Letter to the Editor: Comment on “Oral Monosodium Glutamate Administration Causes Early Onset of Alzheimer’s Disease-like Pathophysiology in APP/PS1 Mice"

14 June 2020

This letter refers to the article “Oral Monosodium Glutamate Administration Causes Early Onset of Alzheimer’s Disease-like Pathophysiology in APP/PS1 Mice” by Fuchsberger et al. [1], reporting that oral administration of MSG accelerated the onset of pathophysiological and behavioral symptoms in APP/PS1 mice, a genetically modified animal model of Alzheimer’s disease (AD). The authors speculate that circulating glutamate originating from dietary monosodium glutamate (MSG) entered the brains of APP/PS1 model mice by crossing the blood-brain barrier (BBB), and exerted an excitotoxic effect that led to AD-related hallmarks such as accumulation of amyloid-β (Aβ), and tau phosphorylation.

It is, however, well established that glutamate ingested with food is metabolized, mainly serving as an energy source for enterocytes [2, 3] and normal dietary glutamate ingestion has just a small effect on blood glutamate concentration in humans [3-5]. Further studies have elucidated that the profile of circadian variation in the glutamate concentration in blood is not significantly different on diets with or without added MSG [6], and that the basal glutamate concentration in humans is not affected by long-term MSG consumption [7]. Rodent studies show that ingestion of a diet containing MSG hardly influences blood glutamate levels and suggest that the macronutrients in diets, particularly carbohydrate, is most effective in attenuating any rise in blood glutamate levels induced by oral administration of MSG [8]. These research results imply that experimental animals including mice and rats, which have free access to a normal diet combined with an MSG solution, will not show any increase in the blood glutamate concentration. In fact, some studies conducted under standard feeding conditions demonstrate that rats provided with water containing MSG did not show any rise in the basal blood concentrations of amino acids. Even if MSG is consumed in large amounts under these conditions, blood levels of amino acids including glutamate were maintained at almost constant levels [9, 10]. As described by the authors, blood concentrations of glutamate ranging from 50 µM to 100 µM are markedly higher than those in cerebral extracellular fluid (CEF) [11]. If the authors’ speculation that blood glutamate enters the brains of APP/PS1 mice by crossing BBB is scientifically accurate, APP/PS1 mice must have a high glutamate concentration in the CEF and exhibit pathophysiological changes at 16 weeks of age independent of the experimental condition. It is therefore quite surprising that only the APP/PS1 mice provided with a 1% MSG solution as drinking water appear to show pathophysiological changes with associated changes in cognitive function.

Many types of murine model have been created using genetic modification and used in studies of AD. These model mice have specific characteristics that depend on the modified genes and have been developed to improve applicability to studies of AD. There have been many reviews [12-15] of the pros and cons of the various models, including APP/PS1 transgenic mice, allowing selection of suitable and valid models in studying AD. In these reviews, the disadvantages of the APP/PS1 transgenic mouse, such as relatively unstable Aβ expression and absence of neurofibrillary tangles are noted. These findings imply that onset of AD-like pathophysiology varies depending on each individual. In the current study, the values of biomarkers related to AD were obtained from just three or four samples. I consider this sample size to be insufficient to evaluate the effects of MSG. To overcome these disadvantages, an adequate sample size is required with each verification. Regarding cognitive function assessment, time in open arms should be measured before an intervention to evaluate effect of the intervention.

Recently, it has been suggested that environmental enrichment can reduce accumulation of Aβ deposits and suppress the progression of memory impairment in AD model animals [16, 17]. Effects on AD of interventions associated with the environment including diet and aroma have also been investigated and amelioration of cognitive function has been observed in many clinical trials. A flavor enhancer, MSG is well known to make food more palatable and enhance the enjoyment of eating. Kouzuki et al. reported that an intervention involving ingestion of meals seasoned with MSG for 12 weeks improved some scores related to cognitive function in patients with dementia, including AD [18]. As the authors mention in the report, the average intake of glutamate as a flavoring agent is estimated to be 0.3-0.5 g/day in European countries and 1.2-1.7 g/day in Asian countries, suggesting that people in Asian countries consume a far greater amount of MSG daily [19]. If the authors’ speculation is scientifically accurate, early onset and prevalence of AD and dementia would be prominent in Asian countries. However, this geographical disproportion in onset and prevalence of AD and dementia has not been observed so far [20, 21]. Therefore, in order to suggest a potential role of dietary glutamate in aggravating AD pathophysiology, further studies seem to be needed and the authors should at least discuss the discrepancies between their study results and clinical and epidemiological facts.

