Inflammatory Blood Biomarker Kynurenine Is Linked With Elevated Neuroinflammation and Neurodegeneration in Older Adults: Evidence From Two 1H-MRS Post-Processing Analysis Methods

Wouter A J Vints, Simona Kušleikiene, Samrat Sheoran, Milda Šarkinaite, Kristina Valatkevičiene, Rymante Gleizniene, Mindaugas Kvedaras, Kazimieras Pukenas, Uwe Himmelreich, Vida J Cesnaitiene, Oron Levin, Jeanine Verbunt, Nerijus Masiulis, Wouter A J Vints, Simona Kušleikiene, Samrat Sheoran, Milda Šarkinaite, Kristina Valatkevičiene, Rymante Gleizniene, Mindaugas Kvedaras, Kazimieras Pukenas, Uwe Himmelreich, Vida J Cesnaitiene, Oron Levin, Jeanine Verbunt, Nerijus Masiulis

Abstract

Rationale and objectives: Pro-inflammatory processes have been argued to play a role in conditions associated with cognitive decline and neurodegeneration, like aging and obesity. Only a limited number of studies have tried to measure both peripheral and central biomarkers of inflammation and examined their interrelationship. The primary aim of this study was to examine the hypothesis that chronic peripheral inflammation would be associated with neurometabolic changes that indicate neuroinflammation (the combined elevation of myoinositol and choline), brain gray matter volume decrease, and lower cognitive functioning in older adults.

Materials and methods: Seventy-four older adults underwent bio-impedance body composition analysis, cognitive testing with the Montreal Cognitive Assessment (MoCA), blood serum analysis of inflammatory markers interleukin-6 (IL-6) and kynurenine, magnetic resonance imaging (MRI), and proton magnetic resonance spectroscopy (1H-MRS) of the brain. Neurometabolic findings from both Tarquin and LCModel 1H-MRS post-processing software packages were compared. The regions of interest for MRI and 1H-MRS measurements were dorsal posterior cingulate cortex (DPCC), left hippocampal cortex (HPC), left medial temporal cortex (MTC), left primary sensorimotor cortex (SM1), and right dorsolateral prefrontal cortex (DLPFC).

Results: Elevated serum kynurenine levels were associated with signs of neuroinflammation, specifically in the DPCC, left SM1 and right DLPFC, and signs of neurodegeneration, specifically in the left HPC, left MTC and left SM1, after adjusting for age, sex and fat percentage (fat%). Elevated serum IL-6 levels were associated with increased Glx levels in left HPC, left MTC, and right DLPFC, after processing the 1H-MRS data with Tarquin. Overall, the agreement between Tarquin and LCModel results was moderate-to-strong for tNAA, tCho, mIns, and tCr, but weak to very weak for Glx. Peripheral inflammatory markers (IL-6 and kynurenine) were not associated with older age, higher fat%, decreased brain gray matter volume loss or decreased cognitive functioning within a cohort of older adults.

Conclusion: Our results suggest that serum kynurenine may be used as a peripheral inflammatory marker that is associated with neuroinflammation and neurodegeneration, although not linked to cognition. Future studies should consider longitudinal analysis to assess the causal inferences between chronic peripheral and neuroinflammation, brain structural and neurometabolic changes, and cognitive decline in aging.

Keywords: aging; brain imaging; cerebral metabolite ratios; cognition; gray matter volume; inflammation; neurochemicals; obesity.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Vints, Kušleikiene, Sheoran, Šarkinaite, Valatkevičiene, Gleizniene, Kvedaras, Pukenas, Himmelreich, Cesnaitiene, Levin, Verbunt and Masiulis.

Figures

Figure 1
Figure 1
(A) Example voxel positions and spectra from a representative subject. (B) Raw (black curve) and fitted (red curve) spectra from LCModel (left hand side panel) and Tarquin (right hand side panel) are illustrated for the left HPC and right DLPFC. Cho, total choline; Cr, creatine + phosphocreatine; DLPFC, dorsolateral prefrontal cortex; DPCC, dorsal posterior cingulate cortex; Glx, glutamate–glutamine complex; HPC, hippocampal cortex; mIns, myoinositol; MTC, medial temporal cortex; NAA, N-acetyl aspartate; SM1, primary sensorimotor cortex.

