The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives
Abstract
:1. Introduction
2. Microglia in Health and Neurodegenerative Disease
3. Microglia Are under the Influence of Peripheral Inflammation
4. Obesity-Associated Chronic Peripheral Inflammation
5. Obesity and Risk of Neurological Disease
6. The Impact of Obesity on Microglia-Mediated Neuroinflammation
7. Mechanisms Mediating Obesity-Associated Neuroinflammation
7.1. The Impact of Obesity on Blood-Brain Barrier Function
7.2. Circulating Monocytes
7.3. Cytokines
7.4. Adipokines
7.5. Dyslipidemia
7.6. Insulin Resistance
7.7. Glucocorticoids
7.8. The Gut-Brain Axis
8. Conclusions and Future Perspectives
Funding
Conflicts of Interest
References
- Bluher, M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298. [Google Scholar] [CrossRef] [PubMed]
- Heymsfield, S.B.; Wadden, T.A. Mechanisms, pathophysiology, and management of obesity. N. Engl. J. Med. 2017, 376, 254–266. [Google Scholar] [CrossRef]
- Kivipelto, M.; Ngandu, T.; Fratiglioni, L.; Viitanen, M.; Kareholt, I.; Winblad, B.; Helkala, E.L.; Tuomilehto, J.; Soininen, H.; Nissinen, A. Obesity and vascular risk factors at midlife and the risk of dementia and Alzheimer disease. Arch. Neurol. 2005, 62, 1556–1560. [Google Scholar] [CrossRef] [Green Version]
- Whitmer, R.A.; Gustafson, D.R.; Barrett-Connor, E.; Haan, M.N.; Gunderson, E.P.; Yaffe, K. Central obesity and increased risk of dementia more than three decades later. Neurology 2008, 71, 1057–1064. [Google Scholar] [CrossRef]
- Whitmer, R.A.; Gunderson, E.P.; Barrett-Connor, E.; Quesenberry, C.P., Jr.; Yaffe, K. Obesity in middle age and future risk of dementia: A 27 year longitudinal population based study. BMJ 2005, 330, 1360. [Google Scholar] [CrossRef] [Green Version]
- Hassing, L.B.; Dahl, A.K.; Thorvaldsson, V.; Berg, S.; Gatz, M.; Pedersen, N.L.; Johansson, B. Overweight in midlife and risk of dementia: A 40-year follow-up study. Int. J. Obes. 2009, 33, 893–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eskelinen, M.H.; Ngandu, T.; Helkala, E.L.; Tuomilehto, J.; Nissinen, A.; Soininen, H.; Kivipelto, M. Fat intake at midlife and cognitive impairment later in life: A population-based CAIDE study. Int. J. Geriatr. Psychiatry 2008, 23, 741–747. [Google Scholar] [CrossRef]
- Miller, A.A.; Spencer, S.J. Obesity and neuroinflammation: A pathway to cognitive impairment. Brain Behav. Immun. 2014, 42, 10–21. [Google Scholar] [CrossRef] [PubMed]
- O’Brien, P.D.; Hinder, L.M.; Callaghan, B.C.; Feldman, E.L. Neurological consequences of obesity. Lancet Neurol. 2017, 16, 465–477. [Google Scholar] [CrossRef]
- Michailidou, Z.; Gomez-Salazar, M.; Alexaki, V.I. Innate immune cells in the adipose tissue in health and metabolic disease. J. Innate Immun. 2021, 1–27. [Google Scholar] [CrossRef] [PubMed]
- Nati, M.; Haddad, D.; Birkenfeld, A.L.; Koch, C.A.; Chavakis, T.; Chatzigeorgiou, A. The role of immune cells in metabolism-related liver inflammation and development of non-alcoholic steatohepatitis (NASH). Rev. Endocrinol. Metab. Disord. 2016, 17, 29–39. [Google Scholar] [CrossRef] [PubMed]
- Guillemot-Legris, O.; Muccioli, G.G. Obesity-induced neuroinflammation: Beyond the hypothalamus. Trends Neurosci. 2017, 40, 237–253. [Google Scholar] [CrossRef] [PubMed]
- Alexaki, V.I.; Fodelianaki, G.; Neuwirth, A.; Mund, C.; Kourgiantaki, A.; Ieronimaki, E.; Lyroni, K.; Troullinaki, M.; Fujii, C.; Kanczkowski, W.; et al. DHEA inhibits acute microglia-mediated inflammation through activation of the TrkA-Akt1/2-CREB-Jmjd3 pathway. Mol. Psychiatry 2018, 23, 1410–1420. [Google Scholar] [CrossRef]
- Yilmaz, C.; Karali, K.; Fodelianaki, G.; Gravanis, A.; Chavakis, T.; Charalampopoulos, I.; Alexaki, V.I. Neurosteroids as regulators of neuroinflammation. Front. Neuroendocrinol. 2019, 55, 100788. [Google Scholar] [CrossRef]
- Prinz, M.; Jung, S.; Priller, J. Microglia biology: One century of evolving concepts. Cell 2019, 179, 292–311. [Google Scholar] [CrossRef]
- Li, Q.; Barres, B.A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 2018, 18, 225–242. [Google Scholar] [CrossRef] [PubMed]
- Bohlen, C.J.; Friedman, B.A.; Dejanovic, B.; Sheng, M. Microglia in brain development, homeostasis, and neurodegeneration. Annu. Rev. Genet. 2019, 53, 263–288. [Google Scholar] [CrossRef]
- Eyo, U.B.; Wu, L.J. Microglia: Lifelong patrolling immune cells of the brain. Prog. Neurobiol. 2019, 179, 101614. [Google Scholar] [CrossRef]
