Mechanisms and applications of the anti-inflammatory effects of photobiomodulation

Michael R Hamblin, Michael R Hamblin

Abstract

Photobiomodulation (PBM) also known as low-level level laser therapy is the use of red and near-infrared light to stimulate healing, relieve pain, and reduce inflammation. The primary chromophores have been identified as cytochrome c oxidase in mitochondria, and calcium ion channels (possibly mediated by light absorption by opsins). Secondary effects of photon absorption include increases in ATP, a brief burst of reactive oxygen species, an increase in nitric oxide, and modulation of calcium levels. Tertiary effects include activation of a wide range of transcription factors leading to improved cell survival, increased proliferation and migration, and new protein synthesis. There is a pronounced biphasic dose response whereby low levels of light have stimulating effects, while high levels of light have inhibitory effects. It has been found that PBM can produce ROS in normal cells, but when used in oxidatively stressed cells or in animal models of disease, ROS levels are lowered. PBM is able to up-regulate anti-oxidant defenses and reduce oxidative stress. It was shown that PBM can activate NF-kB in normal quiescent cells, however in activated inflammatory cells, inflammatory markers were decreased. One of the most reproducible effects of PBM is an overall reduction in inflammation, which is particularly important for disorders of the joints, traumatic injuries, lung disorders, and in the brain. PBM has been shown to reduce markers of M1 phenotype in activated macrophages. Many reports have shown reductions in reactive nitrogen species and prostaglandins in various animal models. PBM can reduce inflammation in the brain, abdominal fat, wounds, lungs, spinal cord.

Keywords: animal studies; chromophores; clinical trials; inflammation; low-level laser therapy; photobiomodulation.

Conflict of interest statement

Conflict of Interest: The author declares no conflict of interest in this paper.

Figures

Figure 1
Figure 1
Chromophores in PBM. Cytochrome c oxidase in respiratory chain absorbs mainly red (and NIR) light by heme and copper; Heat-gated TRP ion channels absorb NIR (and blue light) via structured water; opsins absorb mainly blue/green light via cis-retinal; flavoproteins and cryptochromes absorb mainly blue light via pterin.
Figure 2
Figure 2
NFkB is activated by PBM induced ROS in embryonic fibroblasts. (A) Intracellular ROS measured by DCDHF fluorescence; (B) NF-kB activation measured by a luciferase assay; (C) NF-kB activation is inhibited by antioxidants; (D) ATP increase is not affected by antioxidants. Figure adapted from data in [34].
Figure 3
Figure 3
Dose response of 810 nm laser in cortical neurons. (A) ATP production as a function of fluence; (B) Mitochondrial membrane potential (JC1 red/green ratio); (C) Mitochondrial ROS. Figure adapted from data in [35].
Figure 4
Figure 4
PBM reduces oxidative stress in cortical neurons. Oxidative stress was induced by three different treatments (cobalt chloride, hydrogen peroxide, rotenone) and cells were treated with 3 J/cm2 810 nm. (A) Mitochondrial ROS, (B) Mitochondrial membrane potential (tetramethylrhodamine methyl ester). Figure adapted from data in [39].
Figure 5
Figure 5
PBM reduces inflammatory markers in activated murine DCs in vitro. (A) Flow cytometry was used to measure MHC class II, CD86, CD11c (dexamethasone was used as positive control); (B) Secreted IL12 measured by ELISA. Figure adapted from data in [44].
Figure 6
Figure 6
Animal models in which the anti-inflammatory effects of PBM have been shown. Acute traumatic brain injury; experimental autoimmune encephalomyelitis; spinal cord injury; wound healing; muscle exercise and recovery; inflammatory pain in paw; abdominal fat; lung inflammation; knee arthritis.
Figure 7
Figure 7
Human clinical indications concentrating on anti-inflammatory effects. Autoimmune thyroiditis; muscle exercise and recovery; Achilles tendinopathy; knee arthritis; psoriasis; alopecia areata.

