IMR Press / JIN / Volume 23 / Issue 5 / DOI: 10.31083/j.jin2305092
Open Access Opinion
Photobiomodulation Therapy: A Novel Therapeutic Approach to Alzheimer's Disease Made Possible by the Evidence of a Brain–Gut Interconnection
Show Less
1 REGEnLIFE, 75008 Paris, France
2 FRconsulting, 34800 Clermont l’Hérault, France
3 Vaiomer, 31670 Labège, France
4 Faculty of Medicine, University of Montpellier, 34000, Montpellier, France
*Correspondence: blivet_guillaume@yahoo.fr (Guillaume Blivet)
J. Integr. Neurosci. 2024, 23(5), 92; https://doi.org/10.31083/j.jin2305092
Submitted: 20 December 2023 | Revised: 6 February 2024 | Accepted: 21 February 2024 | Published: 30 April 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

The evidence of brain–gut interconnections in Alzheimer’s disease (AD) opens novel avenues for the treatment of a pathology for which no definitive treatment exists. Gut microbiota and bacterial translocation may produce peripheral inflammation and immune modulation, contributing to brain amyloidosis, neurodegeneration, and cognitive deficits in AD. The gut microbiota can be used as a potential therapeutic target in AD. In particular, photobiomodulation (PBM) can affect the interaction between the microbiota and the immune system, providing a potential explanation for its restorative properties in AD-associated dysbiosis. PBM is a safe, non-invasive, non-ionizing, and non-thermal therapy that uses red or near-infrared light to stimulate the cytochrome c oxidase (CCO, complex IV), the terminal enzyme of the mitochondrial electron transport chain, resulting in adenosine triphosphate synthesis. The association of the direct application of PBM to the head with an abscopal and a systemic treatment through simultaneous application to the abdomen provides an innovative therapeutic approach to AD by targeting various components of this highly complex pathology. As a hypothesis, PBM might have a significant role in the therapeutic options available for the treatment of AD.

Keywords
Alzheimer's disease
neurodegeneration
neuroinflammation
brain–gut axis
microbiota
microbiome
photobiomodulation
low-level laser therapy (LLLT)
oxidative stress
mitochondria
1. Introduction

Recent treatments such as Food and Drug Administration (FDA)-approved anti-amyloid-β monoclonal antibodies have been shown to slow the progression of Alzheimer’s disease (AD) in patients. However, this benefit comes with an increased risk of adverse effects [1, 2]. There is a pressing need for novel treatments for AD ideally offering reduced adverse effects and increased cost-effectiveness. In recent decades, human microbiota have attracted more research attention, and the gut–microbiome–brain axis has been demonstrated to regulate multiple neurophysiological responses [3, 4, 5] through interactions among the autonomic nervous system, enteric neural system, central nervous system (CNS), immune system, and endocrine system. Probiotic supplementation has been used in the treatment of multiple CNS-related diseases and has demonstrated the modulation of numerous genes in the brain [6] with consequences for inflammatory and neuronal processes.

In AD, patients display altered microbial diversity and composition, as indicated by fecal analysis compared with controls [7]. The observed shifts in the composition of the microbiome may lead to an inflammatory condition in the intestine that degrades the epithelial barrier [8]. This could result in an increased translocation of proinflammatory products and bacterial molecules triggering autoimmunity, such as liposaccharides, amyloids, DNA, proteins, and polysaccharides. The systemic inflammation leads to microglia activation, neuroinflammation, and blood–brain barrier impairment [9]. Photobiomodulation (PBM) allows simultaneous transcranial and abdominal application, reaching several important targets for the treatment of such a complex pathology. Transcranial PBM, acting via the cytochrome c oxidase (CCO), could increase adenosine triphosphate (ATP) and influence downstream cellular signaling to reduce oxidative stress and neuroinflammation as well as upregulate synaptogenesis and neurogenesis [10]. Abdominal PBM application could restore mitochondrial normal function in gut neurons. Its potential restorative effects on the gut–microbiome–brain axis may have a significant effect on immune modulation through a reduction of oxidative stress, a decrease in proinflammatory cytokines, and changes in macrophage phenotype [11]. The local effect of PBM on inflammatory pathways most likely has systemic consequences. Circulating immune cells (mast cells, macrophages, etc.), stimulated by PBM [12, 13, 14, 15], could transduce protective signals from distal tissues such as the gut to sites in need of protection such as the brain.

