cartilage disorders.PEMF may be a promising treatment for a variety of bone and joint disorders.
In vitro electroporationhttps://pubmed.ncbi.nlm.nih.gov/27634482/PEMF assisted in vitro electroporation of cells. The study was well-designed and well-conducted, and the results are promising. However, more research is needed to confirm the findings in vivo.PEMF may be a useful tool for gene and drug delivery.
Work-related chronic stresshttps://pubmed.ncbi.nlm.nih.gov/27430621/A randomized controlled trial will be conducted to evaluate the effect of a coaching program combined with light therapy and pulsed electromagnetic field therapy on return to work in workers with work-related chronic stress.
Work-related chronic stresshttps://pubmed.ncbi.nlm.nih.gov/28969697/A randomized controlled trial will be conducted to evaluate the effect of a coaching program combined with light therapy and PEMF therapy on return to work in workers with work-related chronic stress.The results of this trial will help to determine whether this combined approach is an effective treatment for work-related chronic stress.
Wound healinghttps://pubmed.ncbi.nlm.nih.gov/24395219/PEMF promoted early wound healing and myofibroblast proliferation in diabetic rats.PEMF may be a promising treatment for wound healing in diabetic patients.
β-Adrenergic response in beating cardiomyocyteshttps://pubmed.ncbi.nlm.nih.gov/27418252/PEMF counteracted the β-adrenergic response in beating cardiomyocytes.PEMF may have therapeutic potential for cardiovascular diseases.
Various veterinary conditionshttps://pubmed.ncbi.nlm.nih.gov/29775839/Pulsed electromagnetic field therapy (PEMF) has been used for various veterinary applications, such as wound healing, fracture repair, pain relief, and osteoarthritis. The mechanisms of action and the evidence of efficacy and safety of PEMF are discussed.PEMF may be a useful treatment for various veterinary conditions, but more research is needed to optimize the parameters and protocols of PEMF.
Various types of cancerhttps://pubmed.ncbi.nlm.nih.gov/27748048/PEMF has been shown to have anti-cancer effects by modulating various cellular processes, such as apoptosis, proliferation, invasion, angiogenesis, and immune response. The mechanisms of action and the clinical evidence of PEMF in oncology are reviewed.PEMF may be a promising treatment for cancer, but more research is needed to determine the optimal parameters and combinations of PEMF with other therapies.
Fibromyalgiahttps://pubmed.ncbi.nlm.nih.gov/29709070/PEMF therapy improved pain, fatigue, sleep quality, and physical function in patients with fibromyalgia compared to sham therapy. The treatment was well tolerated and no adverse events were reported.PEMF therapy may be an effective treatment for fibromyalgia.
Varioushttps://pubmed.ncbi.nlm.nih.gov/29662800/This article reviews the current state of equipment for PEMFs and highlights worldwide therapeutic achievements. It also discusses the basic science evidence for the potential mechanisms of action of PEMF in osteogenesis stimulation.This article provides a comprehensive overview of PEMF applications and challenges in the corporate perspective.
High intracranial pressure (ICP)https://pubmed.ncbi.nlm.nih.gov/29492540/PEMF stimulation attenuated high ICP-induced pathological microcirculatory changes, tissue hypoxia, BBB degradation and neuronal necrosis in rats by dilating arterioles, increasing capillary blood flow velocity and reducing MVS/capillary ratio.PEMF stimulation may be a beneficial treatment for improving cerebral blood flow and tissue oxygenation in high ICP conditions.
Diabetic woundshttps://pubmed.ncbi.nlm.nih.gov/29324868/PEMF stimulation improved the energy absorption capacity of diabetic wounds in the early healing phase, but impaired the material properties (maximum stress and Young’s modulus) in the remodeling phase. The 10-mT PEMF group also showed increased wound tissue thickness.PEMF stimulation may be a useful treatment for promoting the recovery of structural properties of diabetic wounds, but it may not be applied in the remodeling phase to avoid impairing the recovery of material properties.
Intervertebral disc degenerationhttps://pubmed.ncbi.nlm.nih.gov/23187450/PEMF was more effective than sham treatment in reducing pain and improving range of motion.PEMF may be an effective treatment for intervertebral disc degeneration.