References

    1. Hamblin MR. History of Low-Level Laser (Light) Therapy. In: Hamblin MR, de Sousa MVP, Agrawal T, editors. Handbook of Low-Level Laser Therapy. Singapore: Pan Stanford Publishing; 2017.
    1. Anders JJ, Lanzafame RJ, Arany PR. Low-level light/laser therapy versus photobiomodulation therapy. Photomed Laser Surg. 2015;33:183–184.
    1. Hamblin MR, de Sousa MVP, Agrawal T. Handbook of Low-Level Laser Therapy. Singapore: Pan Stanford Publishing; 2017.
    1. de Freitas LF, Hamblin MR. Proposed mechanisms of photobiomodulation or low-level light therapy. IEEE J Sel Top Quantum Electron. 2016;22:348–364.
    1. Wang Y, Huang YY, Wang Y, et al. Photobiomodulation (blue and green light) encourages osteoblastic-differentiation of human adipose-derived stem cells: role of intracellular calcium and light-gated ion channels. Sci Rep. 2016;6:33719.
    1. Wang L, Jacques SL, Zheng L. MCML—Monte Carlo modeling of light transport in multi-layered tissues. Comput Meth Prog Bio. 1995;47:131–146.
    1. Huang YY, Chen AC, Carroll JD, et al. Biphasic dose response in low level light therapy. Dose Response. 2009;7:358–383.
    1. Huang YY, Sharma SK, Carroll JD, et al. Biphasic dose response in low level light therapy—an update. Dose Response. 2011;9:602–618.
    1. Mason MG, Nicholls P, Cooper CE. Re-evaluation of the near infrared spectra of mitochondrial cytochrome c oxidase: Implications for non invasive in vivo monitoring of tissues. Biochim Biophys Acta. 2014;1837:1882–1891.
    1. Karu TI, Pyatibrat LV, Kolyakov SF, et al. Absorption measurements of a cell monolayer relevant to phototherapy: reduction of cytochrome c oxidase under near IR radiation. J Photochem Photobiol B. 2005;81:98–106.
    1. Karu TI. Multiple roles of cytochrome c oxidase in mammalian cells under action of red and IR-A radiation. IUBMB Life. 2010;62:607–610.
    1. Wong-Riley MT, Liang HL, Eells JT, et al. Photobiomodulation directly benefits primary neurons functionally inactivated by toxins: role of cytochrome c oxidase. J Biol Chem. 2005;280:4761–4771.
    1. Lane N. Cell biology: power games. Nature. 2006;443:901–903.
    1. Pannala VR, Camara AK, Dash RK. Modeling the detailed kinetics of mitochondrial cytochrome c oxidase: Catalytic mechanism and nitric oxide inhibition. J Appl Physiol. 2016;121:1196–1207.
    1. Fernandes AM, Fero K, Driever W, et al. Enlightening the brain: linking deep brain photoreception with behavior and physiology. Bioessays. 2013;35:775–779.
    1. Poletini MO, Moraes MN, Ramos BC, et al. TRP channels: a missing bond in the entrainment mechanism of peripheral clocks throughout evolution. Temperature. 2015;2:522–534.
    1. Caterina MJ, Pang Z. TRP channels in skin biology and pathophysiology. Pharmaceuticals. 2016;9:77.
    1. Montell C. The history of TRP channels, a commentary and reflection. Pflugers Arch. 2011;461:499–506.
    1. Smani T, Shapovalov G, Skryma R, et al. Functional and physiopathological implications of TRP channels. Biochim Biophys Acta. 2015;1853:1772–1782.
    1. Cronin MA, Lieu MH, Tsunoda S. Two stages of light-dependent TRPL-channel translocation in Drosophila photoreceptors. J Cell Sci. 2006;119:2935–2944.
    1. Sancar A. Cryptochrome: the second photoactive pigment in the eye and its role in circadian photoreception. Annu Rev Biochem. 2000;69:31–67.
    1. Weber S. Light-driven enzymatic catalysis of DNA repair: a review of recent biophysical studies on photolyase. Biochim Biophys Acta. 2005;1707:1–23.
    1. Gillette MU, Tischkau SA. Suprachiasmatic nucleus: the brain's circadian clock. Recent Prog Horm Res. 1999;54:33–58.
    1. Kofuji P, Mure LS, Massman LJ, et al. Intrinsically photosensitive petinal ganglion cells (ipRGCs) are necessary for light entrainment of peripheral clocks. PLoS One. 2016;11:e0168651.
    1. Sexton T, Buhr E, Van Gelder RN. Melanopsin and mechanisms of non-visual ocular photoreception. J Biol Chem. 2012;287:1649–1656.
