Preprint / Version 1

The Homeostasis of the Gut Brain Axis in Human and Animal Models of Alzheimer's

##article.authors##

  • Anvi Sinha Polygence

DOI:

https://doi.org/10.58445/rars.438

Keywords:

Alzheimer's, Gut-Brain Axis, Neuroscience

Abstract

The Gut-Brain Axis (GBA) is a bidirectional channel through which the enteric nervous system in the abdomen and the central nervous system in the cranium can communicate. The gut microbiome strongly influences the GBA and is composed of a variety of bacterial strains (e.g. E. col, B. bifidum) which are influenced by a variety of factors during growth and development. In this review, I will summarize studies focused on the influences of psychostimulants, early life adversity, and diet on human and animal microbiota as well as their influence on neurodegeneration in the brain. Drugs, stress, and diet are known to increase inflammation and disrupt the blood-brain barrier (BBB). In turn, activation of pro-inflammatory cytokines lead to neuroinflammation in the brain which have been shown to increase the susceptibility to Alzheimer’s disease (AD) and overall cognitive impairment. 

References

References

Socała, K., et al., The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacological Research, 2021. 172: p. 105840. 2. Góralczyk-Bińkowska A, S.-K.D., Kozłowska The Microbiota-Gut-Brain Axis in Psychiatric Disorders. Int J Mol Sci, 2022.

Agirman G, Y.K., Hsiao EY, Signaling inflammation across the gut-brain axis. Science, 2021.

Zhihai Huang, J.D.J., and Quanguang Zhang, Early life adversity as a risk factor for cognitive impairment and Alzheimer’s disease. Translational Neurodegeneration, 2023(25).

Rebecca S. Hofford, S.J.R., Drew D. Kiraly, Neuroimmune mechanisms of psychostimulant and opioid use disorders. Eur J Neurosci, 2020.

Rutsch A, K.J., Ronchi F., The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front Immunol., 2020.

Benjamin Boutrel, G.F.K., What Keeps Us Awake: the Neuropharmacology of Stimulants and Wakefulness Promoting Medications. Sleep, 2004. 27(6).

M. Douglas Anglin, C.B., Brian Perrochet, Ewa Stamper, Samia Dawud-Noursi, History of the Methamphetamine Problem. 2011.

Peter W. Kalivas, N.D.V., The neural basis of addiction: a pathology of motivation and choice

. The American Journal of Psychiatry, 2005.

Rodrigo Arreola, S.A.-H., Gilberto Pérez-Sánchez, Enrique Becerril-Villanueva, Carlos Cruz-Fuentes, Enrique Octavio Flores-Gutierrez, María Eugenia Garcés-Alvarez, Dora Luz de la Cruz-Aguilera, Emilio Medina-Rivero, Gabriela Hurtado-Alvarado, Saray Quintero-Fabián, Lenin Pavón, Immunomodulatory Effects Mediated by Dopamine. Journal of Immunology Research, 2016.

Northcutt, A., Hutchinson, M., Wang, X. et al, DAT isn’t all that: cocaine reward and reinforcement require Toll-like receptor 4 signaling. Mol Psychiatry 2015. 12. Lacagnina, M., Rivera, P. & Bilbo, S., Glial and Neuroimmune Mechanisms as Critical Modulators of Drug Use and Abuse. Neuropsychopharmacol, 2017.

Thanos, P.K., et al., Chronic Methamphetamine Effects on Brain Structure and Function in Rats. PLoS One, 2016. 11(6): p. e0155457.

Chen, H., T. Uz, and H. Manev, Minocycline affects cocaine sensitization in mice. Neurosci Lett, 2009. 452(3): p. 258-61.

Zhang, L. and C.C. Wang, Inflammatory response of macrophages in infection. Hepatobiliary Pancreat Dis Int, 2014. 13(2): p. 138-52.

Northcutt, A.L., et al., DAT isn't all that: cocaine reward and reinforcement require Toll like receptor 4 signaling. Mol Psychiatry, 2015. 20(12): p. 1525-37.

Brown, K.T., et al., Innate immune signaling in the ventral tegmental area contributes to drug-primed reinstatement of cocaine seeking. Brain Behav Immun, 2018. 67: p. 130- 138.

Tanda, G., et al., Lack of Specific Involvement of (+)-Naloxone and (+)-Naltrexone on the Reinforcing and Neurochemical Effects of Cocaine and Opioids.