The mechanisms considered in this opinion paper are involved in brain–gut interconnections, evidencing PBM as a potential treatment of AD and allowing synergies through multitarget approaches to this multifactorial disease.

2. The Brain–Microbiome–Gut Axis and AD

There is increasing evidence for the contribution of gut microbiota (GM) to the pathogenesis of AD, and it has already been found that AD patients have altered microbiota diversity [16]. Furthermore, GM have been demonstrated as an important player in insulin resistance and type 2 diabetes mellitus [17, 18], which are known to be more frequent in AD patients [19].

Bacterial byproducts such as short-chain fatty acids (SCFAs) exert numerous neuromodulation effects and act directly on gastrointestinal cells, stimulating the synthesis of hormones such as leptin, ghrelin, and glucagon-like peptide 1, peptide YY [20]. These hormones have been shown to exhibit neuroprotective effects [21]. The microbiome has a role in tryptophan metabolism, producing tryptophan catabolites and also other metabolites including neurotransmitters and hormones able to leave the gut lumen and be detected in the circulation to serve as signaling molecules such as catecholamines, serotonin, gamma aminobutyric acid, dopamine, acetylcholine α-melanocyte stimulation hormone (α-MSH), norepinephrine, and melatonin [22, 23, 24]. The afferent neurons of the enteric nervous system can be activated or stimulated by bacteria. The vagal nerve plays a crucial role in facilitating direct neural communication between the gut and the brain [25].

As the source of a large amount of bacterial product, GM may contribute through the disruption of physiological barriers to systemic inflammation and autoimmunity [26]. Bacteria or their products can translocate from the gastrointestinal tract to the CNS. Bacterial amyloids [27] may act as prion protein cross-seeding of misfolding and enhance native amyloid aggregation. Moreover, GM products may prime microglia, enhancing the inflammatory response in the CNS, which, in turn, results in pathologic microglial function, increased neurotoxicity, and impaired amyloid clearance [28].

GM may promote brain inflammation in AD brains and be responsible for the inflammatory reaction featured around amyloid plaques. The possible role of GM was investigated in cognitively impaired AD patients by studying the association of brain amyloidosis, GM taxa with pro-inflammatory or anti-inflammatory properties, and peripheral inflammation [29]. Cognitively impaired patients with amyloidosis revealed higher expression levels of blood pro-inflammatory cytokines such as interleukine-6 (IL-6), chemokine ligand 2 (CXCL2), nucleotide oligomerization domain (NOD)-like receptor protein 3 (NLRP3), and interleukin-1β (IL-1β) and a reduction of the anti-inflammatory cytokine interleukin-10 (IL-10) compared with both controls and cognitively impaired patients without amyloidosis. The pro-inflammatory cytokines IL-1β, NLRP3, and CXCL2 were positively correlated to the relative abundance of the Escherichia-Shigella genus, which includes mostly species known for their pro-inflammatory properties, but negatively correlated to Eubacterium rectale species, known for its anti-inflammatory properties. This indicates a possible association of a peripheral inflammatory state in patients with cognitive impairment and brain amyloidosis. Thus, GM may be responsible for peripheral inflammation, favoring brain amyloidosis and, as a possible consequence, neurodegeneration and cognitive impairment in AD.

3. Gut Microbiome Modification as a Therapeutic Approach for the Treatment of AD

Modifying the gut microbiome exhibits promise in treating AD and other neurological conditions [30, 31]. While interventions like diet, probiotics, and fecal microbiota transplantation (FMT) have had some success, they may not be sufficient for a complete treatment. Recently, it has been demonstrated in aged rats that the mixture VSL#3 containing eight strains of probiotics modulates the expression of several genes in the brain cortex, with positive inflammatory and neuronal consequences [6]. A recent clinical phase-3 trial [32] has shown that GV-971, a sodium oligomannate that is able to remodel gut microbiota, suppressing gut dysbiosis and the associated phenylalanine/isoleucine accumulation, reverses cognition impairment in patients with mild cognitive impairment due to AD [33]. FMT, approved for certain intestinal diseases including recurrent Clostridium difficile infection [34], is being explored for neurodegenerative diseases such as Parkinson’s and other non-intestinal disorders [35]. Studies suggest FMT can improve cognitive symptoms in AD patients [36, 37]. Achieving a healthy microbiome seems crucial for balancing key compounds and influencing the progression of neurodegenerative diseases. However, clinical trials are still lacking and are essential for more conclusive results.