Yoshida Shintaro, PhD, DVM
International Glutamate Technical Committee
E-mail: secretariat@e-igtc.org

References:
[1] Fuchsberger T, Yuste R, Martinez-Bellver S, Blanco-Gandia MC, Torres-Cuevas I, Blasco-Serra A, Arango R, Miñarro J, Rodríguez-Arias M, Teruel-Marti V, Lloret A, Viña J (2019) Oral monosodium glutamate administration causes early onset of Alzheimer's disease-like pathophysiology in APP/PS1 mice. J Alzheimers Dis 72, 957-975.
[2] Wu G (1998) Intestinal mucosal amino acid catabolism. J Nutr 128, 1249-1252.
[3] Reeds PJ, Burrin DG, Stoll, B, Jahoor F (2000) Intestinal glutamate metabolism. J Nutr 130, 978S-982S.
[4] Stegink LD, Filer LJ, Baker GL (1985) Effect of starch ingestion on plasma glutamate concentration in human ingesting monosodium L-glutamate in soup. J Nutr 115, 211-218.
[5] Ghezzi P, Bianchi M, Gianera L, Salmona M, Garattini S (1985) Kinetics of monosodium glutamate in human volunteers under different experimental conditions. Food Chem Toxicol 23, 975-978.
[6] Tsai PJ, Huang PC (1999) Circadian variations in plasma and erythrocyte concentrations of glutamate, glutamine, and alanine in men on a diet without and with added monosodium glutamate. Metabolism 48, 1455-1460.
[7] Tanphaichitr V, Leelahagul P, Suwan K (2000) Plasma amino acid pattern and visceral protein status in users and nonusers of monosodium glutamate. J Nutr 130, 1005S-1006S.
[8] Fernstrom MH, Patil VP, Fernstrom JD (2002) The effect of dietary carbohydrate on the rise in plasma glutamate concentrations following oral glutamate ingestion in rats. J Nutr Biochem 13, 734-746.
[9] Kondoh T, Torii K (2008) MSG intake suppresses weight gain, fat deposition, and plasma leptin levels in male Sprague–Dawley rats. Physiol Behav 95, 135-144.
[10] Nishigaki R, Yokoyama Y, Shimizu Y, Marumoto R, Misumi S, Ueda Y, Ishida A, Shibuya Y, Hida H (2018) Monosodium glutamate ingestion during the development period reduces aggression mediated by the vagus nerve in a rat model of attention deficit-hyperactivity disorder. Brain Res 1690, 40-50.
[11] Hawkins RA (2009) The blood-brain barrier and glutamate. Am J Clin Nutr 90, 867S-874S.
[12] Li H, Wei Y, Wang Z, Wang Q (2015) Application of APP/PS1 transgenic mouse model for Alzheimer’s disease. J Alzheimers Dis Parkinsonism 5, 201.
[13] Wu X, Li J, Zhou W, Tam K (2015) Animal models for Alzheimer’s disease: a focused review of transgenic rodent models and behavioral assessment methods. ADMET DMPK 3, 242-253.
[14] Drummond E, Wisniewski T (2017) Alzheimer’s disease: experimental models and reality. Acta Neuropathol 133, 155-175.
[15] Jankowsky JL, Zheng H (2017) Practical consideration for choosing mouse model of Alzheimer’s disease. Mol Neurodegener 12, 89.
[16] Lazarov O, Robinson J, Tang YP, Hairston IS, Korade-Mirnics Z, Lee VM, Hersh LB, Sapolsky RM, Mirnics K, Sisodia SS (2005) Environmental enrichment reduces Abeta levels and amyloid deposition in transgenic mice. Cell 120, 701-713.
[17] Jankowsky JL, Melnikova T, Fadale DJ, Xu GM, Slunt HH, Gonzales V, Younkin LH, Younkin SG, Borchelt DR, Savonenko AV (2005) Environmental enrichment mitigates cognitive deficits in a mouse model of Alzheimer's disease. J Neurosci 25, 5217-5224.
[18] Kouzuki M, Taniguchi M, Suzuki T, Nagano M, Nakamura S, Katsumata Y, Matsumoto H, Urakami K (2019) Effect of monosodium L-glutamate (umami substance) on cognitive function in people with dementia. Eur J Clin Nutr 73, 266-275.
[19] Beyreuther K, Biesalski HK, Fernstrom JD, Grimm P, Hammes W P, Heinemann U, Kempski O, Stehle P, Steinhart H, Walker R (2007) Consensus metting: Monosodium glutamate – an update. Eur J Clin Nutr 61, 304-313.
[20] Fratiglioni L, Ronchi DD, Agüero-Torres H (1999) Worldwide prevalence and incidence of dementia. Drugs Aging 15, 365-375.
[21] Prince M, Wimo A, Guerchet M, Ali GC, Wu YT, Prina M (2015) World Alzheimer Report 2015: The Global Impact of Dementia: An analysis of prevalence, incidence, cost and trends. Alzheimer’s Disease International, London.

Comments

We have carefully read the comments of Dr. Shinora on our article. We thank the editor for publishing our response alongside.

Dr. Yoshida cites various studies that found no significant effect on plasma glutamate levels after MSG ingestion. It should be stated in this context that there is, however, much controversy in the literature over this topic. Other studies did report an increase in plasma glutamate levels after ingestion of MSG in humans [1-3] and rodents [4]. It is therefore possible that MSG supplementation does increase plasma glutamate levels, at least under certain circumstances. However, in our study we did not address this question, but focused on long-term effects of sustained MSG ingestion several weeks after MSG was administered. Indeed, in our paper we stated that glutamate may have entered the brain through the blood-brain barrier (BBB) as a possible explanation for our findings, but this is subject to future research. Since breakdown of the BBB has been described in the aging brain [5], in early Alzheimer’s disease [6], and even in people with the ApoE4 allele [7], we still regard this as the most likely hypothesis. In any case, whether future findings will or will not support this hypothesis, does not invalidate any of the major conclusion of the present paper. Analyzing BBB permeability to glutamate was not the main aim of this paper.