References

    1. Dall TM, Gallo PD, Chakrabarti R, West T, Semilla AP, Storm M V. An aging population and growing disease burden will require a large and specialized health care workforce by 2025. Health Aff. (2013) 32:2013–20. 10.1377/hlthaff.2013.0714
    1. Mattiuzzi C, Lippi G. Worldwide disease epidemiology in the older persons. Eur Geriatr Med. (2020) 11:147–53. 10.1007/S41999-019-00265-2/TABLES/4
    1. Mayeux R, Stern Y. Epidemiology of Alzheimer disease. Cold Spring Harb Perspect Med. (2012) 2:6239. 10.1101/CSHPERSPECT.A006239
    1. Haththotuwa RN, Wijeyaratne CN, Senarath U. Worldwide epidemic of obesity. In: Mahmood TA, Arulkumaran S, Chervenak FA, editors. Obesity and Obstetrics, 2nd ed. Netherlands: Elsevier; (2020) 3–8. 10.1016/B978-0-12-817921-5.00001-1
    1. Cournot M, Marquié JC, Ansiau D, Martinaud C, Fonds H, Ferrières J, et al. . Relation between body mass index and cognitive function in healthy middle-aged men and women. Neurology. (2006) 67:1208–14. 10.1212/01.WNL.0000238082.13860.50
    1. Mrak RE. Alzheimer-type neuropathological changes in morbidly obese elderly individuals. Clin Neuropathol. (2009) 28:40–5. 10.5414/NPP28040
    1. Rakesh G, Szabo ST, Alexopoulos GS, Zannas AS. Strategies for dementia prevention: latest evidence and implications. Ther Adv Chronic Dis. (2017) 8:121–36. 10.1177/2040622317712442
    1. Sartori AC, Vance DE, Slater LZ, Crowe M. The impact of inflammation on cognitive function in older adults: implications for healthcare practice and research. J Neurosci Nurs. (2012) 44:206–17. 10.1097/JNN.0b013e3182527690
    1. Tang Y, Purkayastha S, Cai D. Hypothalamic microinflammation: a common basis of metabolic syndrome and aging. Trends Neurosci. (2015) 38:36–44. 10.1016/J.TINS.2014.10.002
    1. Beilharz JE, Maniam J, Morris MJ. Short-term exposure to a diet high in fat and sugar, or liquid sugar, selectively impairs hippocampal-dependent memory, with differential impacts on inflammation. Behav Brain Res. (2016) 306:1–7. 10.1016/J.BBR.2016.03.018
    1. Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging. An evolutionary perspective on immunosenescence. in Annals of the New York Academy of Sciences (New York Academy of Sciences; ):244–254. 10.1111/j.1749-6632.2000.tb06651.x
    1. Borodkina A V, Deryabin PI, Giukova AA, Nikolsky NN. “Social life” of senescent cells: what is sasp and why study it? Acta Naturae. (2018) 10:4–14. Available online at: (accessed October 25, 2020).
    1. Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, et al. . A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. (1995) 92:9363–7. 10.1073/pnas.92.20.9363
    1. Campisi J, D'Adda Di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. (2007) 8:729–40. 10.1038/nrm2233
    1. Yudkin JS. Inflammation, obesity, and the metabolic syndrome. in Hormone and Metabolic Research. New York:© Georg Thieme Verlag KG Stuttgart ·, 707–9. 10.1055/s-2007-985898
    1. Woods JA, Wilund KR, Martin SA, Kistler BM. Exercise, inflammation and aging. Aging Dis. (2012) 3:130–140. Available online at: /pmc/articles/PMC3320801/?report=abstract (Accessed October 20, 2020).