4. PBM Effects on the Brain and Microbiome

The mechanisms involved in PBM exposure to produce its positive effects on AD symptoms are not fully understood. Transcranial PBM for stimulation of the brain in AD patients has shown improvement of cognitive functions [38, 39, 40], quality of life and patient independence [41], and enhancement of prefrontal oxygenation [39, 42].

However, the exact mechanism by which light interacts with the microbiome remains to be elucidated. Beyond the chromophores located in mammalian cells, which could respond to PBM, there is also a diverse range of bacterial species (both Gram-positive and Gram-negative) and fungal (including yeast) cells that have been demonstrated to respond to PBM [43, 44]. In general, an increased proliferation of the microbial cells was observed, but at higher doses, inhibition was also seen, resulting from a biphasic dose-response curve of the PBM [45, 46]. In vitro study [47] has indicated that PBM inhibits the growth of Pseudomonas aeruginosa and Escherichia coli, two Gram-negative bacteria that infect skin ulcers. However, the changes in the microbiome composition observed in the mouse experiments [48] may be due to other effects of PBM on the murine inflammatory system. Indeed, PBM has well-known anti-inflammatory and redox signaling effects, thus reducing the level of pro-inflammatory cytokines such as IL-6, tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) [49] and changing the activity of macrophages and neutrophils [11]. Importantly, PBM can alter the polarization state of macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 lineage [50].

It has been posited that PBM delivered to the abdomen of healthy mice can significantly modify the gut microbiome composition [51]. Recent data has signified that an Amyloid β (Aβ25-35) peptide central injection in mice produces significant changes in the gut microbiome [48], and the dysbiosis observed in this AD murine model is similar to what has been reported in AD patients [52]. Abdominal PBM application reversed the decline of Firmicutes phylum (Gram-positive), reversed the increase of Tenericutes (Gram-negative) and Bacteroidetes (Gram-negative) phyla [53], and reduced the expression of Deferribacteres produced by Aβ25-35 peptide injections. These results suggest that PBM could reduce the overexpression of Gram-negative bacteria that have been shown responsible for the inflammatory processes occurring in AD [28]. The findings were confirmed in the same mouse model with red and near-infrared light directed to the abdomen [54], resulting in a decrease of the relative abundance of Helicobacter, a genus previously identified as a risk factor for AD [55]. In APP/PS1 mice, a restoration of the microbiome composition was observed after mid-infrared light was directed to the entire body, producing an increase in Akkermansia [56], which is known for its protective effects on amyloid pathology [57].

5. Discussion

During the last several years, more evidence has been accumulated demonstrating the involvement of microbiota in various diseases such as cancer [58], diabetes [59], neurological disorders, and gastrointestinal disorders [60]. Furthermore, the manipulation of the microbiota in the human body can be a strategy for disease treatment. In view of the complexity of AD, the discovery of a single, unique molecule with an unambiguous mechanism of action able to prevent or cure this pathology as found in other cases of drug discovery becomes increasingly elusive. As a result, novel clinical trials are being designed by combining the action of several pathways to obtain stronger effects with fewer side effects [61], and this type of drug development is promoted by the US FDA [62]. In this context, PBM emerges as a therapeutic opportunity because it can target the CNS through transcranial application concomitantly with abscopal effects through abdominal application.

The direct effect of PBM on mitochondria to activate CCO is of importance as a possibility of information exchange exists between the GM and neural mitochondria [18, 63]. Furthermore, there is an increasing amount of data demonstrating the involvement of an aberrant metabolism and defective mitochondrial bioenergetics [64, 65, 66, 67, 68] in AD onset and progression. These mitochondrial dysfunctions trigger impaired synaptic activities in AD such as calcium signaling, synaptic energy, and neurotransmission [69, 70]. Maintaining optimal neuronal and synaptic function is crucial in AD and is closely linked to mitochondria [67, 69, 70].Thus, the suggestion emerges that targeting mitochondria could be a promising approach for developing new treatments. As PBM is strongly hypothesized to target mitochondria function, it may be considered as a novel promising therapeutic tool for treating AD.