- Kierdorf, K.; Erny, D.; Goldmann, T.; Sander, V.; Schulz, C.; Perdiguero, E.G.; Wieghofer, P.; Heinrich, A.; Riemke, P.; Holscher, C.; et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 2013, 16, 273–280. [Google Scholar] [CrossRef]
- Huang, Y.; Xu, Z.; Xiong, S.; Sun, F.; Qin, G.; Hu, G.; Wang, J.; Zhao, L.; Liang, Y.X.; Wu, T.; et al. Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion. Nat. Neurosci. 2018, 21, 530–540. [Google Scholar] [CrossRef]
- Fuger, P.; Hefendehl, J.K.; Veeraraghavalu, K.; Wendeln, A.C.; Schlosser, C.; Obermuller, U.; Wegenast-Braun, B.M.; Neher, J.J.; Martus, P.; Kohsaka, S.; et al. Microglia turnover with aging and in an Alzheimer’s model via long-term in vivo single-cell imaging. Nat. Neurosci. 2017, 20, 1371–1376. [Google Scholar] [CrossRef]
- Tay, T.L.; Mai, D.; Dautzenberg, J.; Fernandez-Klett, F.; Lin, G.; Sagar Datta, M.; Drougard, A.; Stempfl, T.; Ardura-Fabregat, A.; Staszewski, O.; et al. A new fate mapping system reveals context-dependent random or clonal expansion of microglia. Nat. Neurosci. 2017, 20, 793–803. [Google Scholar] [CrossRef] [PubMed]
- Askew, K.; Li, K.; Olmos-Alonso, A.; Garcia-Moreno, F.; Liang, Y.; Richardson, P.; Tipton, T.; Chapman, M.A.; Riecken, K.; Beccari, S.; et al. Coupled proliferation and apoptosis maintain the rapid turnover of microglia in the adult brain. Cell Rep. 2017, 18, 391–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Butovsky, O.; Jedrychowski, M.P.; Moore, C.S.; Cialic, R.; Lanser, A.J.; Gabriely, G.; Koeglsperger, T.; Dake, B.; Wu, P.M.; Doykan, C.E.; et al. Identification of a unique TGF-beta-dependent molecular and functional signature in microglia. Nat. Neurosci. 2014, 17, 131–143. [Google Scholar] [CrossRef] [Green Version]
- Gosselin, D.; Skola, D.; Coufal, N.G.; Holtman, I.R.; Schlachetzki, J.C.M.; Sajti, E.; Jaeger, B.N.; O’Connor, C.; Fitzpatrick, C.; Pasillas, M.P.; et al. An environment-dependent transcriptional network specifies human microglia identity. Science 2017, 356, 6344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galatro, T.F.; Holtman, I.R.; Lerario, A.M.; Vainchtein, I.D.; Brouwer, N.; Sola, P.R.; Veras, M.M.; Pereira, T.F.; Leite, R.E.P.; Moller, T.; et al. Transcriptomic analysis of purified human cortical microglia reveals age-associated changes. Nat. Neurosci. 2017, 20, 1162–1171. [Google Scholar] [CrossRef] [PubMed]
- Gosselin, D.; Link, V.M.; Romanoski, C.E.; Fonseca, G.J.; Eichenfield, D.Z.; Spann, N.J.; Stender, J.D.; Chun, H.B.; Garner, H.; Geissmann, F.; et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 2014, 159, 1327–1340. [Google Scholar] [CrossRef] [PubMed]
- Grabert, K.; Michoel, T.; Karavolos, M.H.; Clohisey, S.; Baillie, J.K.; Stevens, M.P.; Freeman, T.C.; Summers, K.M.; McColl, B.W. Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nat. Neurosci. 2016, 19, 504–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Butovsky, O.; Weiner, H.L. Microglial signatures and their role in health and disease. Nat. Rev. Neurosci. 2018, 19, 622–635. [Google Scholar] [CrossRef]
- Bennett, M.L.; Bennett, F.C.; Liddelow, S.A.; Ajami, B.; Zamanian, J.L.; Fernhoff, N.B.; Mulinyawe, S.B.; Bohlen, C.J.; Adil, A.; Tucker, A.; et al. New tools for studying microglia in the mouse and human CNS. Proc. Natl. Acad. Sci. USA 2016, 113, E1738–E1746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zrzavy, T.; Hametner, S.; Wimmer, I.; Butovsky, O.; Weiner, H.L.; Lassmann, H. Loss of ‘homeostatic’ microglia and patterns of their activation in active multiple sclerosis. Brain 2017, 140, 1900–1913. [Google Scholar] [CrossRef]
- Villa, A.; Gelosa, P.; Castiglioni, L.; Cimino, M.; Rizzi, N.; Pepe, G.; Lolli, F.; Marcello, E.; Sironi, L.; Vegeto, E.; et al. Sex-specific features of microglia from adult mice. Cell Rep. 2018, 23, 3501–3511. [Google Scholar] [CrossRef]
- Davalos, D.; Grutzendler, J.; Yang, G.; Kim, J.V.; Zuo, Y.; Jung, S.; Littman, D.R.; Dustin, M.L.; Gan, W.B. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 2005, 8, 752–758. [Google Scholar] [CrossRef]
- Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef] [Green Version]
- Miyamoto, A.; Wake, H.; Ishikawa, A.W.; Eto, K.; Shibata, K.; Murakoshi, H.; Koizumi, S.; Moorhouse, A.J.; Yoshimura, Y.; Nabekura, J. Microglia contact induces synapse formation in developing somatosensory cortex. Nat. Commun. 2016, 7, 12540. [Google Scholar] [CrossRef] [Green Version]