    1. Scheiblich H, Trombly M, Ramirez A, Heneka MT. neuroimmune connections in aging and neurodegenerative diseases. Trends Immunol. (2020) 41:300–12. 10.1016/j.it.2020.02.002
    1. Guillemot-Legris O, Muccioli GG. Obesity-induced neuroinflammation: beyond the hypothalamus. Trends Neurosci. (2017) 40:237–53. 10.1016/J.TINS.2017.02.005
    1. Terry RD, DeTeresa R, Hansen LA. Neocortical cell counts in normal human adult aging. Ann Neurol. (1987) 21:530–9. 10.1002/ana.410210603
    1. Perry VH, Cunningham C, Holmes C. Systemic infections and inflammation affect chronic neurodegeneration. Nat Rev Immunol. (2007) 7:161–7. 10.1038/nri2015
    1. Cohen J, Torres C. Astrocyte senescence: evidence and significance. Aging Cell. (2019) 18:12937. 10.1111/acel.12937
    1. Lu J, Wu D. mei, Zheng Y lin, Hu B, Cheng W, Zhang Z feng, Shan Q. Ursolic acid improves high fat diet-induced cognitive impairments by blocking endoplasmic reticulum stress and IκB kinase β/nuclear factor-κB-mediated inflammatory pathways in mice. Brain Behav Immun. (2011) 25:1658–67. 10.1016/J.BBI.2011.06.009
    1. Tapia-González S, García-Segura LM, Tena-Sempere M, Frago LM, Castellano JM, Fuente-Martín E, et al. . Activation of microglia in specific hypothalamic nuclei and the cerebellum of adult rats exposed to neonatal overnutrition. J Neuroendocrinol. (2011) 23:365–70. 10.1111/J.1365-2826.2011.02113.X
    1. Guillemot-Legris O, Masquelier J, Everard A, Cani PD, Alhouayek M, Muccioli GG. High-fat diet feeding differentially affects the development of inflammation in the central nervous system. J Neuroinflammation. (2016) 13:1–11. 10.1186/S12974-016-0666-8/FIGURES/5
    1. van Dam AM, Bauer J, Tilders FJH, Berkenbosch F. Endotoxin-induced appearance of immunoreactive interleukin-1 beta in ramified microglia in rat brain: a light and electron microscopic study. Neuroscience. (1995) 65:815–26. 10.1016/0306-4522(94)00549-K
    1. Layé S, Goujon E, Combe C, VanHoy R, Kelley KW, Parnet P, et al. . Effects of lipopolysaccharide and glucocorticoids on expression of interleukin-1β converting enzyme in the pituitary and brain of mice. J Neuroimmunol. (1996) 68:61–6. 10.1016/0165-5728(96)00066-5
    1. Nguyen KT, Deak T, Owens SM, Kohno T, Fleshner M, Watkins LR, et al. . Exposure to acute stress induces brain interleukin-1β protein in the rat. J Neurosci. (1998) 18:2246. 10.1523/JNEUROSCI.18-06-02239.1998
    1. Turrin NP, Gayle D, Ilyin SE, Flynn MC, Langhans W, Schwartz GJ, et al. . Pro-inflammatory and anti-inflammatory cytokine mRNA induction in the periphery and brain following intraperitoneal administration of bacterial lipopolysaccharide. Brain Res Bull. (2001) 54:443–53. 10.1016/S0361-9230(01)00445-2
    1. Bourgognon JM, Cavanagh J. The role of cytokines in modulating learning and memory and brain plasticity. Brain Neurosci Adv. (2020) 4:239821282097980. 10.1177/2398212820979802
    1. Dinarello CA, Cannon JG, Wolff SM. New Concepts on the Pathogenesis of Fever. Rev Infect Dis. (1988) 10:168–89. 10.1093/CLINIDS/10.1.168