Preclinical data obtained in mice in the Aβ25-35model of AD has signaled that daily concomitant application of PBM at a pulsed-wave mode both on the head and abdomen for 10 minutes produced a neuroprotective impact on the neurotoxic effects of Aβ25-35 peptide injection by normalizing all the modified behavioral and biochemical parameters [71]. The efficacy observed with this PBM exposure was not seen when the head or abdomen alone was exposed in similar experimental conditions. GM composition induced by toxic peptide injection was also restored by PBM application to the head and abdomen [48]. A pilot clinical study proved that the therapy is safe and well tolerated and confirmed that dual application of PBM therapy to both the head and the abdomen led to an improvement of cognitive functions in patients with mild to moderate AD [72]. In another clinical study, significant changes in microbiome diversity were observed in a patient after PBM abdominal treatment [73]. A multicentric, double-blind, randomized, sham-controlled, pivotal clinical trial was initiated in June 2023 at the Toulouse University Hospital Gerontopole, which included 108 patients with the National Institute on Aging–Alzheimer’s Association (NIA-AA) clinical diagnosis of AD [74]. The primary endpoint of this ongoing pivotal clinical trial is the evolution of patients’ cognition after 26 weeks of brain–gut PBM therapy, as measured by the Alzheimer’s Disease Assessment Scale–Cognitive Subscale (ADAS-Cog).

Transcranial PBM is emerging as a potential treatment and cognitive enhancement method for various neurodegenerative pathologies. It has also been shown to help increase the potential of pharmacological therapies by modulating the blood–brain barrier permeability, opening innovative avenues for non-invasive therapeutic interventions in the CNS [75]. The evidence that abdominal PBM is able to activate mechanisms of brain neuronal rescue by means of the brain–microbiome–gut axis confirms the interest in associating transcranial to abdominal PBM in clinical practice.

6. Conclusion

PBM appears to be a promising non-invasive, non-pharmacological therapeutic strategy for AD, able to mobilize multiple mechanisms in synergy through the association of transcranial and transabdominal application for optimal treatment efficacy. Due to its affordability, safety profile, and ability to be administered both at home and in hospitals, brain–gut PBM has the potential to become widely accessible and integrated into the treatment of AD.

Author Contributions

FJR and GB performed the literature searches, designed and wrote the paper and contributed to the editorial changes in the manuscript. BL, CR, and JT contributed to its analysis, its critical review, and its final version approval. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

GB is an employee of REGEnLIFE and owns equity in the company. FJR is the director of FR Consulting. BL and CR are employees of Vaiomer. BL is a shareholder of Vaiomer. The authors declare no conflict of interest and the writing is not influenced by this relationship.