- Wake, H.; Moorhouse, A.J.; Jinno, S.; Kohsaka, S.; Nabekura, J. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J. Neurosci. 2009, 29, 3974–3980. [Google Scholar] [CrossRef] [Green Version]
- Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef] [Green Version]
- Schafer, D.P.; Lehrman, E.K.; Kautzman, A.G.; Koyama, R.; Mardinly, A.R.; Yamasaki, R.; Ransohoff, R.M.; Greenberg, M.E.; Barres, B.A.; Stevens, B. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 2012, 74, 691–705. [Google Scholar] [CrossRef] [Green Version]
- Stevens, B.; Allen, N.J.; Vazquez, L.E.; Howell, G.R.; Christopherson, K.S.; Nouri, N.; Micheva, K.D.; Mehalow, A.K.; Huberman, A.D.; Stafford, B.; et al. The classical complement cascade mediates CNS synapse elimination. Cell 2007, 131, 1164–1178. [Google Scholar] [CrossRef] [Green Version]
- Stephan, A.H.; Barres, B.A.; Stevens, B. The complement system: An unexpected role in synaptic pruning during development and disease. Annu. Rev. Neurosci. 2012, 35, 369–389. [Google Scholar] [CrossRef] [Green Version]
- Selkoe, D.J. Alzheimer’s disease is a synaptic failure. Science 2002, 298, 789–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vasek, M.J.; Garber, C.; Dorsey, D.; Durrant, D.M.; Bollman, B.; Soung, A.; Yu, J.; Perez-Torres, C.; Frouin, A.; Wilton, D.K.; et al. A complement-microglial axis drives synapse loss during virus-induced memory impairment. Nature 2016, 534, 538–543. [Google Scholar] [CrossRef] [Green Version]
- Parkhurst, C.N.; Yang, G.; Ninan, I.; Savas, J.N.; Yates, J.R., 3rd; Lafaille, J.J.; Hempstead, B.L.; Littman, D.R.; Gan, W.B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 2013, 155, 1596–1609. [Google Scholar] [CrossRef] [Green Version]
- Kettenmann, H.; Kirchhoff, F.; Verkhratsky, A. Microglia: New roles for the synaptic stripper. Neuron 2013, 77, 10–18. [Google Scholar] [CrossRef] [Green Version]
- Fodelianaki, G.; Lansing, F.; Bhattarai, P.; Troullinaki, M.; Zeballos, M.A.; Charalampopoulos, I.; Gravanis, A.; Mirtschink, P.; Chavakis, T.; Alexaki, V.I. Nerve growth factor modulates LPS-induced microglial glycolysis and inflammatory responses. Exp. Cell Res. 2019, 377, 10–16. [Google Scholar] [CrossRef] [PubMed]
- Rizzi, C.; Tiberi, A.; Giustizieri, M.; Marrone, M.C.; Gobbo, F.; Carucci, N.M.; Meli, G.; Arisi, I.; D’Onofrio, M.; Marinelli, S.; et al. NGF steers microglia toward a neuroprotective phenotype. Glia 2018, 66, 1395–1416. [Google Scholar] [CrossRef] [Green Version]
- Wu, S.Y.; Pan, B.S.; Tsai, S.F.; Chiang, Y.T.; Huang, B.M.; Mo, F.E.; Kuo, Y.M. BDNF reverses aging-related microglial activation. J. Neuroinflamm. 2020, 17, 210. [Google Scholar] [CrossRef]
- Harley, S.B.R.; Willis, E.F.; Shaikh, S.N.; Blackmore, D.G.; Sah, P.; Ruitenberg, M.J.; Bartlett, P.F.; Vukovic, J. Selective ablation of BDNF from microglia reveals novel roles in self-renewal and hippocampal neurogenesis. J. Neurosci. 2021, 41, 4172–4186. [Google Scholar] [CrossRef]
- Hines, D.J.; Hines, R.M.; Mulligan, S.J.; Macvicar, B.A. Microglia processes block the spread of damage in the brain and require functional chloride channels. Glia 2009, 57, 1610–1618. [Google Scholar] [CrossRef]
- Szalay, G.; Martinecz, B.; Lenart, N.; Kornyei, Z.; Orsolits, B.; Judak, L.; Csaszar, E.; Fekete, R.; West, B.L.; Katona, G.; et al. Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke. Nat. Commun. 2016, 7, 11499. [Google Scholar] [CrossRef] [Green Version]
- David, S.; Kroner, A. Repertoire of microglial and macrophage responses after spinal cord injury. Nat. Rev. Neurosci. 2011, 12, 388–399. [Google Scholar] [CrossRef]
- Kuno, R.; Wang, J.; Kawanokuchi, J.; Takeuchi, H.; Mizuno, T.; Suzumura, A. Autocrine activation of microglia by tumor necrosis factor-alpha. J. Neuroimmunol. 2005, 162, 89–96. [Google Scholar] [CrossRef] [PubMed]
- Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Munch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed]
- Galloway, D.A.; Phillips, A.E.M.; Owen, D.R.J.; Moore, C.S. Phagocytosis in the brain: Homeostasis and disease. Front. Immunol. 2019, 10, 790. [Google Scholar] [CrossRef] [Green Version]
- Mike, J.K.; Ferriero, D.M. Efferocytosis mediated modulation of injury after neonatal brain hypoxia-ischemia. Cells 2021, 10, 1025. [Google Scholar] [CrossRef] [PubMed]
- Hill, R.A.; Li, A.M.; Grutzendler, J. Lifelong cortical myelin plasticity and age-related degeneration in the live mammalian brain. Nat. Neurosci. 2018, 21, 683–695. [Google Scholar] [CrossRef]