    1. Ericsson A, Kovács KJ, Sawchenko PE. A functional anatomical analysis of central pathways subserving the effects of interleukin-1 on stress-related neuroendocrine neurons. J Neurosci. (1994) 14:897–913. 10.1523/JNEUROSCI.14-02-00897.1994
    1. Goehler LE, Relton JK, Dripps D, Kiechle R, Tartaglia N, Maier SF, et al. . Vagal paraganglia bind biotinylated interleukin-1 receptor antagonist: a possible mechanism for immune-to-brain communication. Brain Res Bull. (1997) 43:357–64. 10.1016/S0361-9230(97)00020-8
    1. Gaykema RPA, Goehler LE, Tilders FJH, Bol JGJM, McGorry M, Fleshner M, et al. . Bacterial endotoxin induces fos immunoreactivity in primary afferent neurons of the vagus nerve. Neuroimmunomodulation. (1998) 5:234–40. 10.1159/000026343
    1. Banks WA, Kastin AJ, Brennan JM, Vallance KL. Adsorptive endocytosis of HIV-1gp120 by blood-brain barrier is enhanced by lipopolysaccharide. Exp Neurol. (1999) 156:165–71. 10.1006/EXNR.1998.7011
    1. Maier SF. Bi-directional immune-brain communication: implications for understanding stress, pain, and cognition. Brain Behav Immun. (2003) 17:69–85. 10.1016/S0889-1591(03)00032-1
    1. Bachstetter AD, Morganti JM, Jernberg J, Schlunk A, Mitchell SH, Brewster KW, et al. . Fractalkine and CX 3 CR1 regulate hippocampal neurogenesis in adult and aged rats. Neurobiol Aging. (2011) 32:2030–44. 10.1016/J.NEUROBIOLAGING.2009.11.022
    1. Barrientos RM, Kitt MM, Watkins LR, Maier SF. Neuroinflammation in the normal aging hippocampus. Neuroscience. (2015) 309:84–99. 10.1016/j.neuroscience.2015.03.007
    1. Maier SF, Watkins LR. Cytokines for psychologists: implications of bidirectional immune-to-brain communication for understanding behavior, mood, and cognition. Psychol Rev. (1998) 105:83–107. 10.1037/0033-295X.105.1.83
    1. Campbell SJ, Jiang Y, Davis AEM, Farrands R, Holbrook J, Leppert D, et al. . Immunomodulatory effects of etanercept in a model of brain injury act through attenuation of the acute-phase response. J Neurochem. (2007) 103:2245–55. 10.1111/J.1471-4159.2007.04928.X
    1. Jiang Y, Deacon R, Anthony DC, Campbell SJ. Inhibition of peripheral TNF can block the malaise associated with CNS inflammatory diseases. Neurobiol Dis. (2008) 32:125–32. 10.1016/J.NBD.2008.06.017
    1. Campbell SJ, Hughes PM, Iredale JP, Wilcockson DC, Waters S, Docagne F, et al. . CINC-1 is an acute-phase protein induced by focal brain injury causing leukocyte mobilization and liver injury. FASEB J. (2003) 17:1168–70. 10.1096/FJ.02-0757FJE
    1. Campbell SJ, Perry VH, Pitossi FJ, Butchart AG, Chertoff M, Waters S, et al. . Central nervous system injury triggers hepatic CC and CXC chemokine expression that is associated with leukocyte mobilization and recruitment to both the central nervous system and the liver. Am J Pathol. (2005) 166:1487–97. 10.1016/S0002-9440(10)62365-6