Neuropsychopharmacology, 2016. 41(11): p. 2772-81.

Wohleb, E.S., T. Franklin, M. Iwata, and R.S. Duman, Integrating neuroimmune systems in the neurobiology of depression. Nat Rev Neurosci, 2016. 17(8): p. 497-511. 20. Lewitus, G.M., et al., Microglial TNF-α Suppresses Cocaine-Induced Plasticity and Behavioral Sensitization. Neuron, 2016. 90(3): p. 483-91.

Guyon, A., et al., Long term exposure to the chemokine CCL2 activates the nigrostriatal dopamine system: a novel mechanism for the control of dopamine release. Neuroscience, 2009. 162(4): p. 1072-80.

Trocello, J.M., et al., Implication of CCR2 chemokine receptor in cocaine-induced sensitization. J Mol Neurosci, 2011. 44(3): p. 147-51.

Trecki, J. and E.M. Unterwald, Modulation of cocaine-induced activity by intracerebral administration of CXCL12. Neuroscience, 2009. 161(1): p. 13-22.

Pedraz, M., et al., Sex differences in psychiatric comorbidity and plasma biomarkers for cocaine addiction in abstinent cocaine-addicted subjects in outpatient settings. Front Psychiatry, 2015. 6: p. 17.

Levandowski, M.L., A.R. Hess, R. Grassi-Oliveira, and R.M. de Almeida, Plasma interleukin-6 and executive function in crack cocaine-dependent women. Neurosci Lett, 2016. 628: p. 85-90.

Fox, H.C., et al., Immune system inflammation in cocaine dependent individuals: implications for medications development. Hum Psychopharmacol, 2012. 27(2): p. 156- 66.

Hodes, G.E., et al., Neuroimmune mechanisms of depression. Nat Neurosci, 2015. 18(10): p. 1386-93.

Little, K.Y., et al., Decreased brain dopamine cell numbers in human cocaine users. Psychiatry Res, 2009. 168(3): p. 173-80.

Sekine, Y., et al., Methamphetamine causes microglial activation in the brains of human abusers. J Neurosci, 2008. 28(22): p. 5756-61.

Narendran, R., et al., Cocaine abuse in humans is not associated with increased microglial activation: an 18-kDa translocator protein positron emission tomography imaging study with [11C]PBR28. J Neurosci, 2014. 34(30): p. 9945-50.

Fung, T.C., C.A. Olson, and E.Y. Hsiao, Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci, 2017. 20(2): p. 145-155. 32. Foster, J.A., L. Rinaman, and J.F. Cryan, Stress & the gut-brain axis: Regulation by the microbiome. Neurobiol Stress, 2017. 7: p. 124-136.

Kiraly, D.D., et al., Alterations of the Host Microbiome Affect Behavioral Responses to Cocaine. Sci Rep, 2016. 6: p. 35455.

Bercik, P., et al., The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology, 2011. 141(2): p. 599-609, 609.e1-3.

Volpe, G.E., et al., Associations of cocaine use and HIV infection with the intestinal microbiota, microbial translocation, and inflammation. J Stud Alcohol Drugs, 2014. 75(2): p. 347-57.

Gareau, M.G., et al., Bacterial infection causes stress-induced memory dysfunction in mice. Gut, 2011. 60(3): p. 307-17.

Carlino, D., M. De Vanna, and E. Tongiorgi, Is altered BDNF biosynthesis a general feature in patients with cognitive dysfunctions? Neuroscientist, 2013. 19(4): p. 345-53. 38. Neufeld, K.M., N. Kang, J. Bienenstock, and J.A. Foster, Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil, 2011. 23(3): p. 255-64, e119.

Ogbonnaya, E.S., et al., Adult Hippocampal Neurogenesis Is Regulated by the Microbiome. Biol Psychiatry, 2015. 78(4): p. e7-9.

Erny, D., et al., Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci, 2015. 18(7): p. 965-77.

Sudo, N., et al., Postnatal microbial colonization programs the hypothalamic-pituitary adrenal system for stress response in mice. J Physiol, 2004. 558(Pt 1): p. 263-75. 42. Lakhan, S.E., M. Caro, and N. Hadzimichalis, NMDA Receptor Activity in Neuropsychiatric Disorders. Front Psychiatry, 2013. 4: p. 52.