- Safaiyan, S.; Kannaiyan, N.; Snaidero, N.; Brioschi, S.; Biber, K.; Yona, S.; Edinger, A.L.; Jung, S.; Rossner, M.J.; Simons, M. Age-related myelin degradation burdens the clearance function of microglia during aging. Nat. Neurosci. 2016, 19, 995–998. [Google Scholar] [CrossRef]
- Robinson, S.; Miller, R.H. Contact with central nervous system myelin inhibits oligodendrocyte progenitor maturation. Dev. Biol. 1999, 216, 359–368. [Google Scholar] [CrossRef] [Green Version]
- Kotter, M.R.; Li, W.W.; Zhao, C.; Franklin, R.J. Myelin impairs CNS remyelination by inhibiting oligodendrocyte precursor cell differentiation. J. Neurosci. 2006, 26, 328–332. [Google Scholar] [CrossRef] [PubMed]
- Mawuenyega, K.G.; Sigurdson, W.; Ovod, V.; Munsell, L.; Kasten, T.; Morris, J.C.; Yarasheski, K.E.; Bateman, R.J. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science 2010, 330, 1774. [Google Scholar] [CrossRef] [Green Version]
- Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [Green Version]
- Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms underlying inflammation in neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hefendehl, J.K.; Neher, J.J.; Suhs, R.B.; Kohsaka, S.; Skodras, A.; Jucker, M. Homeostatic and injury-induced microglia behavior in the aging brain. Aging Cell 2014, 13, 60–69. [Google Scholar] [CrossRef]
- Sierra, A.; Gottfried-Blackmore, A.C.; McEwen, B.S.; Bulloch, K. Microglia derived from aging mice exhibit an altered inflammatory profile. Glia 2007, 55, 412–424. [Google Scholar] [CrossRef] [PubMed]
- Floden, A.M.; Combs, C.K. Microglia demonstrate age-dependent interaction with amyloid-beta fibrils. J. Alzheimers Dis. 2011, 25, 279–293. [Google Scholar] [CrossRef] [PubMed]
- Cantuti-Castelvetri, L.; Fitzner, D.; Bosch-Queralt, M.; Weil, M.T.; Su, M.; Sen, P.; Ruhwedel, T.; Mitkovski, M.; Trendelenburg, G.; Lutjohann, D.; et al. Defective cholesterol clearance limits remyelination in the aged central nervous system. Science 2018, 359, 684–688. [Google Scholar] [CrossRef] [Green Version]
- Streit, W.J.; Sammons, N.W.; Kuhns, A.J.; Sparks, D.L. Dystrophic microglia in the aging human brain. Glia 2004, 45, 208–212. [Google Scholar] [CrossRef]
- Savage, J.C.; Carrier, M.; Tremblay, M.E. Morphology of microglia across contexts of health and disease. Methods Mol. Biol. 2019, 2034, 13–26. [Google Scholar] [PubMed]
- Streit, W.J. Microglial senescence: Does the brain’s immune system have an expiration date? Trends Neurosci. 2006, 29, 506–510. [Google Scholar] [CrossRef] [PubMed]
- Bussian, T.J.; Aziz, A.; Meyer, C.F.; Swenson, B.L.; van Deursen, J.M.; Baker, D.J. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 2018, 562, 578–582. [Google Scholar] [CrossRef]
- Streit, W.J.; Braak, H.; Xue, Q.S.; Bechmann, I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 2009, 118, 475–485. [Google Scholar] [CrossRef] [Green Version]
- Angelova, D.M.; Brown, D.R. Microglia and the aging brain: Are senescent microglia the key to neurodegeneration? J. Neurochem. 2019, 151, 676–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kapralov, A.A.; Yang, Q.; Dar, H.H.; Tyurina, Y.Y.; Anthonymuthu, T.S.; Kim, R.; St. Croix, C.M.; Mikulska-Ruminska, K.; Liu, B.; Shrivastava, I.H.; et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat. Chem. Biol. 2020, 16, 278–290. [Google Scholar] [CrossRef]
- Perry, V.H.; Holmes, C. Microglial priming in neurodegenerative disease. Nat. Rev. Neurol. 2014, 10, 217–224. [Google Scholar] [CrossRef] [PubMed]
- Song, W.M.; Colonna, M. The identity and function of microglia in neurodegeneration. Nat. Immunol. 2018, 19, 1048–1058. [Google Scholar] [CrossRef]
- Keren-Shaul, H.; Spinrad, A.; Weiner, A.; Matcovitch-Natan, O.; Dvir-Szternfeld, R.; Ulland, T.K.; David, E.; Baruch, K.; Lara-Astaiso, D.; Toth, B.; et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 2017, 169, 1276–1290.e17. [Google Scholar] [CrossRef]
- Jordao, M.J.C.; Sankowski, R.; Brendecke, S.M.; Sagar Locatelli, G.; Tai, Y.H.; Tay, T.L.; Schramm, E.; Armbruster, S.; Hagemeyer, N.; Gross, O.; et al. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 2019, 363, 6425. [Google Scholar] [CrossRef]