    1. Campbell SJ, Deacon RMJ, Jiang Y, Ferrari C, Pitossi FJ, Anthony DC. Overexpression of IL-1beta by adenoviral-mediated gene transfer in the rat brain causes a prolonged hepatic chemokine response, axonal injury and the suppression of spontaneous behaviour. Neurobiol Dis. (2007) 27:151–63. 10.1016/J.NBD.2007.04.013
    1. Lind A, Boraxbekk C-J, Petersen ET, Paulson OB, Siebner HR, Marsman A. Regional myo-inositol, creatine, and choline levels are higher at older age and scale negatively with visuospatial working memory: a cross-sectional proton mr spectroscopy study at 7 tesla on normal cognitive ageing. J Neurosci. (2020) 40:8149–59. 10.1523/JNEUROSCI.2883-19.2020
    1. Lind A, Boraxbekk CJ, Petersen ET, Paulson OB, Andersen O, Siebner HR, et al. . Do glia provide the link between low-grade systemic inflammation and normal cognitive ageing? A 1H magnetic resonance spectroscopy study at 7 tesla. J Neurochem. (2021) 159:185–96. 10.1111/JNC.15456
    1. Ershler WB. Interleukin-6: a cytokine for gerontolgists. J Am Geriatr Soc. (1993) 41:176–81. 10.1111/J.1532-5415.1993.TB02054.X
    1. Maggio M, Guralnik JM, Longo DL, Ferrucci L. Interleukin-6 in aging and chronic disease: a magnificent pathway. Journals Gerontol - Ser A Biol Sci Med Sci. (2006) 61:575–84. 10.1093/gerona/61.6.575
    1. Bradburn S, Sarginson J, Murgatroyd CA. Association of peripheral interleukin-6 with global cognitive decline in non-demented adults: a meta-analysis of prospective studies. Front Aging Neurosci. (2018) 9:438. 10.3389/fnagi.2017.00438
    1. Smith LL, Miles MP. “Exercise-Induced Muscle Injury and Inflammation,” in Exercise and Sport Science, eds. Garret WE, Kirkendall DT (Philadelphia: Lippincott Williams & Wilkins; ), 401–12. Available online at: (accessed October 20, 2020)
    1. Allison DJ, Josse AR, Gabriel DA, Klentrou P, Ditor DS. Targeting inflammation to influence cognitive function following spinal cord injury: a randomized clinical trial. Spinal Cord. (2017) 55:26–32. 10.1038/sc.2016.96
    1. Chiarugi A, Calvani M, Meli E, Traggiai E, Moroni F. Synthesis and release of neurotoxic kynurenine metabolites by human monocyte-derived macrophages. J Neuroimmunol. (2001) 120:190–8. 10.1016/S0165-5728(01)00418-0
    1. Lustgarten MS, Fielding RA. Metabolites associated with circulating interleukin-6 in older adults. J Gerontol Ser A. (2017) 72:1277–83. 10.1093/GERONA/GLW039
    1. Kindler J, Lim CK, Weickert CS, Boerrigter D, Galletly C, et al. . Dysregulation of kynurenine metabolism is related to proinflammatory cytokines, attention, and prefrontal cortex volume in schizophrenia. Mol Psychiatry. (2019) 25:2860–2872. 10.1038/s41380-019-0401-9
    1. Pedraz-Petrozzi B, Elyamany O, Rummel C, Mulert C. Effects of inflammation on the kynurenine pathway in schizophrenia - a systematic review. J Neuroinflammation. (2020) 17:1–17. 10.1186/S12974-020-1721-Z/TABLES/4
    1. Wichers MC, Maes M. The role of indoleamine 2,3-dioxygenase (IDO) in the pathophysiology of interferon-α-induced depression. J Psychiatry Neurosci. (2004) 29:11. Available online at: (Accessed December 16, 2021).