    1. Ho MW. Illuminating water and life: Emilio Del Giudice. Electromagn Biol Med. 2015;34:113–122.
    1. Inoue S, Kabaya M. Biological activities caused by far-infrared radiation. Int J Biometeorol. 1989;33:145–150.
    1. Damodaran S. Water at biological phase boundaries: its role in interfacial activation of enzymes and metabolic pathways. Subcell Biochem. 2015;71:233–261.
    1. Chai B, Yoo H, Pollack GH. Effect of radiant energy on near-surface water. J Phys Chem B. 2009;113:13953–13958.
    1. Pollack GH, Figueroa X, Zhao Q. Molecules, water, and radiant energy: new clues for the origin of life. Int J Mol Sci. 2009;10:1419–1429.
    1. Sommer AP, Haddad M, Fecht HJ. Light effect on water viscosity: implication for ATP biosynthesis. Sci Rep. 2015;5:12029.
    1. FDA. Code of Federal Regulations 21CFR890.5500. 2016;8 Title 21.
    1. Chen ACH, Huang YY, Arany PR, et al. Role of reactive oxygen species in low level light therapy. Proc SPIE. 2009;7165:716502–716511.
    1. Chen AC, Arany PR, Huang YY, et al. Low-level laser therapy activates NF-kB via generation of reactive oxygen species in mouse embryonic fibroblasts. PLoS One. 2011;6:e22453.
    1. Sharma SK, Kharkwal GB, Sajo M, et al. Dose response effects of 810 nm laser light on mouse primary cortical neurons. Lasers Surg Med. 2011;43:851–859.
    1. Tatmatsu-Rocha JC, Ferraresi C, Hamblin MR, et al. Low-level laser therapy (904 nm) can increase collagen and reduce oxidative and nitrosative stress in diabetic wounded mouse skin. J Photochem Photobiol B. 2016;164:96–102.
    1. De Marchi T, Leal Junior EC, Bortoli C, et al. Low-level laser therapy (LLLT) in human progressive-intensity running: effects on exercise performance, skeletal muscle status, and oxidative stress. Lasers Med Sci. 2012;27:231–236.
    1. Fillipin LI, Mauriz JL, Vedovelli K, et al. Low-level laser therapy (LLLT) prevents oxidative stress and reduces fibrosis in rat traumatized Achilles tendon. Lasers Surg Med. 2005;37:293–300.
    1. Huang YY, Nagata K, Tedford CE, et al. Low-level laser therapy (LLLT) reduces oxidative stress in primary cortical neurons in vitro. J Biophotonics. 2013;6:829–838.
    1. Hervouet E, Cizkova A, Demont J, et al. HIF and reactive oxygen species regulate oxidative phosphorylation in cancer. Carcinogenesis. 2008;29:1528–1537.
    1. Madungwe NB, Zilberstein NF, Feng Y, et al. Critical role of mitochondrial ROS is dependent on their site of production on the electron transport chain in ischemic heart. Am J Cardiovasc Dis. 2016;6:93–108.
    1. Martins DF, Turnes BL, Cidral-Filho FJ, et al. Light-emitting diode therapy reduces persistent inflammatory pain: Role of interleukin 10 and antioxidant enzymes. Neuroscience. 2016;324:485–495.
    1. Macedo AB, Moraes LH, Mizobuti DS, et al. Low-level laser therapy (LLLT) in dystrophin-deficient muscle cells: effects on regeneration capacity, inflammation response and oxidative stress. PLoS One. 2015;10:e0128567.
    1. Chen AC, Huang YY, Sharma SK, et al. Effects of 810-nm laser on murine bone-marrow-derived dendritic cells. Photomed Laser Surg. 2011;29:383–389.
    1. Yamaura M, Yao M, Yaroslavsky I, et al. Low level light effects on inflammatory cytokine production by rheumatoid arthritis synoviocytes. Lasers Surg Med. 2009;41:282–290.
    1. Hwang MH, Shin JH, Kim KS, et al. Low level light therapy modulates inflammatory mediators secreted by human annulus fibrosus cells during intervertebral disc degeneration in vitro. Photochem Photobiol. 2015;91:403–410.
    1. Imaoka A, Zhang L, Kuboyama N, et al. Reduction of IL-20 expression in rheumatoid arthritis by linear polarized infrared light irradiation. Laser Ther. 2014;23:109–114.
    1. Lim W, Choi H, Kim J, et al. Anti-inflammatory effect of 635 nm irradiations on in vitro direct/indirect irradiation model. J Oral Pathol Med. 2015;44:94–102.