Tzeng, N.S., et al., Association between amphetamine-related disorders and dementia-a nationwide cohort study in Taiwan. Ann Clin Transl Neurol, 2020. 7(8): p. 1284-1295. 44. Yuan, B., J. Li, K. Li, and M. Chen, Longstanding health risk across the life course: The influence of early-life experience on health status throughout the life span. J Biosoc Sci, 2023. 55(4): p. 708-734.

Kundakovic, M. and F.A. Champagne, Early-life experience, epigenetics, and the developing brain. Neuropsychopharmacology, 2015. 40(1): p. 141-53.

Cameron, J.L., et al., Social Origins of Developmental Risk for Mental and Physical Illness. J Neurosci, 2017. 37(45): p. 10783-10791.

Tottenham, N., Early Adversity and the Neotenous Human Brain. Biol Psychiatry, 2020. 87(4): p. 350-358.

Hoeijmakers, L., et al., A preclinical perspective on the enhanced vulnerability to Alzheimer's disease after early-life stress. Neurobiol Stress, 2018. 8: p. 172-185. 49. Vanaelst, B., et al., Epidemiological approaches to measure childhood stress. Paediatr Perinat Epidemiol, 2012. 26(3): p. 280-97.

Lacey, R.E. and H. Minnis, Practitioner Review: Twenty years of research with adverse childhood experience scores - Advantages, disadvantages and applications to practice. J Child Psychol Psychiatry, 2020. 61(2): p. 116-130.

Mersky, J.P., J. Topitzes, and A.J. Reynolds, Impacts of adverse childhood experiences on health, mental health, and substance use in early adulthood: a cohort study of an urban, minority sample in the U.S. Child Abuse Negl, 2013. 37(11): p. 917-25.

Felitti, V.J., et al., Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The Adverse Childhood Experiences (ACE) Study. Am J Prev Med, 1998. 14(4): p. 245-58.

Garner, A.S. and J.P. Shonkoff, Early childhood adversity, toxic stress, and the role of the pediatrician: translating developmental science into lifelong health. Pediatrics, 2012. 129(1): p. e224-31.

Lopez, M., et al., The social ecology of childhood and early life adversity. Pediatr Res, 2021. 89(2): p. 353-367.

Christian, C.W., The evaluation of suspected child physical abuse. Pediatrics, 2015. 135(5): p. e1337-54.

Benmhammed, H., et al., Animal Models of Early-Life Adversity. Methods Mol Biol, 2019. 2011: p. 143-161.

Levine, S., Infantile experience and resistance to physiological stress. Science, 1957. 126(3270): p. 405.

Ellenbroek, B.A., N. Derks, and H.J. Park, Early maternal deprivation retards neurodevelopment in Wistar rats. Stress, 2005. 8(4): p. 247-57.

Levis, S.C., T.Z. Baram, and S.V. Mahler, Neurodevelopmental origins of substance use disorders: Evidence from animal models of early-life adversity and addiction. Eur J Neurosci, 2022. 55(9-10): p. 2170-2195.

Zardooz, H., F. Sadeghimahalli, and F. Khodagholi, Early postnatal stress impairs insulin secretion in response to psychological stress in adult rats. J Endocrinol Invest, 2021. 44(2): p. 277-286.

Abbink, M.R., E.F.G. Naninck, P.J. Lucassen, and A. Korosi, Early-life stress diminishes the increase in neurogenesis after exercise in adult female mice. Hippocampus, 2017. 27(8): p. 839-844.

Liu, D., et al., Maternal care, hippocampal synaptogenesis and cognitive development in rats. Nat Neurosci, 2000. 3(8): p. 799-806.

Yajima, H., et al., Early-life stress induces cognitive disorder in middle-aged mice. Neurobiol Aging, 2018. 64: p. 139-146.

Wang, A., et al., Early-Life Stress Alters Synaptic Plasticity and mTOR Signaling: Correlation With Anxiety-Like and Cognition-Related Behavior. Front Genet, 2020. 11: p. 590068.

Marco, E.M., et al., Maternal deprivation effects on brain plasticity and recognition memory in adolescent male and female rats. Neuropharmacology, 2013. 68: p. 223-31. 66. Wang, X.D., et al., Forebrain CRF₁ modulates early-life stress-programmed cognitive deficits. J Neurosci, 2011. 31(38): p. 13625-34.