- Krasemann, S.; Madore, C.; Cialic, R.; Baufeld, C.; Calcagno, N.; El Fatimy, R.; Beckers, L.; O’Loughlin, E.; Xu, Y.; Fanek, Z.; et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 2017, 47, 566–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Srinivasan, K.; Friedman, B.A.; Etxeberria, A.; Huntley, M.A.; van der Brug, M.P.; Foreman, O.; Paw, J.S.; Modrusan, Z.; Beach, T.G.; Serrano, G.E.; et al. Alzheimer’s patient microglia exhibit enhanced aging and unique transcriptional activation. Cell Rep. 2020, 31, 107843. [Google Scholar] [CrossRef]
- Sala Frigerio, C.; Wolfs, L.; Fattorelli, N.; Thrupp, N.; Voytyuk, I.; Schmidt, I.; Mancuso, R.; Chen, W.T.; Woodbury, M.E.; Srivastava, G.; et al. The major risk factors for Alzheimer’s disease: Age, sex, and genes modulate the microglia response to abeta plaques. Cell Rep. 2019, 27, 1293–1306.e6. [Google Scholar] [CrossRef] [Green Version]
- Ransohoff, R.M. How neuroinflammation contributes to neurodegeneration. Science 2016, 353, 777–783. [Google Scholar] [CrossRef]
- Salter, M.W.; Stevens, B. Microglia emerge as central players in brain disease. Nat. Med. 2017, 23, 1018–1027. [Google Scholar] [CrossRef]
- Dokalis, N.; Prinz, M. Resolution of neuroinflammation: Mechanisms and potential therapeutic option. Semin. Immunopathol. 2019, 41, 699–709. [Google Scholar] [CrossRef] [PubMed]
- Cunha, M.I.; Su, M.; Cantuti-Castelvetri, L.; Muller, S.A.; Schifferer, M.; Djannatian, M.; Alexopoulos, I.; van der Meer, F.; Winkler, A.; van Ham, T.J.; et al. Pro-inflammatory activation following demyelination is required for myelin clearance and oligodendrogenesis. J. Exp. Med. 2020, 217, e20191390. [Google Scholar] [CrossRef] [Green Version]
- Dantzer, R. Neuroimmune Interactions: From the brain to the immune system and vice versa. Physiol. Rev. 2018, 98, 477–504. [Google Scholar] [CrossRef]
- Chrousos, G.P. The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N. Engl. J. Med. 1995, 332, 1351–1362. [Google Scholar] [CrossRef]
- Rivest, S. Regulation of innate immune responses in the brain. Nat. Rev. Immunol. 2009, 9, 429–439. [Google Scholar] [CrossRef]
- Banks, W.A.; Kastin, A.J.; Broadwell, R.D. Passage of cytokines across the blood-brain barrier. Neuroimmunomodulation 1995, 2, 241–248. [Google Scholar] [CrossRef]
- Pan, W.; Kastin, A.J. TNFalpha transport across the blood-brain barrier is abolished in receptor knockout mice. Exp. Neurol. 2002, 174, 193–200. [Google Scholar] [CrossRef]
- Blecharz-Lang, K.G.; Wagner, J.; Fries, A.; Nieminen-Kelha, M.; Rosner, J.; Schneider, U.C.; Vajkoczy, P. Interleukin 6-mediated endothelial barrier disturbances can be attenuated by blockade of the IL6 receptor expressed in brain microvascular endothelial cells. Transl. Stroke Res. 2018, 9, 631–642. [Google Scholar] [CrossRef]
- Wendeln, A.C.; Degenhardt, K.; Kaurani, L.; Gertig, M.; Ulas, T.; Jain, G.; Wagner, J.; Hasler, L.M.; Wild, K.; Skodras, A.; et al. Innate immune memory in the brain shapes neurological disease hallmarks. Nature 2018, 556, 332–338. [Google Scholar] [CrossRef]
- Bras, J.P.; Bravo, J.; Freitas, J.; Barbosa, M.A.; Santos, S.G.; Summavielle, T.; Almeida, M.I. TNF-alpha-induced microglia activation requires miR-342: Impact on NF-kB signaling and neurotoxicity. Cell Death Dis. 2020, 11, 415. [Google Scholar] [CrossRef]
- Garner, K.M.; Amin, R.; Johnson, R.W.; Scarlett, E.J.; Burton, M.D. Microglia priming by interleukin-6 signaling is enhanced in aged mice. J. Neuroimmunol. 2018, 324, 90–99. [Google Scholar] [CrossRef]
- Verma, S.; Nakaoke, R.; Dohgu, S.; Banks, W.A. Release of cytokines by brain endothelial cells: A polarized response to lipopolysaccharide. Brain Behav. Immun. 2006, 20, 449–455. [Google Scholar] [CrossRef]
- Banks, W.A.; Robinson, S.M. Minimal penetration of lipopolysaccharide across the murine blood-brain barrier. Brain Behav. Immun. 2010, 24, 102–109. [Google Scholar] [CrossRef] [Green Version]
- Xaio, H.; Banks, W.A.; Niehoff, M.L.; Morley, J.E. Effect of LPS on the permeability of the blood-brain barrier to insulin. Brain Res. 2001, 896, 36–42. [Google Scholar] [CrossRef]
- Banks, W.A.; Gray, A.M.; Erickson, M.A.; Salameh, T.S.; Damodarasamy, M.; Sheibani, N.; Meabon, J.S.; Wing, E.E.; Morofuji, Y.; Cook, D.G.; et al. Lipopolysaccharide-induced blood-brain barrier disruption: Roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit. J. Neuroinflamm. 2015, 12, 223. [Google Scholar] [CrossRef] [Green Version]