    1. Vécsei L, Szalárdy L, Fülöp F, Toldi J. Kynurenines in the CNS: recent advances and new questions. Nat Rev Drug Discov. (2013) 12:64–82. 10.1038/nrd3793
    1. Mangge H, Summers KL, Meinitzer A, Zelzer S, Almer G, Prassl R, et al. . Obesity-related dysregulation of the Tryptophan–Kynurenine metabolism: role of age and parameters of the metabolic syndrome. Obesity. (2014) 22:195–201. 10.1002/OBY.20491
    1. Solvang SEH, Nordrehaug JE, Tell GS, Nygård O, McCann A, Ueland PM, et al. . The kynurenine pathway and cognitive performance in community-dwelling older adults. The Hordaland health study. Brain Behav Immun. (2019) 75:155–62. 10.1016/j.bbi.2018.10.003
    1. Skorobogatov K, De Picker L, Verkerk R, Coppens V, Leboyer M, et al. . Brain vs. blood: a systematic review on the concordance between peripheral and central kynurenine pathway measures in psychiatric disorders. Front Immunol. (2021) 12:980. 10.3389/FIMMU.2021.716980
    1. Zwilling D, Huang SY, Sathyasaikumar K V, Notarangelo FM, Guidetti P, Wu HQ, et al. . Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell. (2011) 145:863–74. 10.1016/J.CELL.2011.05.020
    1. Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, Collin I, et al. . The montreal cognitive assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc. (2005) 53:695–9. 10.1111/j.1532-5415.2005.53221.x
    1. Bruijnen CJWH, Dijkstra BAG, Walvoort SJW, Budy MJJ, Beurmanjer H, De Jong CAJ, et al. . Psychometric properties of the Montreal Cognitive Assessment (MoCA) in healthy participants aged 18–70. Int J Psychiatry Clin Pract. (2020) 24:293–300. 10.1080/13651501.2020.1746348
    1. Malek-Ahmadi M, Powell JJ, Belden CM, Oconnor K, Evans L, Coon DW, et al. . Age- and education-adjusted normative data for the montreal cognitive assessment (MoCA) in older adults age 70–99. Neuropsychol Dev Cogn B Aging Neuropsychol Cogn. (2015) 22:755–61. 10.1080/13825585.2015.1041449
    1. He X, Li Z, Tang X, Zhang L, Wang L, He Y, Jin T, Yuan D. Age- and sex-related differences in body composition in healthy subjects aged 18 to 82 years. Medicine. (2018) 97:11152. 10.1097/MD.0000000000011152
    1. Zöllner HJ, PovaŽan M, Hui SCN, Tapper S, Edden RAE, Oeltzschner G. Comparison of different linear-combination modeling algorithms for short-TE proton spectra. NMR Biomed. (2021) 34:4482. 10.1002/NBM.4482
    1. Kanowski M, Kaufmann J, Braun J, Bernarding J, Tempelmann C. Quantitation of simulated short echo time 1H human brain spectra by LCModel and AMARES. Magn Reson Med. (2004) 51:904–12. 10.1002/MRM.20063
    1. Near J, Harris AD, Juchem C, Kreis R, Marjańska M, Öz G, Slotboom J, Wilson M, Gasparovic C. Preprocessing, analysis and quantification in single-voxel magnetic resonance spectroscopy: experts' consensus recommendations. NMR Biomed. (2021) 34:457. 10.1002/NBM.4257
    1. Glanville NT, Byers DM, Cook HW, Spence MW, Palmer FBSC. Differences in the metabolism of inositol and phosphoinositides by cultured cells of neuronal and glial origin. Biochim Biophys Acta (BBA)/Lipids Lipid Metab. (1989) 1004:169–79. 10.1016/0005-2760(89)90265-8
    1. Urenjak J, Williams SR, Gadian DG, Noble M. Proton nuclear magnetic resonance spectroscopy unambiguously identifies different neural cell types. J Neurosci. (1993) 13:981–9. 10.1523/jneurosci.13-03-00981.1993
    1. Bitsch A, Bruhn H, Vougioukas V, Stringaris A, Lassmann H, Frahm J, Brück W. Inflammatory CNS demyelination: histopathologic correlation with in vivo quantitative proton MR spectroscopy. Am J Neuroradiol. (1999) 20:1619–27.