Naninck, E.F., et al., Chronic early life stress alters developmental and adult neurogenesis and impairs cognitive function in mice. Hippocampus, 2015. 25(3): p. 309- 28.

Lu, C.Y., et al., Effects of Traumatic Stress Induced in the Juvenile Period on the Expression of Gamma-Aminobutyric Acid Receptor Type A Subunits in Adult Rat Brain. Neural Plast, 2017. 2017: p. 5715816.

Avital, A. and G. Richter-Levin, Exposure to juvenile stress exacerbates the behavioural consequences of exposure to stress in the adult rat. Int J Neuropsychopharmacol, 2005. 8(2): p. 163-73.

McLaughlin, K.A., Future Directions in Childhood Adversity and Youth Psychopathology. J Clin Child Adolesc Psychol, 2016. 45(3): p. 361-82.

Loria, A.S., D.H. Ho, and J.S. Pollock, A mechanistic look at the effects of adversity early in life on cardiovascular disease risk during adulthood. Acta Physiol (Oxf), 2014. 210(2): p. 277-87.

Bethell, C.D., et al., Methods to Assess Adverse Childhood Experiences of Children and Families: Toward Approaches to Promote Child Well-being in Policy and Practice. Acad Pediatr, 2017. 17(7s): p. S51-s69.

Oral, R., et al., Adverse childhood experiences and trauma informed care: the future of health care. Pediatr Res, 2016. 79(1-2): p. 227-33.

Holmes, T.H. and R.H. Rahe, The Social Readjustment Rating Scale. J Psychosom Res, 1967. 11(2): p. 213-8.

Rutter, M., Family, area and school influences in the genesis of conduct disorders. Book Suppl J Child Psychol Psychiatr, 1978(1): p. 95-113.

Sameroff, A., R. Seifer, M. Zax, and R. Barocas, Early indicators of developmental risk: Rochester Longitudinal Study. Schizophr Bull, 1987. 13(3): p. 383-94.

Dong, M., et al., The relationship of exposure to childhood sexual abuse to other forms of abuse, neglect, and household dysfunction during childhood. Child Abuse Negl, 2003. 27(6): p. 625-39.

Brown, D.W., et al., Adverse childhood experiences are associated with the risk of lung cancer: a prospective cohort study. BMC Public Health, 2010. 10: p. 20.

Thompson, L.A., et al., Specific adverse childhood experiences and their association with other adverse childhood experiences, asthma and emotional, developmental and behavioral problems in childhood. Pediatr Res, 2020. 88(1): p. 100-109.

Felitti, V.J., Adverse childhood experiences and adult health. Acad Pediatr, 2009. 9(3): p. 131-2.

Lobo, F.M. and E. Lunkenheimer, Understanding the parent-child coregulation patterns shaping child self-regulation. Dev Psychol, 2020. 56(6): p. 1121-1134.

Paley, B. and N.J. Hajal, Conceptualizing Emotion Regulation and Coregulation as Family-Level Phenomena. Clin Child Fam Psychol Rev, 2022. 25(1): p. 19-43. 83. Pesonen, A.K., et al., Cognitive ability and decline after early life stress exposure. Neurobiol Aging, 2013. 34(6): p. 1674-9.

Wade, M., C.H. Zeanah, N.A. Fox, and C.A. Nelson, Global deficits in executive functioning are transdiagnostic mediators between severe childhood neglect and psychopathology in adolescence. Psychol Med, 2020. 50(10): p. 1687-1694.

Saleh, A., et al., Effects of early life stress on depression, cognitive performance and brain morphology. Psychol Med, 2017. 47(1): p. 171-181.

Cryan, J.F., et al., The Microbiota-Gut-Brain Axis. Physiol Rev, 2019. 99(4): p. 1877- 2013.

Osadchiy, V., C.R. Martin, and E.A. Mayer, The Gut-Brain Axis and the Microbiome: Mechanisms and Clinical Implications. Clin Gastroenterol Hepatol, 2019. 17(2): p. 322- 332.

Clarke, G., et al., The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry, 2013. 18(6): p. 666-73.

Deng, Y., et al., Involvement of the microbiota-gut-brain axis in chronic restraint stress: disturbances of the kynurenine metabolic pathway in both the gut and brain. Gut Microbes, 2021. 13(1): p. 1-16.