Dr. Yoshida also deems it as ‘surprising’ that only APP/PS1 with 1% MSG and not in other experimental conditions—that is, 0.5% MSG—show impairments after treatment. We cannot follow the logic of the argumentation why it would be surprising that only the higher concentration should in fact induce impairments in APP/PS1 animals, while the lower MSG concentration would not.

Furthermore, Dr. Yoshida raises concerns about the sample sizes we used in this study, given that pathophysiological development can vary between individuals in the APP/PS1 mouse model. The statistical tests that were applied demonstrated that the observed effects were significant, meaning that they were above chance level. Let us point out that is in fact the very essence of statistical parametric testing such as that resting upon Student’s t- or Snedecor’s F-distributions: by merely assuming normality and by taking into account within-sample variability, these tests allow for valid statistical inferences however little the sample size may be. A fortiori, the consistency and prominence of the mean difference exhibited between the groups (e.g., there was 4 times more Aβ present in the MSG-supplemented group, Fig. 2), turned out for our sample sizes to be sufficient. Besides, for some experiments larger sample sizes were indeed used (over 10 animals in each group), and we observed consistent effects in the MSG-1%-group, which correlated between different sets of experiments carried out by different researchers. Therefore, from a scientific standpoint, having applied well established statistical testing in a careful and responsible way, we are very much confident that our conclusions are reliable and consistent under the significance levels reported.

Genetic variability and several environmental factors can influence the pathophysiology of AD, making it therefore difficult to relate our findings directly to epidemiological tendencies. We agree with Dr. Yoshida that more studies will be required to address whether MSG could have an effect in a certain subpopulation in humans (like carriers of the ApoE4 mutation, for example) and—as suggested in our discussion—this will be an interesting topic for further research.

Tanja Fuchsberger, Jose Viña and Ana Lloret

References
[1] Marina M, Graham TE (2002) Glutamate ingestion and its effects at rest and during exercise in humans. J Appl Physiol 93, 1251–1259.
[2] Graham TE, Sgro V, Friars D, Gibala MJ (2000) Glutamate ingestion: the plasma and muscle free amino acid pools of resting humans. Am J Physiol Endocrinol Metab 278, E83–E89.
[3] Stegink LD, Filer LJ Jr, Baker GL, Bell EF (1986) Plasma glutamate concentrations in 1-year-old infants and adults ingesting monosodium L-glutamate in consommi. Pediatr Res 20, 53-58.
[4] McLaughlan JM, Neel FJ, Botting HG, Knipfel JE (1970) Blood and brain levels of glutamic acid in young rats given monosodium glutamate. Nutr Rep Int 1, 131-138.
[5] Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, Toga AW, Jacobs RE, Liu CY, Amezcua L, Harrington MG, Chui HC, Law M, Zlokovic BV (2015) Blood-brain barrier breakdown in the aging human hippocampus. Neuron 85, 296-302.
[6] van de Haar HJ, Burgmans S, Jansen JF, van Osch MJ, van Buchem MA, Muller M, Hofman PA, Verhey FR, Backes WH (2016) Blood–brain barrier leakage in patients with early Alzheimer disease. Radiology 281, 527–535.
[7] Montagne A, Nation DA, Sagare AP, Barisano G, Sweeney MD, Chakhoyan A, Pachicano M, Joe E, Nelson AR, D'Orazio LM, Buennagel DP, Harrington MG, Benzinger TLS, Fagan AM, Ringman JM, Schneider LS, Morris JC, Reiman EM, Caselli RJ, Chui HC, Tcw J, Chen Y, Pa J, Conti PS, Law M, Toga AW, Zlokovic BV (2020) APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature 581, 71-76.

Dear Sir;

Our article which was published by Plos ONE in 2010 presented that homocysteic acid (HCA) was a pathogen of 3xTg-AD model mice (1). And also we published that blood HCA was the pathogen of human AD (2). This HCA is the strong agonist of glutamate, which suggests MSG is the same glutamate.

References

1. Hasegawa T, Mikoda N, Kitazawa M, LaFerla FM (2010) Treatment of Alzheimer’s Disease with Anti-Homocysteic Acid Antibody in 3xTg-AD Male Mice. PLoS ONE 5(1): e8593. doi:10.1371/journal.pone.0008593

2. Tohru Hasegawa, Masayoshi Ichiba, Shin-ei Matsumoto, Koji Kasanuki, Taku Hatano, Hiroshige Fujishiro, Eizo Iseki, Nobutaka Hattori, Tatsuo Yamada, Takeshi Tabira (2012) Urinary Homocysteic Acid Levels Correlate with Mini-Mental State Examination Scores in Alzheimer’s Disease Patients. Journal of Alzheimer’s Disease 31, 59–64.