    1. Cichocka M, Bereś A. From fetus to older age: a review of brain metabolic changes across the lifespan. Ageing Res Rev. (2018) 46:60–73. 10.1016/j.arr.2018.05.005
    1. Waragai M, Moriya M, Nojo T. Decreased N-acetyl aspartate/myo-inositol ratio in the posterior cingulate cortex shown by magnetic resonance spectroscopy may be one of the risk markers of preclinical alzheimer's disease: a 7-year follow-up study. J Alzheimer's Dis. (2017) 60:1411–27. 10.3233/JAD-170450
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. (1995) 57:289–300. 10.1111/J.2517-6161.1995.TB02031.X
    1. Rosen Y, Lenkinski RE. Recent advances in magnetic resonance neurospectroscopy. Neurotherapeutics. (2007) 4:330–45. 10.1016/j.nurt.2007.04.009
    1. Graff-Radford J, Kantarci K. Magnetic resonance spectroscopy in Alzheimer's disease. Neuropsychiatr Dis Treat. (2013) 9:687–96. 10.2147/NDT.S35440
    1. Ge X, Xu XY, Feng CH, Wang Y, Li YL, Feng B. Relationships among serum C-reactive protein, receptor for advanced glycation products, metabolic dysfunction, and cognitive impairments. BMC Neurol. (2013) 13:110. 10.1186/1471-2377-13-110
    1. Danielsen ER, Ross B. eds. Magnetic Resonance Spectroscopy Diagnosis of Neurological Diseases. 1st ed. New York: Marcel Dekker Inc; (1999). Available online at: (accessed October 24, 2020).
    1. Ho TC, Teresi GI, Segarra JR, Ojha A, Walker JC, Gu M, et al. . Higher levels of pro-inflammatory cytokines are associated with higher levels of glutamate in the anterior cingulate cortex in depressed adolescents. Front Psychiatry. (2021) 12:346. 10.3389/FPSYT.2021.642976/BIBTEX
    1. Ali NS, Hashem AHH, Hassan AM, Saleh AA, El-Baz HN. Serum interleukin-6 is related to lower cognitive functioning in elderly patients with major depression. Aging Ment Health. (2018) 22:655–61. 10.1080/13607863.2017.1293005
    1. Fayed N, Andrés E, Viguera L, Modrego PJ, Garcia-Campayo J. Higher glutamate+glutamine and reduction of N-acetylaspartate in posterior cingulate according to age range in patients with cognitive impairment and/or pain. Acad Radiol. (2014) 21:1211–7. 10.1016/J.ACRA.2014.04.009
    1. Lewis CP, Port JD, Blacker CJ, Sonmez AI, Seewoo BJ, Leffler JM, et al. . Altered anterior cingulate glutamatergic metabolism in depressed adolescents with current suicidal ideation. Transl Psychiatry. (2020) 10:119. 10.1038/S41398-020-0792-Z
    1. Káradóttir R, Attwell D. Neurotransmitter receptors in the life and death of oligodendrocytes. Neuroscience. (2007) 145:1426–38. 10.1016/J.NEUROSCIENCE.2006.08.070
    1. Matute C. Glutamate and ATP signalling in white matter pathology. J Anat. (2011) 219:53–64. 10.1111/J.1469-7580.2010.01339.X
    1. Moriguchi S, Takamiya A, Noda Y, Horita N, Wada M, et al. . Glutamatergic neurometabolite levels in major depressive disorder: a systematic review and meta-analysis of proton magnetic resonance spectroscopy studies. Mol Psychiatry. (2018) 24:952–64. 10.1038/s41380-018-0252-9
    1. Oeltzschner G, Wijtenburg SA, Mikkelsen M, Edden RAE, Barker PB, Joo JH, et al. . Neurometabolites and associations with cognitive deficits in mild cognitive impairment: a magnetic resonance spectroscopy study at 7 Tesla. Neurobiol Aging. (2019) 73:211–8. 10.1016/J.NEUROBIOLAGING.2018.09.027
    1. Harris JL, Yeh H-W, Swerdlow RH, Choi I-Y, Lee P, Brooks WM. High-field proton magnetic resonance spectroscopy reveals metabolic effects of normal brain aging. Neurobiol Aging. (2014) 35:1686–94. 10.1016/j.neurobiolaging.2014.01.018