O'Mahony, S.M., et al., Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol Psychiatry, 2009. 65(3): p. 263-7.

Enqi, W., S. Jingzhu, P. Lingpeng, and L. Yaqin, Comparison of the Gut Microbiota Disturbance in Rat Models of Irritable Bowel Syndrome Induced by Maternal Separation and Multiple Early-Life Adversity. Front Cell Infect Microbiol, 2020. 10: p. 581974.

Rincel, M., et al., Multi-hit early life adversity affects gut microbiota, brain and behavior in a sex-dependent manner. Brain Behav Immun, 2019. 80: p. 179-192.

Park, H.J., S.A. Kim, W.S. Kang, and J.W. Kim, Early-Life Stress Modulates Gut Microbiota and Peripheral and Central Inflammation in a Sex-Dependent Manner. Int J Mol Sci, 2021. 22(4).

Jašarević, E., C.L. Howerton, C.D. Howard, and T.L. Bale, Alterations in the Vaginal Microbiome by Maternal Stress Are Associated With Metabolic Reprogramming of the Offspring Gut and Brain. Endocrinology, 2015. 156(9): p. 3265-76.

Bailey, M.T. and C.L. Coe, Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Dev Psychobiol, 1999. 35(2): p. 146-55. 96. Amini-Khoei, H., et al., On the role of corticosterone in behavioral disorders, microbiota composition alteration and neuroimmune response in adult male mice subjected to maternal separation stress. Int Immunopharmacol, 2019. 66: p. 242-250.

Pusceddu, M.M., et al., N-3 Polyunsaturated Fatty Acids (PUFAs) Reverse the Impact of Early-Life Stress on the Gut Microbiota. PLoS One, 2015. 10(10): p. e0139721. 98. Bailey, M.T., et al., Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation. Brain Behav Immun, 2011. 25(3): p. 397-407.

Galley, J.D. and M.T. Bailey, Impact of stressor exposure on the interplay between commensal microbiota and host inflammation. Gut Microbes, 2014. 5(3): p. 390-6. 100. Allen, R.G., et al., The intestinal microbiota are necessary for stressor-induced enhancement of splenic macrophage microbicidal activity. Brain Behav Immun, 2012. 26(3): p. 371-82.

Rincel, M., et al., Pharmacological restoration of gut barrier function in stressed neonates partially reverses long-term alterations associated with maternal separation. Psychopharmacology (Berl), 2019. 236(5): p. 1583-1596.

El Aidy, S., et al., Serotonin Transporter Genotype Modulates the Gut Microbiota Composition in Young Rats, an Effect Augmented by Early Life Stress. Front Cell Neurosci, 2017. 11: p. 222.

Hantsoo, L., et al., Childhood adversity impact on gut microbiota and inflammatory response to stress during pregnancy. Brain Behav Immun, 2019. 75: p. 240-250. 104. D'Agata, A.L., et al., Effects of early life NICU stress on the developing gut microbiome. Dev Psychobiol, 2019. 61(5): p. 650-660.

Flannery, J.E., et al., Gut Feelings Begin in Childhood: the Gut Metagenome Correlates with Early Environment, Caregiving, and Behavior. mBio, 2020. 11(1).

Callaghan, B.L., et al., Mind and gut: Associations between mood and gastrointestinal distress in children exposed to adversity. Dev Psychopathol, 2020. 32(1): p. 309-328. 107. Coley, E.J.L., et al., Early life adversity predicts brain-gut alterations associated with increased stress and mood. Neurobiol Stress, 2021. 15: p. 100348.

Zhuang, Z.Q., et al., Gut Microbiota is Altered in Patients with Alzheimer's Disease. J Alzheimers Dis, 2018. 63(4): p. 1337-1346.

Li, B., et al., Mild cognitive impairment has similar alterations as Alzheimer's disease in gut microbiota. Alzheimers Dement, 2019. 15(10): p. 1357-1366.

Hoeijmakers, L., et al., Early-life stress lastingly alters the neuroinflammatory response to amyloid pathology in an Alzheimer's disease mouse model. Brain Behav Immun, 2017. 63: p. 160-175.

Lesuis, S.L., et al., Targeting glucocorticoid receptors prevents the effects of early life stress on amyloid pathology and cognitive performance in APP/PS1 mice. Transl Psychiatry, 2018. 8(1): p. 53.