- Hoover, D.B. Cholinergic modulation of the immune system presents new approaches for treating inflammation. Pharmacol. Ther. 2017, 179, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Ano, Y.; Ohya, R.; Yamazaki, T.; Takahashi, C.; Taniguchi, Y.; Kondo, K.; Takashima, A.; Uchida, K.; Nakayama, H. Hop bitter acids containing a beta-carbonyl moiety prevent inflammation-induced cognitive decline via the vagus nerve and noradrenergic system. Sci. Rep. 2020, 10, 20028. [Google Scholar] [CrossRef]
- Huffman, W.J.; Subramaniyan, S.; Rodriguiz, R.M.; Wetsel, W.C.; Grill, W.M.; Terrando, N. Modulation of neuroinflammation and memory dysfunction using percutaneous vagus nerve stimulation in mice. Brain Stimul. 2019, 12, 19–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaczmarczyk, R.; Tejera, D.; Simon, B.J.; Heneka, M.T. Microglia modulation through external vagus nerve stimulation in a murine model of Alzheimer’s disease. J. Neurochem. 2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frasch, M.G.; Szynkaruk, M.; Prout, A.P.; Nygard, K.; Cao, M.; Veldhuizen, R.; Hammond, R.; Richardson, B.S. Decreased neuroinflammation correlates to higher vagus nerve activity fluctuations in near-term ovine fetuses: A case for the afferent cholinergic anti-inflammatory pathway? J. Neuroinflamm. 2016, 13, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sousa, C.; Golebiewska, A.; Poovathingal, S.K.; Kaoma, T.; Pires-Afonso, Y.; Martina, S.; Coowar, D.; Azuaje, F.; Skupin, A.; Balling, R.; et al. Single-cell transcriptomics reveals distinct inflammation-induced microglia signatures. EMBO Rep. 2018, 19, e46171. [Google Scholar] [CrossRef] [PubMed]
- Bodea, L.G.; Wang, Y.; Linnartz-Gerlach, B.; Kopatz, J.; Sinkkonen, L.; Musgrove, R.; Kaoma, T.; Muller, A.; Vallar, L.; Di Monte, D.A.; et al. Neurodegeneration by activation of the microglial complement-phagosome pathway. J. Neurosci. 2014, 34, 8546–8556. [Google Scholar] [CrossRef] [Green Version]
- Forsberg, A.; Cervenka, S.; Jonsson Fagerlund, M.; Rasmussen, L.S.; Zetterberg, H.; Erlandsson Harris, H.; Stridh, P.; Christensson, E.; Granstrom, A.; Schening, A.; et al. The immune response of the human brain to abdominal surgery. Ann. Neurol. 2017, 81, 572–582. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Liu, T.; Chen, X.; Li, L.; Feng, M.; Zhang, Y.; Wan, L.; Zhang, C.; Yao, W. Microglia induce the transformation of A1/A2 reactive astrocytes via the CXCR7/PI3K/Akt pathway in chronic post-surgical pain. J. Neuroinflamm. 2020, 17, 211. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.N.; Sha, H.H.; Wang, Y.W.; Zhou, Q.; Bhuiyan, P.; Li, N.N.; Qian, Y.N.; Dong, H.Q. Histamine 2/3 receptor agonists alleviate perioperative neurocognitive disorders by inhibiting microglia activation through the PI3K/AKT/FoxO1 pathway in aged rats. J. Neuroinflamm. 2020, 17, 217. [Google Scholar] [CrossRef]
- Raffaele, S.; Lombardi, M.; Verderio, C.; Fumagalli, M. TNF production and release from microglia via extracellular vesicles: Impact on brain functions. Cells 2020, 9, 2145. [Google Scholar] [CrossRef]
- Rizzo, F.R.; Musella, A.; de Vito, F.; Fresegna, D.; Bullitta, S.; Vanni, V.; Guadalupi, L.; Stampanoni Bassi, M.; Buttari, F.; Mandolesi, G.; et al. Tumor necrosis factor and interleukin-1beta modulate synaptic plasticity during neuroinflammation. Neural Plast. 2018, 2018, 8430123. [Google Scholar] [CrossRef] [Green Version]
- Nelson, T.E.; Olde Engberink, A.; Hernandez, R.; Puro, A.; Huitron-Resendiz, S.; Hao, C.; de Graan, P.N.; Gruol, D.L. Altered synaptic transmission in the hippocampus of transgenic mice with enhanced central nervous systems expression of interleukin-6. Brain Behav. Immun. 2012, 26, 959–971. [Google Scholar] [CrossRef] [Green Version]
- Riazi, K.; Galic, M.A.; Kuzmiski, J.B.; Ho, W.; Sharkey, K.A.; Pittman, Q.J. Microglial activation and TNFalpha production mediate altered CNS excitability following peripheral inflammation. Proc. Natl. Acad. Sci. USA 2008, 105, 17151–17156. [Google Scholar] [CrossRef] [Green Version]
- Riazi, K.; Galic, M.A.; Kentner, A.C.; Reid, A.Y.; Sharkey, K.A.; Pittman, Q.J. Microglia-dependent alteration of glutamatergic synaptic transmission and plasticity in the hippocampus during peripheral inflammation. J. Neurosci. 2015, 35, 4942–4952. [Google Scholar] [CrossRef] [Green Version]
- Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferrari, C.C.; Tarelli, R. Parkinson’s disease and systemic inflammation. Parkinsons Dis. 2011, 2011, 436813. [Google Scholar] [CrossRef] [Green Version]