References
[1]
Budd Haeberlein S, Aisen PS, Barkhof F, Chalkias S, Chen T, Cohen S, et al. Two Randomized Phase 3 Studies of Aducanumab in Early Alzheimer’s Disease. The Journal of Prevention of Alzheimer’s Disease. 2022; 9: 197–210.
[2]
van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, Gee M, et al. Lecanemab in Early Alzheimer’s Disease. The New England Journal of Medicine. 2023; 388: 9–21.
[3]
Bhattacharjee S, Lukiw WJ. Alzheimer’s disease and the microbiome. Frontiers in Cellular Neuroscience. 2013; 7: 153.
[4]
Kang SS, Jeraldo PR, Kurti A, Miller MEB, Cook MD, Whitlock K, et al. Diet and exercise orthogonally alter the gut microbiome and reveal independent associations with anxiety and cognition. Molecular Neurodegeneration. 2014; 9: 36.
[5]
Wang Y, Kasper LH. The role of microbiome in central nervous system disorders. Brain, Behavior, and Immunity. 2014; 38: 1–12.
[6]
Distrutti E, O’Reilly JA, McDonald C, Cipriani S, Renga B, Lynch MA, et al. Modulation of intestinal microbiota by the probiotic VSL#3 resets brain gene expression and ameliorates the age-related deficit in LTP. PLoS ONE. 2014; 9: e106503.
[7]
Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer’s disease. Scientific Reports. 2017; 7: 13537.
[8]
Heston MB, Hanslik KL, Zarbock KR, Harding SJ, Davenport-Sis NJ, Kerby RL, et al. Gut inflammation associated with age and Alzheimer’s disease pathology: a human cohort study. Scientific Reports. 2023; 13: 18924.
[9]
Lee RL, Funk KE. Imaging blood-brain barrier disruption in neuroinflammation and Alzheimer’s disease. Frontiers in Aging Neuroscience. 2023; 15: 1144036.
[10]
Cassano P, Petrie SR, Hamblin MR, Henderson TA, Iosifescu DV. Review of transcranial photobiomodulation for major depressive disorder: targeting brain metabolism, inflammation, oxidative stress, and neurogenesis. Neurophotonics. 2016; 3: 031404.
[11]
Hamblin MR. Mechanisms and applications of the anti-inflammatory effects of photobiomodulation. AIMS Biophysics. 2017; 4: 337–361.
[12]
Muili KA, Gopalakrishnan S, Meyer SL, Eells JT, Lyons JA. Amelioration of experimental autoimmune encephalomyelitis in C57BL/6 mice by photobiomodulation induced by 670 nm light. PLoS ONE. 2012; 7: e30655.
[13]
Bagheri M, Amini A, Abdollahifar MA, Ghoreishi SK, Piryaei A, Pouriran R, et al. Effects of Photobiomodulation on Degranulation and Number of Mast Cells and Wound Strength in Skin Wound Healing of Streptozotocin-Induced Diabetic Rats. Photomedicine and Laser Surgery. 2018; 36: 415–423.
[14]
Khuman J, Zhang J, Park J, Carroll JD, Donahue C, Whalen MJ. Low-level laser light therapy improves cognitive deficits and inhibits microglial activation after controlled cortical impact in mice. Journal of Neurotrauma. 2012; 29: 408–417.
[15]
Song JW, Li K, Liang ZW, Dai C, Shen XF, Gong YZ, et al. Low-level laser facilitates alternatively activated macrophage/microglia polarization and promotes functional recovery after crush spinal cord injury in rats. Scientific Reports. 2017; 7: 620.
[16]
Faulin TDES, Estadella D. Alzheimer’s disease and its relationship with the microbiota-gut-brain axis. Arquivos De Gastroenterologia. 2023; 60: 144–154.
[17]
Deng K, Shuai M, Zhang Z, Jiang Z, Fu Y, Shen L, et al. Temporal relationship among adiposity, gut microbiota, and insulin resistance in a longitudinal human cohort. BMC Medicine. 2022; 20: 171.
[18]
Zhou Z, Sun B, Yu D, Zhu C. Gut Microbiota: An Important Player in Type 2 Diabetes Mellitus. Frontiers in Cellular and Infection Microbiology. 2022; 12: 834485.
[19]
Burillo J, Marqués P, Jiménez B, González-Blanco C, Benito M, Guillén C. Insulin Resistance and Diabetes Mellitus in Alzheimer’s Disease. Cells. 2021; 10: 1236.
[20]