    1. Choi H, Lim W, Kim I, et al. Inflammatory cytokines are suppressed by light-emitting diode irradiation of P. gingivalis LPS-treated human gingival fibroblasts: inflammatory cytokine changes by LED irradiation. Lasers Med Sci. 2012;27:459–467.
    1. Sakurai Y, Yamaguchi M, Abiko Y. Inhibitory effect of low-level laser irradiation on LPS-stimulated prostaglandin E2 production and cyclooxygenase-2 in human gingival fibroblasts. Eur J Oral Sci. 2000;108:29–34.
    1. Nomura K, Yamaguchi M, Abiko Y. Inhibition of interleukin-1beta production and gene expression in human gingival fibroblasts by low-energy laser irradiation. Lasers Med Sci. 2001;16:218–223.
    1. Briken V, Mosser DM. Editorial: switching on arginase in M2 macrophages. J Leukoc Biol. 2011;90:839–841.
    1. Whyte CS, Bishop ET, Ruckerl D, et al. Suppressor of cytokine signaling (SOCS)1 is a key determinant of differential macrophage activation and function. J Leukoc Biol. 2011;90:845–854.
    1. Xu H, Wang Z, Li J, et al. The polarization states of microglia in TBI: A new paradigm for pharmacological intervention. Neural Plast. 2017;2017:5405104.
    1. Lu J, Xie L, Liu C, et al. PTEN/I3k/AKT regulates macrophage polarization in emphysematous mice. Scand J Immunol 2017
    1. Saha B, Kodys K, Szabo G. Hepatitis C virus-induced monocyte differentiation into polarized M2 macrophages promotes stellate cell activation via TGF-beta. Cell Mol Gastroenterol Hepatol. 2016;2:302–316.
    1. Fernandes KP, Souza NH, Mesquita-Ferrari RA, et al. Photobiomodulation with 660-nm and 780-nm laser on activated J774 macrophage-like cells: Effect on M1 inflammatory markers. J Photochem Photobiol B. 2015;153:344–351.
    1. Silva IH, de Andrade SC, de Faria AB, et al. Increase in the nitric oxide release without changes in cell viability of macrophages after laser therapy with 660 and 808 nm lasers. Lasers Med Sci. 2016;31:1855–1862.
    1. von Leden RE, Cooney SJ, Ferrara TM, et al. 808 nm wavelength light induces a dose-dependent alteration in microglial polarization and resultant microglial induced neurite growth. Lasers Surg Med. 2013;45:253–263.
    1. Sousa KB, de Santana Araujo L, Pedroso NM, et al. Photobiomodulation effects on gene and protein expression of proinflammatory chemokines and cytokines by J774 macrophages polarized to M1 phenotype. Lasers Surg Med. 2017;49:36.
    1. de Lima FJ, de Oliveira Neto OB, Barbosa FT, et al. Is there a protocol in experimental skin wounds in rats using low-level diode laser therapy (LLDLT) combining or not red and infrared wavelengths? Systematic review. Lasers Med Sci. 2016;31:779–787.
    1. Tchanque-Fossuo CN, Ho D, Dahle SE, et al. Low-level light therapy for treatment of diabetic foot ulcer: a review of clinical experiences. J Drugs Dermatol. 2016;15:843–848.
    1. Gupta A, Keshri GK, Yadav A, et al. Superpulsed (Ga-As, 904 nm) low-level laser therapy (LLLT) attenuates inflammatory response and enhances healing of burn wounds. J Biophotonics. 2015;8:489–501.
    1. Weylandt KH, Chiu CY, Gomolka B, et al. Omega-3 fatty acids and their lipid mediators: towards an understanding of resolvin and protectin formation. Prostag Oth Lipid M. 2012;97:73–82.
    1. Tang Y, Zhang MJ, Hellmann J, et al. Proresolution therapy for the treatment of delayed healing of diabetic wounds. Diabetes. 2013;62:618–627.
    1. Bohr S, Patel SJ, Sarin D, et al. Resolvin D2 prevents secondary thrombosis and necrosis in a mouse burn wound model. Wound Repair Regen. 2013;21:35–43.
    1. Castano AP, Dai T, Yaroslavsky I, et al. Low-level laser therapy for zymosan-induced arthritis in rats: Importance of illumination time. Lasers Surg Med. 2007;39:543–550.
    1. Moriyama Y, Moriyama EH, Blackmore K, et al. In vivo study of the inflammatory modulating effects of low-level laser therapy on iNOS expression using bioluminescence imaging. Photochem Photobiol. 2005;81:1351–1355.
    1. Pallotta RC, Bjordal JM, Frigo L, et al. Infrared (810-nm) low-level laser therapy on rat experimental knee inflammation. Lasers Med Sci. 2012;27:71–78.