    1. Cleeland C, Pipingas A, Scholey A, White D. Neurochemical changes in the aging brain: a systematic review. Neurosci Biobehav Rev. (2019) 98:306–19. 10.1016/j.neubiorev.2019.01.003
    1. Wilson M, Reynolds G, Kauppinen RA, Arvanitis TN, Peet AC. A constrained least-squares approach to the automated quantitation of in vivo 1H magnetic resonance spectroscopy data. Magn Reson Med. (2011) 65:1–12. 10.1002/MRM.22579
    1. Mosconi E, Sima DM, Osorio Garcia MI, Fontanella M, Fiorini S, Van Huffel S, et al. . Different quantification algorithms may lead to different results : a comparison using proton MRS lipid signals. NMR Biomed. (2014) 27:431–43. 10.1002/NBM.3079
    1. Scott J, Underwood J, Garvey LJ, Mora-Peris B, Winston A. A comparison of two post-processing analysis methods to quantify cerebral metabolites measured via proton magnetic resonance spectroscopy in HIV disease. Br J Radiol. (2016) 89:20150979. 10.1259/BJR.20150979
    1. Kossowski B, Orzeł J, Bogorodzki P, Wilson M, Setkowicz Z. P. Gazdzinski S Follow-up analyses on the effects of long-term use of high fat diet on hippocampal metabolite concentrations in Wistar rats: Comparing Tarquin quantification of 70T rat metabolites to LCModel. Biol Eng Med. (2017) 2:1–7. 10.15761/BEM.1000129
    1. Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat Rev Endocrinol. (2012) 8:457–65. 10.1038/nrendo.2012.49
    1. Carey AL, Bruce CR, Sacchetti M, Anderson MJ, Olsen DB, Saltin B, et al. . Interleukin-6 and tumor necrosis factor-α are not increased in patients with Type 2 diabetes: evidence that plasma interleukin-6 is related to fat mass and not insulin responsiveness. Diabetologia. (2004) 47:1029–37. 10.1007/s00125-004-1403-x
    1. Kline R, Wong E, Haile M, Didehvar S. Peri-operative inflammatory cytokines in plasma of the elderly correlate in prospective study with postoperative changes in cognitive test scores. Int J Anesthesiol Res. (2016) 4:313–21. 10.19070/2332-2780-1600065
    1. Puzianowska-Kuznicka M, Owczarz M, Wieczorowska-Tobis K, Nadrowski P, Chudek J, Slusarczyk P, et al. . Interleukin-6 and C-reactive protein, successful aging, and mortality: the PolSenior study. Immun Ageing. (2016) 13:76. 10.1186/s12979-016-0076-x
    1. Paine NJ, Bosch JA, Ring C, Drayson MT. Veldhuijzen van Zanten JJCS. Induced mild systemic inflammation is associated with impaired ability to improve cognitive task performance by practice. Psychophysiology. (2015) 52:333–41. 10.1111/psyp.12360
    1. Shivakumar V, Debnath M, Venugopal D, Rajasekaran A, Kalmady S V, Subbanna M, et al. . Influence of correlation between HLA-G polymorphism and Interleukin-6 (IL6) gene expression on the risk of schizophrenia. Cytokine. (2018) 107:59–64. 10.1016/j.cyto.2017.11.016
    1. Marsland AL, Gianaros PJ, Kuan DCH, Sheu LK, Krajina K, Manuck SB. Brain morphology links systemic inflammation to cognitive function in midlife adults. Brain Behav Immun. (2015) 48:195–204. 10.1016/j.bbi.2015.03.015
    1. Thielen JW, Kärgel C, Müller BW, Rasche I, Genius J, Bus B, Maderwald S, Norris DG, Wiltfang J, Tendolkar I. Aerobic activity in the healthy elderly is associated with larger plasticity in memory related brain structures and lower systemic inflammation. Front Aging Neurosci. (2016) 8:319. 10.3389/fnagi.2016.00319
    1. Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-α production in humans. FASEB J. (2003) 17:1–10. 10.1096/fj.02-0670fje

Source: PubMed

3
Subscribe