Bonfili L, Cecarini V, Berardi S, Scarpona S, Suchodolski JS, Nasuti C, et al. Microbiota modulation counteracts Alzheimer’s disease progression influencing neuronal proteolysis and gut hormones plasma levels. Scientific Reports. 2017; 7: 2426.
[21]
Gomes S, Martins I, Fonseca ACRG, Oliveira CR, Resende R, Pereira CMF. Protective effect of leptin and ghrelin against toxicity induced by amyloid-β oligomers in a hypothalamic cell line. Journal of Neuroendocrinology. 2014; 26: 176–185.
[22]
Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nature Communications. 2018; 9: 3294.
[23]
Anderson G, Vaillancourt C, Maes M, Reiter RJ. Breastfeeding and the gut-brain axis: is there a role for melatonin? Biomolecular Concepts. 2017; 8: 185–195.
[24]
Tetel MJ, de Vries GJ, Melcangi RC, Panzica G, O’Mahony SM. Steroids, stress and the gut microbiome-brain axis. Journal of Neuroendocrinology. 2018; 30: e12548.
[25]
Forsythe P, Bienenstock J, Kunze WA. Vagal pathways for microbiome-brain-gut axis communication. Advances in Experimental Medicine and Biology. 2014; 817: 115–133.
[26]
Pei Y, Lu Y, Li H, Jiang C, Wang L. Gut microbiota and intestinal barrier function in subjects with cognitive impairments: a cross-sectional study. Frontiers in Aging Neuroscience. 2023; 15: 1174599.
[27]
Nicastro L, Tükel Ç. Bacterial Amyloids: The Link between Bacterial Infections and Autoimmunity. Trends in Microbiology. 2019; 27: 954–963.
[28]
Sharma A, Martins IJ. The role of Microbiota in the Pathogenesis of Alzheimer’s Disease. Acta Scientific Nutritional Health. 2023; 7: 108–118.
[29]
Cattaneo A, Cattane N, Galluzzi S, Provasi S, Lopizzo N, Festari C, et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiology of Aging. 2017; 49: 60–68.
[30]
Claeysen S. Modulation du microbiote intestinal comme nouvelle voie thérapeutique contre la maladie d’Alzheimer. Revue Neurologique. 2022; 178: S149.
[31]
Bicknell B, Liebert A, Borody T, Herkes G, McLachlan C, Kiat H. Neurodegenerative and Neurodevelopmental Diseases and the Gut-Brain Axis: The Potential of Therapeutic Targeting of the Microbiome. International Journal of Molecular Sciences. 2023; 24: 9577.
[32]
Xiao S, Chan P, Wang T, Hong Z, Wang S, Kuang W, et al. A 36-week multicenter, randomized, double-blind, placebo-controlled, parallel-group, phase 3 clinical trial of sodium oligomannate for mild-to-moderate Alzheimer’s dementia. Alzheimer’s Research & Therapy. 2021; 13: 62.
[33]
Wang X, Sun G, Feng T, Zhang J, Huang X, Wang T, et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Research. 2019; 29: 787–803.
[34]
Paramsothy S, Kamm MA, Kaakoush NO, Walsh AJ, van den Bogaerde J, Samuel D, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet. 2017; 389: 1218–1228.
[35]
Sun MF, Zhu YL, Zhou ZL, Jia XB, Xu YD, Yang Q, et al. Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: Gut microbiota, glial reaction and TLR4/TNF-α signaling pathway. Brain, Behavior, and Immunity. 2018; 70: 48–60.
[36]
Hazan S. Rapid improvement in Alzheimer’s disease symptoms following fecal microbiota transplantation: a case report. The Journal of International Medical Research. 2020; 48: 300060520925930.
[37]
Park SH, Lee JH, Shin J, Kim JS, Cha B, Lee S, et al. Cognitive function improvement after fecal microbiota transplantation in Alzheimer’s dementia patient: a case report. Current Medical Research and Opinion. 2021; 37: 1739–1744.
[38]
Blanco NJ, Maddox WT, Gonzalez-Lima F. Improving executive function using transcranial infrared laser stimulation. Journal of Neuropsychology. 2017; 11: 14–25.
[39]
Holmes E, Barrett DW, Saucedo CL, O’Connor P, Liu H, Gonzalez-Lima F. Cognitive Enhancement by Transcranial Photobiomodulation Is Associated With Cerebrovascular Oxygenation of the Prefrontal Cortex. Frontiers in Neuroscience. 2019; 13: 1129.