Male fertilityhttps://pubmed.ncbi.nlm.nih.gov/28323003/PEMF induced apoptosis in mouse spermatogenic cells in vitro.PEMF may have a negative impact on male fertility.
Tendon regenerationhttps://pubmed.ncbi.nlm.nih.gov/26617513/PEMF and extracorporeal shock wave therapy (ESWT) were both effective in promoting tendon regeneration.PEMF and ESWT may be promising treatments for tendon regeneration.
Peripheral nerve injuryhttps://pubmed.ncbi.nlm.nih.gov/25271799/Co-treatment with PEMF and human dental pulp stromal cells and FK506 was more effective than human dental pulp stromal cells and FK506 alone in promoting the regeneration of crush-injured rat sciatic nerve.PEMF may be a promising treatment for peripheral nerve injury.
Osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/29251361/PEMF improved subchondral bone microstructure in knee osteoarthritis rats through a Wnt/β-catenin signaling-associated mechanism.PEMF may be a promising treatment for osteoarthritis.
Peripheral nerve injuryhttps://pubmed.ncbi.nlm.nih.gov/29451219/Low-frequency pulsed electromagnetic field pretreated bone marrow-derived mesenchymal stem cells promoted the regeneration of crush-injured rat mental nerve.PEMF may be a promising treatment for peripheral nerve injury.
Chondrocyte functionhttps://pubmed.ncbi.nlm.nih.gov/26955182/Low dose short duration PEMF had a positive effect on the proliferation and differentiation of cultured human chondrocytes.PEMF may be a promising treatment for chondrocyte-related disorders, such as osteoarthritis.
Shoulder impingement syndromehttps://pubmed.ncbi.nlm.nih.gov/24139986/PEMF and exercises were more effective than sham treatment in reducing pain and improving function in patients with shoulder impingement syndrome.PEMF and exercises may be an effective treatment for shoulder impingement syndrome.
Sciatic nerve injuryhttps://pubmed.ncbi.nlm.nih.gov/33797305/PEMF improved the recovery of nerve function in rats with experimentally induced sciatic nerve injury.PEMF may be a promising treatment for sciatic nerve injury.
Knee osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/28026095/PEMF at different stages of knee osteoarthritis in rats improved subchondral trabecular bone microarchitecture and reduced cartilage degradation.PEMF may be a promising treatment for knee osteoarthritis.
Stem cell differentiationhttps://pubmed.ncbi.nlm.nih.gov/25196518/PEMF promoted the differentiation of HUES-17 human embryonic stem cells into osteoblasts, chondrocytes, and adipocytes.PEMF may be a promising tool for directing stem cell differentiation.
Ischemia/reperfusion injuryhttps://pubmed.ncbi.nlm.nih.gov/27780890/PEMF reduced infarct size and improved cardiac function in rats with ischemia/reperfusion injury.PEMF may be a promising treatment for ischemia/reperfusion injury.
Osteoblast differentiationhttps://pubmed.ncbi.nlm.nih.gov/33630907/PEMF induced osteoblast differentiation through activation of adenosine receptors A2A and A3.PEMF may be a promising treatment to promote bone regeneration.
Osteoblast differentiationhttps://pubmed.ncbi.nlm.nih.gov/28512472/PEMF regulated MicroRNA 21 expression to activate TGF-β signaling in human bone marrow stromal cells, which enhanced osteoblast differentiation.PEMF may be a promising treatment to promote bone regeneration.
Parkinson’s diseasehttps://pubmed.ncbi.nlm.nih.gov/30252895/PEMF improved functional rate of force development and movement speed in persons with Parkinson’s disease.T-PEMF may be a promising treatment for Parkinson’s disease.
Glutamate-induced excitotoxicityhttps://pubmed.ncbi.nlm.nih.gov/28220060/PEMF protected against glutamate-induced excitotoxicity by modulating the endocannabinoid system.PEMF may be a promising treatment for neurodegenerative disorders caused by glutamate-induced excitotoxicity.