    1. Ferraresi C, Hamblin MR, Parizotto NA. Low-level laser (light) therapy (LLLT) on muscle tissue: performance, fatigue and repair benefited by the power of light. Photonics Lasers Med. 2012;1:267–286.
    1. Ferraresi C, Huang YY, Hamblin MR. Photobiomodulation in human muscle tissue: an advantage in sports performance? J Biophotonics 2016
    1. Ferraresi C, de Sousa MV, Huang YY, et al. Time response of increases in ATP and muscle resistance to fatigue after low-level laser (light) therapy (LLLT) in mice. Lasers Med Sci. 2015;30:1259–1267.
    1. Silveira PC, Scheffer Dda L, Glaser V, et al. Low-level laser therapy attenuates the acute inflammatory response induced by muscle traumatic injury. Free Radic Res. 2016;50:503–513.
    1. Pires de Sousa MV, Ferraresi C, Kawakubo M, et al. Transcranial low-level laser therapy (810 nm) temporarily inhibits peripheral nociception: photoneuromodulation of glutamate receptors, prostatic acid phophatase, and adenosine triphosphate. Neurophotonics. 2016;3:015003.
    1. de Sousa MV, Ferraresi C, de Magalhaes AC, et al. Building, testing and validating a set of home-made von Frey filaments: A precise, accurate and cost effective alternative for nociception assessment. J Neurosci Methods. 2014;232:1–5.
    1. Kobiela Ketz A, Byrnes KR, Grunberg NE, et al. Characterization of Macrophage/Microglial activation and effect of photobiomodulation in the spared nerve injury model of neuropathic pain. Pain Med. 2016:pnw144.
    1. Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain. 2000;87:149–158.
    1. de Lima FM, Vitoretti L, Coelho F, et al. Suppressive effect of low-level laser therapy on tracheal hyperresponsiveness and lung inflammation in rat subjected to intestinal ischemia and reperfusion. Lasers Med Sci. 2013;28:551–564.
    1. Silva VR, Marcondes P, Silva M, et al. Low-level laser therapy inhibits bronchoconstriction, Th2 inflammation and airway remodeling in allergic asthma. Respir Physiol Neurobiol. 2014;194:37–48.
    1. Rigonato-Oliveira N, Brito A, Vitoretti L, et al. Effect of low-level laser therapy on chronic lung inflammation in experimental model of asthma: A comparative study of doses. Lasers Surg Med. 2017;49:36.
    1. Huang YY, Gupta A, Vecchio D, et al. Transcranial low level laser (light) therapy for traumatic brain injury. J Biophotonics. 2012;5:827–837.
    1. Thunshelle C, Hamblin MR. Transcranial low-level laser (light) therapy for brain injury. Photomed Laser Surg. 2016;34:587–598.
    1. Hamblin MR. Shining light on the head: Photobiomodulation for brain disorders. BBA Clin. 2016;6:113–124.
    1. Khuman J, Zhang J, Park J, et al. Low-level laser light therapy improves cognitive deficits and inhibits microglial activation after controlled cortical impact in mice. J Neurotrauma. 2012;29:408–417.
    1. Veronez S, Assis L, Del Campo P, et al. Effects of different fluences of low-level laser therapy in an experimental model of spinal cord injury in rats. Lasers Med Sci. 2017;32:343–349.
    1. Muili KA, Gopalakrishnan S, Eells JT, et al. Photobiomodulation induced by 670 nm light ameliorates MOG35-55 induced EAE in female C57BL/6 mice: a role for remediation of nitrosative stress. PLoS One. 2013;8:e67358.
    1. Yoshimura TM, Sabino CP, Ribeiro MS. Photobiomodulation reduces abdominal adipose tissue inflammatory infiltrate of diet-induced obese and hyperglycemic mice. J Biophotonics. 2016;9:1255–1262.
    1. Bjordal JM, Lopes-Martins RA, Iversen VV. A randomised, placebo controlled trial of low level laser therapy for activated Achilles tendinitis with microdialysis measurement of peritendinous prostaglandin E2 concentrations. Br J Sports Med. 2006;40:76–80.
    1. Hofling DB, Chavantes MC, Juliano AG, et al. Low-level laser therapy in chronic autoimmune thyroiditis: a pilot study. Lasers Surg Med. 2010;42:589–596.