[40]
Chan AS, Lee TL, Hamblin MR, Cheung MC. Photobiomodulation Enhances Memory Processing in Older Adults with Mild Cognitive Impairment: A Functional Near-Infrared Spectroscopy Study. Journal of Alzheimer’s Disease. 2021; 83: 1471–1480.
[41]
Nizamutdinov D, Qi X, Berman MH, Dougal G, Dayawansa S, Wu E, et al. Transcranial Near Infrared Light Stimulations Improve Cognition in Patients with Dementia. Aging and Disease. 2021; 12: 954–963.
[42]
Dougal G, Ennaceur A, Chazot PL. Effect of Transcranial Near-Infrared Light 1068 nm Upon Memory Performance in Aging Healthy Individuals: A Pilot Study. Photobiomodulation, Photomedicine, and Laser Surgery. 2021; 39: 654–660.
[43]
Karu TI, Riabykh TP, Fedoseeva GE, Puchkova NI. Effect of He-Ne laser radiation on the chemiluminescence of mouse spleen cells. Radiobiologiia. 1989; 29: 230–234. (In Russian)
[44]
Tiflova OA, Karu TI. Effect of He-Ne laser radiation on the bacteriophage T4-Escherichia coli system. Radiobiologiia. 1989; 29: 278–280.
[45]
Huang YY, Chen ACH, Carroll JD, Hamblin MR. Biphasic dose response in low level light therapy. Dose-Response. 2009; 7: 358–383.
[46]
Huang YY, Sharma SK, Carroll J, Hamblin MR. Biphasic dose response in low level light therapy - an update. Dose-Response. 2011; 9: 602–618.
[47]
de Sousa NTA, Gomes RC, Santos MF, Brandino HE, Martinez R, de Jesus Guirro RR. Red and infrared laser therapy inhibits in vitro growth of major bacterial species that commonly colonize skin ulcers. Lasers in Medical Science. 2016; 31: 549–556.
[48]
Touchon J AL, Meunier J, Ceolin L, Roman FJ, Burcelin R, Blivet GJ. The neuroprotective effect of a new photobiomodulation technique on Aβ25-35 peptide-induced toxicity dramatically impact gut microbiota dysbiosis. The Journal of Prevention of Alzheimer’s Disease. 2018; 5: S29–S30.
[49]
Fukuda TY, Tanji MM, Silva SR, Sato MN, Plapler H. Infrared low-level diode laser on inflammatory process modulation in mice: pro- and anti-inflammatory cytokines. Lasers in Medical Science. 2013; 28: 1305–1313.
[50]
Fernandes KPS, Souza NHC, Mesquita-Ferrari RA, Silva DDFTD, Rocha LA, Alves AN, et al. Photobiomodulation with 660-nm and 780-nm laser on activated J774 macrophage-like cells: Effect on M1 inflammatory markers. Journal of Photochemistry and Photobiology. B, Biology. 2015; 153: 344–351.
[51]
Bicknell B, Liebert A, Johnstone D, Kiat H. Photobiomodulation of the microbiome: implications for metabolic and inflammatory diseases. Lasers in Medical Science. 2019; 34: 317–327.
[52]
Benichou Haziot C, Birak KS. Therapeutic Potential of Microbiota Modulation in Alzheimer’s Disease: A Review of Preclinical Studies. Journal of Alzheimer’s Disease Reports. 2023; 7: 415–431.
[53]
Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, et al. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms. 2019; 7: 14.
[54]
Chen Q, Wu J, Dong X, Yin H, Shi X, Su S, et al. Gut flora-targeted photobiomodulation therapy improves senile dementia in an Aß-induced Alzheimer’s disease animal model. Journal of Photochemistry and Photobiology. B, Biology. 2021; 216: 112152.
[55]
Roubaud Baudron C, Varon C, Mégraud F, Salles N. Alzheimer’s disease and Helicobacter pylori infection: a possible link? Geriatrie et Psychologie Neuropsychiatrie Du Vieillissement. 2016; 14: 86–94. (In French)
[56]
Wang M, Cao J, Amakye WK, Gong C, Li Q, Ren J. Mid infrared light treatment attenuates cognitive decline and alters the gut microbiota community in APP/PS1 mouse model. Biochemical and Biophysical Research Communications. 2020; 523: 60–65.
[57]
Ou Z, Deng L, Lu Z, Wu F, Liu W, Huang D, et al. Protective effects of Akkermansia muciniphila on cognitive deficits and amyloid pathology in a mouse model of Alzheimer’s disease. Nutrition & Diabetes. 2020; 10: 12.