- Culibrk, R.A.; Hahn, M.S. The role of chronic inflammatory bone and joint disorders in the pathogenesis and progression of Alzheimer’s disease. Front. Aging Neurosci. 2020, 12, 583884. [Google Scholar] [CrossRef] [PubMed]
- Diniz Pereira, J.; Gomes Fraga, V.; Morais Santos, A.L.; Carvalho, M.D.G.; Caramelli, P.; Braga Gomes, K. Alzheimer’s disease and type 2 diabetes mellitus: A systematic review of proteomic studies. J. Neurochem. 2021, 156, 753–776. [Google Scholar] [CrossRef]
- Hayden, M.R. Type 2 diabetes mellitus increases the risk of late-onset Alzheimer’s disease: Ultrastructural remodeling of the neurovascular unit and diabetic gliopathy. Brain Sci. 2019, 9, 262. [Google Scholar] [CrossRef] [Green Version]
- Kempuraj, D.; Thangavel, R.; Selvakumar, G.P.; Zaheer, S.; Ahmed, M.E.; Raikwar, S.P.; Zahoor, H.; Saeed, D.; Natteru, P.A.; Iyer, S.; et al. Brain and peripheral atypical inflammatory mediators potentiate neuroinflammation and neurodegeneration. Front. Cell Neurosci. 2017, 11, 216. [Google Scholar] [CrossRef]
- Walker, K.A.; Ficek, B.N.; Westbrook, R. Understanding the role of systemic inflammation in Alzheimer’s disease. ACS Chem. Neurosci. 2019, 10, 3340–3342. [Google Scholar] [CrossRef] [Green Version]
- Park, J.C.; Han, S.H.; Mook-Jung, I. Peripheral inflammatory biomarkers in Alzheimer’s disease: A brief review. BMB Rep. 2020, 53, 10–19. [Google Scholar] [CrossRef]
- Walker, K.A.; Hoogeveen, R.C.; Folsom, A.R.; Ballantyne, C.M.; Knopman, D.S.; Windham, B.G.; Jack, C.R., Jr.; Gottesman, R.F. Midlife systemic inflammatory markers are associated with late-life brain volume: The ARIC study. Neurology 2017, 89, 2262–2270. [Google Scholar] [CrossRef]
- Walker, K.A.; Windham, B.G.; Power, M.C.; Hoogeveen, R.C.; Folsom, A.R.; Ballantyne, C.M.; Knopman, D.S.; Selvin, E.; Jack, C.R., Jr.; Gottesman, R.F. The association of mid-to late-life systemic inflammation with white matter structure in older adults: The atherosclerosis risk in communities study. Neurobiol. Aging 2018, 68, 26–33. [Google Scholar] [CrossRef]
- Hoeijmakers, L.; Heinen, Y.; van Dam, A.M.; Lucassen, P.J.; Korosi, A. Microglial priming and Alzheimer’s disease: A possible role for (early) immune challenges and epigenetics? Front. Hum. Neurosci. 2016, 10, 398. [Google Scholar] [CrossRef] [Green Version]
- Dilger, R.N.; Johnson, R.W. Aging, microglial cell priming, and the discordant central inflammatory response to signals from the peripheral immune system. J. Leuk. Biol. 2008, 84, 932–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sparkman, N.L.; Johnson, R.W. Neuroinflammation associated with aging sensitizes the brain to the effects of infection or stress. Neuroimmunomodulation 2008, 15, 323–330. [Google Scholar] [CrossRef] [Green Version]
- Barrientos, R.M.; Higgins, E.A.; Biedenkapp, J.C.; Sprunger, D.B.; Wright-Hardesty, K.J.; Watkins, L.R.; Rudy, J.W.; Maier, S.F. Peripheral infection and aging interact to impair hippocampal memory consolidation. Neurobiol. Aging 2006, 27, 723–732. [Google Scholar] [CrossRef]
- Godbout, J.P.; Moreau, M.; Lestage, J.; Chen, J.; Sparkman, N.L.; O’Connor, J.; Castanon, N.; Kelley, K.W.; Dantzer, R.; Johnson, R.W. Aging exacerbates depressive-like behavior in mice in response to activation of the peripheral innate immune system. Neuropsychopharmacology 2008, 33, 2341–2351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Godbout, J.P.; Chen, J.; Abraham, J.; Richwine, A.F.; Berg, B.M.; Kelley, K.W.; Johnson, R.W. Exaggerated neuroinflammation and sickness behavior in aged mice following activation of the peripheral innate immune system. FASEB J. 2005, 19, 1329–1331. [Google Scholar] [CrossRef] [PubMed]
- Henry, C.J.; Huang, Y.; Wynne, A.M.; Godbout, J.P. Peripheral lipopolysaccharide (LPS) challenge promotes microglial hyperactivity in aged mice that is associated with exaggerated induction of both pro-inflammatory IL-1beta and anti-inflammatory IL-10 cytokines. Brain Behav. Immun. 2009, 23, 309–317. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Buchanan, J.B.; Sparkman, N.L.; Godbout, J.P.; Freund, G.G.; Johnson, R.W. Neuroinflammation and disruption in working memory in aged mice after acute stimulation of the peripheral innate immune system. Brain Behav. Immun. 2008, 22, 301–311. [Google Scholar] [CrossRef] [Green Version]
- Schrepf, A.; Kaplan, C.M.; Ichesco, E.; Larkin, T.; Harte, S.E.; Harris, R.E.; Murray, A.D.; Waiter, G.D.; Clauw, D.J.; Basu, N. A multi-modal MRI study of the central response to inflammation in rheumatoid arthritis. Nat. Commun. 2018, 9, 2243. [Google Scholar] [CrossRef]