Hui, J., et al., Maternal separation exacerbates Alzheimer's disease-like behavioral and pathological changes in adult APPswe/PS1dE9 mice. Behav Brain Res, 2017. 318: p. 18- 23.

Tanaka, T., et al., Early-life stress induces the development of Alzheimer's disease pathology via angiopathy. Exp Neurol, 2021. 337: p. 113552.

Kim, M.S., et al., Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer's disease animal model. Gut, 2020. 69(2): p. 283-294.

Ma, X., et al., Early-Life Intervention Using Exogenous Fecal Microbiota Alleviates Gut Injury and Reduce Inflammation Caused by Weaning Stress in Piglets. Front Microbiol, 2021. 12: p. 671683.

Roque, A., A. Ochoa-Zarzosa, and L. Torner, Maternal separation activates microglial cells and induces an inflammatory response in the hippocampus of male rat pups, independently of hypothalamic and peripheral cytokine levels. Brain Behav Immun, 2016. 55: p. 39-48.

Gómez-González, B. and A. Escobar, Altered functional development of the blood-brain barrier after early life stress in the rat. Brain Res Bull, 2009. 79(6): p. 376-87. 118. Norton, M.C., et al., Early parental death and remarriage of widowed parents as risk factors for Alzheimer disease: the Cache County study. Am J Geriatr Psychiatry, 2011. 19(9): p. 814-24.

Radford, K., et al., Childhood Stress and Adversity is Associated with Late-Life Dementia in Aboriginal Australians. Am J Geriatr Psychiatry, 2017. 25(10): p. 1097-1106. 120. Donley, G.A.R., E. Lönnroos, T.P. Tuomainen, and J. Kauhanen, Association of childhood stress with late-life dementia and Alzheimer's disease: the KIHD study. Eur J Public Health, 2018. 28(6): p. 1069-1073.

Tani, Y., T. Fujiwara, and K. Kondo, Association Between Adverse Childhood Experiences and Dementia in Older Japanese Adults. JAMA Netw Open, 2020. 3(2): p. e1920740.

Tani, Y., T. Fujiwara, and K. Kondo, Adverse Childhood Experiences and Dementia: Interactions With Social Capital in the Japan Gerontological Evaluation Study Cohort. Am J Prev Med, 2021. 61(2): p. 225-234.

Fernandez-Real, J.M., et al., Gut Microbiota Interacts With Brain Microstructure and Function. J Clin Endocrinol Metab, 2015. 100(12): p. 4505-13.

Akbari, E., et al., Effect of Probiotic Supplementation on Cognitive Function and Metabolic Status in Alzheimer's Disease: A Randomized, Double-Blind and Controlled Trial. Front Aging Neurosci, 2016. 8: p. 256.

Roman, P., et al., A Pilot Randomized Controlled Trial to Explore Cognitive and Emotional Effects of Probiotics in Fibromyalgia. Sci Rep, 2018. 8(1): p. 10965. 126. Malatji, B.G., et al., The GC-MS metabolomics signature in patients with fibromyalgia syndrome directs to dysbiosis as an aspect contributing factor of FMS pathophysiology. Metabolomics, 2019. 15(4): p. 54.

Braak, H. and E. Braak, Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol, 1991. 82(4): p. 239-59.

Radde, R., et al., Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep, 2006. 7(9): p. 940-6.

Harach, T., et al., Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci Rep, 2017. 7: p. 41802.

Kumar, D.K., et al., Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer's disease. Sci Transl Med, 2016. 8(340): p. 340ra72. 131. Stilling, R.M. and J.F. Cryan, Host response: A trigger for neurodegeneration? Nat Microbiol, 2016. 1(8): p. 16129.

Vogt, N.M., et al., Gut microbiome alterations in Alzheimer's disease. Sci Rep, 2017. 7(1): p. 13537.

Arnold, S.E., et al., Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol, 2018. 14(3): p. 168-181.

Manderino, L., et al., Preliminary Evidence for an Association Between the Composition of the Gut Microbiome and Cognitive Function in Neurologically Healthy Older Adults. J Int Neuropsychol Soc, 2017. 23(8): p. 700-705.

Cattaneo, A., et al., Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging, 2017. 49: p. 60-68.

Downloads

Posted

2023-09-13