[58]
Long Y, Tang L, Zhou Y, Zhao S, Zhu H. Causal relationship between gut microbiota and cancers: a two-sample Mendelian randomisation study. BMC Medicine. 2023; 21: 66.
[59]
Leiva-Gea I, Sánchez-Alcoholado L, Martín-Tejedor B, Castellano-Castillo D, Moreno-Indias I, Urda-Cardona A, et al. Gut Microbiota Differs in Composition and Functionality Between Children With Type 1 Diabetes and MODY2 and Healthy Control Subjects: A Case-Control Study. Diabetes Care. 2018; 41: 2385–2395.
[60]
Shaikh SD, Sun N, Canakis A, Park WY, Weber HC. Irritable Bowel Syndrome and the Gut Microbiome: A Comprehensive Review. Journal of Clinical Medicine. 2023; 12: 2558.
[61]
Cummings JL, Tong G, Ballard C. Treatment Combinations for Alzheimer’s Disease: Current and Future Pharmacotherapy Options. Journal of Alzheimer’s Disease: JAD. 2019; 67: 779–794.
[62]
US Food and Drug Administration, Guidance for Industry: codevelopment of two or more unmarketed investigational drugs for use in combination. 2018. Available at: https://www.fda.gov/downloads/drugs/guidances/ucm236669.pdf (Accessed: 5 June 2020).
[63]
Borbolis F, Mytilinaiou E, Palikaras K. The Crosstalk between Microbiome and Mitochondrial Homeostasis in Neurodegeneration. Cells. 2023; 12: 429.
[64]
Picard M, McEwen BS. Mitochondria impact brain function and cognition. Proceedings of the National Academy of Sciences of the United States of America. 2014; 111: 7–8.
[65]
Castro JP, Wardelmann K, Grune T, Kleinridders A. Mitochondrial Chaperones in the Brain: Safeguarding Brain Health and Metabolism? Frontiers in Endocrinology. 2018; 9: 196.
[66]
Wang W, Zhao F, Ma X, Perry G, Zhu X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: recent advances. Molecular Neurodegeneration. 2020; 15: 30.
[67]
Morton H, Kshirsagar S, Orlov E, Bunquin LE, Sawant N, Boleng L, et al. Defective mitophagy and synaptic degeneration in Alzheimer’s disease: Focus on aging, mitochondria and synapse. Free Radical Biology & Medicine. 2021; 172: 652–667.
[68]
Bano D, Ehninger D, Bagetta G. Decoding metabolic signatures in Alzheimer’s disease: a mitochondrial perspective. Cell Death Discovery. 2023; 9: 432.
[69]
Oliver DMA, Reddy PH. Molecular Basis of Alzheimer’s Disease: Focus on Mitochondria. Journal of Alzheimer’s Disease. 2019; 72: S95–S116.
[70]
John A, Reddy PH. Synaptic basis of Alzheimer’s disease: Focus on synaptic amyloid beta, P-tau and mitochondria. Ageing Research Reviews. 2021; 65: 101208.
[71]
Blivet G, Meunier J, Roman FJ, Touchon J. Neuroprotective effect of a new photobiomodulation technique against Aβ25-35 peptide-induced toxicity in mice: Novel hypothesis for therapeutic approach of Alzheimer’s disease suggested. Alzheimer’s & Dementia. 2018; 4: 54–63.
[72]
Blivet G, Relano-Gines A, Wachtel M, Touchon J. A Randomized, Double-Blind, and Sham-Controlled Trial of an Innovative Brain-Gut Photobiomodulation Therapy: Safety and Patient Compliance. Journal of Alzheimer’s Disease. 2022; 90: 811–822.
[73]
Bicknell B, Laakso EL, Liebert A, Kiat H. Modifying the Microbiome as a Potential Mechanism of Photobiomodulation: A Case Report. Photobiomodulation, Photomedicine, and Laser Surgery. 2022; 40: 88–97.
[74]
Efficacy of RGn600 in Patients With Mild-to-moderate Alzheimer’s Disease (LIGHT4LIFE). 2023. Available at: https://classic.clinicaltrials.gov/ct2/show/NCT05926011 (Accessed: 30 July 2023).
[75]
Li X, Vemireddy V, Cai Q, Xiong H, Kang P, Li X, et al. Reversibly Modulating the Blood-Brain Barrier by Laser Stimulation of Molecular-Targeted Nanoparticles. Nano Letters. 2021; 21: 9805–9815.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top