    1. Hofling DB, Chavantes MC, Juliano AG, et al. Low-level laser in the treatment of patients with hypothyroidism induced by chronic autoimmune thyroiditis: a randomized, placebo-controlled clinical trial. Lasers Med Sci. 2013;28:743–753.
    1. Hofling DB, Chavantes MC, Juliano AG, et al. Assessment of the effects of low-level laser therapy on the thyroid vascularization of patients with autoimmune hypothyroidism by color Doppler ultrasound. ISRN Endocrinol. 2012;2012:126720.
    1. Hofling DB, Chavantes MC, Acencio MM, et al. Effects of low-level laser therapy on the serum TGF-beta1 concentrations in individuals with autoimmune thyroiditis. Photomed Laser Surg. 2014;32:444–449.
    1. Hofling D, Chavantes MC, Buchpiguel CA, et al. Long-term follow-up of patients with hypothyroidism induced by autoimmune thyroiditis submitted to low-level laser therapy. Lasers Surg Med. 2017;49:36.
    1. Ferraresi C, Beltrame T, Fabrizzi F, et al. Muscular pre-conditioning using light-emitting diode therapy (LEDT) for high-intensity exercise: a randomized double-blind placebo-controlled trial with a single elite runner. Physiother Theory Pract. 2015:1–8.
    1. Ferraresi C, Dos Santos RV, Marques G, et al. Light-emitting diode therapy (LEDT) before matches prevents increase in creatine kinase with a light dose response in volleyball players. Lasers Med Sci. 2015;30:1281–1287.
    1. Pinto HD, Vanin AA, Miranda EF, et al. Photobiomodulation therapy improves performance and accelerates recovery of high-level rugby players in field test: A randomized, crossover, double-blind, placebo-controlled clinical study. J Strength Cond Res. 2016;30:3329–3338.
    1. Ferraresi C, Bertucci D, Schiavinato J, et al. Effects of light-emitting diode therapy on muscle hypertrophy, gene expression, performance, damage, and delayed-onset muscle soreness: case-control study with a pair of identical twins. Am J Phys Med Rehabil. 2016;95:746–757.
    1. Johnston A, Xing X, Wolterink L, et al. IL-1 and IL-36 are dominant cytokines in generalized pustular psoriasis. J Allergy Clin Immunol 2016
    1. Ablon G. Combination 830-nm and 633-nm light-emitting diode phototherapy shows promise in the treatment of recalcitrant psoriasis: preliminary findings. Photomed Laser Surg. 2010;28:141–146.
    1. Choi M, Na SY, Cho S, et al. Low level light could work on skin inflammatory disease: a case report on refractory acrodermatitis continua. J Korean Med Sci. 2011;26:454–456.
    1. Hamblin MR. Can osteoarthritis be treated with light? Arthritis Res Ther. 2013;15:120.
    1. Ip D. Does addition of low-level laser therapy (LLLT) in conservative care of knee arthritis successfully postpone the need for joint replacement? Lasers Med Sci. 2015;30:2335–2339.
    1. Brosseau L, Robinson V, Wells G, et al. Low level laser therapy (Classes I, II and III) for treating rheumatoid arthritis. Cochrane Database Syst Rev. 2005;19:CD002049.
    1. Brosseau L, Welch V, Wells G, et al. Low level laser therapy (Classes I, II and III) for treating osteoarthritis. Cochrane Database Syst Rev. 2004:CD002046.
    1. Barabas K, Bakos J, Zeitler Z, et al. Effects of laser treatment on the expression of cytosolic proteins in the synovium of patients with osteoarthritis. Lasers Surg Med. 2014;46:644–649.
    1. Gregoriou S, Papafragkaki D, Kontochristopoulos G, et al. Cytokines and other mediators in alopecia areata. Mediators Inflamm. 2010;2010:928030.
    1. Avci P, Gupta GK, Clark J, et al. Low-level laser (light) therapy (LLLT) for treatment of hair loss. Lasers Surg Med. 2014;46:144–151.
    1. Gupta AK, Foley KA. A critical assessment of the evidence for low-level laser therapy in the treatment of hair loss. Dermatol Surg. 2017;43:188–197.
    1. Yamazaki M, Miura Y, Tsuboi R, et al. Linear polarized infrared irradiation using Super Lizer is an effective treatment for multiple-type alopecia areata. Int J Dermatol. 2003;42:738–740.

Source: PubMed

3
Prenumerera