Osteoblastogenesishttps://pubmed.ncbi.nlm.nih.gov/29909008/PEMF induced Ca2+-dependent osteoblastogenesis in C3H10T1/2 mesenchymal cells through the Wnt-Ca2+/Wnt-β-catenin signaling pathway.PEMF may be a promising treatment to promote bone regeneration.
Vaginal laxityhttps://pubmed.ncbi.nlm.nih.gov/34327510/Nonablative, noncoagulative multipolar radiofrequency and pulsed electromagnetic field treatment improved vaginal laxity and sexual function in women.Nonablative, noncoagulative multipolar radiofrequency and pulsed electromagnetic field treatment may be an effective treatment for vaginal laxity.
Major depressionhttps://pubmed.ncbi.nlm.nih.gov/29573167/T-PEMF improved quality of life and depressive symptoms in patients with major depression.T-PEMF may be a promising treatment for major depression.
Knee osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/24106421/PEMF therapy was effective in reducing pain, stiffness, and improving physical function in patients with knee osteoarthritis.PEMF therapy may be an effective treatment for knee osteoarthritis.
Glioblastomahttps://pubmed.ncbi.nlm.nih.gov/27793904/PEMF with temozolomide induced an epigenetic pro-apoptotic effect on glioblastoma T98G cells.PEMF with temozolomide may be a promising treatment for glioblastoma.
Mandibular fracturehttps://pubmed.ncbi.nlm.nih.gov/24764277/PEMF treatment enhanced healing callus biomechanical properties in patients with mandibular fracture.PEMF treatment may be a promising treatment to improve the outcomes of mandibular fracture.
Hypoxia and inflammation damage in neuron-like and microglial cellshttps://pubmed.ncbi.nlm.nih.gov/27639248/PEMF exposure reduced hypoxia and inflammation damage in neuron-like and microglial cells.PEMF may be a promising treatment for neurodegenerative disorders caused by hypoxia and inflammation.
Ovariectomy-induced osteoporosis in ratshttps://pubmed.ncbi.nlm.nih.gov/27711964/Combined treatment with ibandronate and pulsed electromagnetic field was more effective than ibandronate alone in preventing bone loss and improving bone microstructure in rats with ovariectomy-induced osteoporosis.Combined treatment with ibandronate and pulsed electromagnetic field may be a promising treatment for osteoporosis.
Angiogenesishttps://pubmed.ncbi.nlm.nih.gov/33675261/PEMF stimulated angiogenesis in human umbilical vein endothelial cells by activating the AKT/mTOR pathway.PEMF may be a promising treatment for enhancing angiogenesis.
Intervertebral disc degenerationhttps://pubmed.ncbi.nlm.nih.gov/24131391/PEMF reduced intervertebral disc cell apoptosis in rats with intervertebral disc degeneration.PEMF may be a promising treatment for intervertebral disc degeneration.
Cartilage degenerationhttps://pubmed.ncbi.nlm.nih.gov/27856169/PEMF ameliorated cartilage degeneration by inhibiting mitogen-activated protein kinases in rats with osteoarthritis.PEMF may be a promising treatment for osteoarthritis.
Food sensitivityhttps://pubmed.ncbi.nlm.nih.gov/24712751/PEMF therapy was effective in reducing food sensitivity symptoms in patients with food sensitivity.PEMF therapy may be a promising treatment for food sensitivity.
Osteoinductionhttps://pubmed.ncbi.nlm.nih.gov/29902240/PEMF exposure promoted the osteoinduction of human mesenchymal stem cells cultured on nano-TiO2 surfaces.PEMF exposure may be a promising way to promote bone regeneration in implants and other biomedical applications.
Multidifferentiation of human mesenchymal stem cellshttps://pubmed.ncbi.nlm.nih.gov/27332788/PEMF exposure enhanced the multidifferentiation of human mesenchymal stem cells cultured on reduced graphene oxide substrates.PEMF exposure may be a promising way to control the differentiation of stem cells for regenerative medicine applications.
Osteoarthritishttps://pubmed.ncbi.nlm.nih.gov/24501089/Two different pulsed electromagnetic field frequencies were both effective in reducing cartilage degeneration and improving function in rats with osteoarthritis.Pulsed electromagnetic field therapy may be a promising treatment for osteoarthritis.
Scroll to Top