- Kyrkanides, S.; Tallents, R.H.; Miller, J.N.; Olschowka, M.E.; Johnson, R.; Yang, M.; Olschowka, J.A.; Brouxhon, S.M.; O’Banion, M.K. Osteoarthritis accelerates and exacerbates Alzheimer’s disease pathology in mice. J. Neuroinflamm. 2011, 8, 112. [Google Scholar] [CrossRef] [Green Version]
- Suss, P.; Rothe, T.; Hoffmann, A.; Schlachetzki, J.C.M.; Winkler, J. The joint-brain axis: Insights from rheumatoid arthritis on the crosstalk between chronic peripheral inflammation and the brain. Front. Immunol. 2020, 11, 612104. [Google Scholar] [CrossRef]
- Hauser, W.; Janke, K.H.; Klump, B.; Hinz, A. Anxiety and depression in patients with inflammatory bowel disease: Comparisons with chronic liver disease patients and the general population. Inflamm. Bowel Dis. 2011, 17, 621–632. [Google Scholar] [CrossRef]
- D’Mello, C.; Swain, M.G. Liver-brain interactions in inflammatory liver diseases: Implications for fatigue and mood disorders. Brain Behav. Immun. 2014, 35, 9–20. [Google Scholar] [CrossRef]
- Lasselin, J.; Laye, S.; Dexpert, S.; Aubert, A.; Gonzalez, C.; Gin, H.; Capuron, L. Fatigue symptoms relate to systemic inflammation in patients with type 2 diabetes. Brain Behav. Immun. 2012, 26, 1211–1219. [Google Scholar] [CrossRef]
- Saltiel, A.R.; Olefsky, J.M. Inflammatory mechanisms linking obesity and metabolic disease. J. Clin. Investig. 2017, 127, 1–4. [Google Scholar] [CrossRef]
- Li, C.; Xu, M.M.; Wang, K.; Adler, A.J.; Vella, A.T.; Zhou, B. Macrophage polarization and meta-inflammation. Transl. Res. 2018, 191, 29–44. [Google Scholar] [CrossRef]
- Alexaki, V.I.; Chavakis, T. The role of innate immunity in the regulation of brown and beige adipogenesis. Rev. Endocrinol. Metab. Disord. 2016, 17, 41–49. [Google Scholar] [CrossRef]
- Chmelar, J.; Chung, K.J.; Chavakis, T. The role of innate immune cells in obese adipose tissue inflammation and development of insulin resistance. Thromb. Haemost. 2013, 109, 399–406. [Google Scholar]
- Brestoff, J.R.; Artis, D. Immune regulation of metabolic homeostasis in health and disease. Cell 2015, 161, 146–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cancello, R.; Tordjman, J.; Poitou, C.; Guilhem, G.; Bouillot, J.L.; Hugol, D.; Coussieu, C.; Basdevant, A.; Bar Hen, A.; Bedossa, P.; et al. Increased infiltration of macrophages in omental adipose tissue is associated with marked hepatic lesions in morbid human obesity. Diabetes 2006, 55, 1554–1561. [Google Scholar] [CrossRef] [Green Version]
- Harman-Boehm, I.; Bluher, M.; Redel, H.; Sion-Vardy, N.; Ovadia, S.; Avinoach, E.; Shai, I.; Kloting, N.; Stumvoll, M.; Bashan, N.; et al. Macrophage infiltration into omental versus subcutaneous fat across different populations: Effect of regional adiposity and the comorbidities of obesity. J. Clin. Endocrinol. Metab. 2007, 92, 2240–2247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stefan, N.; Haring, H.U.; Hu, F.B.; Schulze, M.B. Metabolically healthy obesity: Epidemiology, mechanisms, and clinical implications. Lancet Diabetes Endocrinol. 2013, 1, 152–162. [Google Scholar] [CrossRef]
- Ghaben, A.L.; Scherer, P.E. Adipogenesis and metabolic health. Nat. Rev. Mol. Cell Biol. 2019, 20, 242–258. [Google Scholar] [CrossRef] [PubMed]
- Spalding, K.L.; Arner, E.; Westermark, P.O.; Bernard, S.; Buchholz, B.A.; Bergmann, O.; Blomqvist, L.; Hoffstedt, J.; Naslund, E.; Britton, T.; et al. Dynamics of fat cell turnover in humans. Nature 2008, 453, 783–787. [Google Scholar] [CrossRef]
- Fasshauer, M.; Bluher, M. Adipokines in health and disease. Trends Pharmacol. Sci. 2015, 36, 461–470. [Google Scholar] [CrossRef]
- Mancuso, P. The role of adipokines in chronic inflammation. Immunotargets Ther. 2016, 5, 47–56. [Google Scholar] [CrossRef] [Green Version]
- Pan, W.W.; Myers, M.G., Jr. Leptin and the maintenance of elevated body weight. Nat. Rev. Neurosci. 2018, 19, 95–105. [Google Scholar] [CrossRef]
- Chung, K.J.; Nati, M.; Chavakis, T.; Chatzigeorgiou, A. Innate immune cells in the adipose tissue. Rev. Endocrinol. Metab. Disord. 2018, 19, 283–292. [Google Scholar] [CrossRef] [PubMed]
- Hill, A.A.; Reid Bolus, W.; Hasty, A.H. A decade of progress in adipose tissue macrophage biology. Immunol. Rev. 2014, 262, 134–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chung, K.J.; Chatzigeorgiou, A.; Economopoulou, M.; Garcia-Martin, R.; Alexaki, V.I.; Mitroulis, I.; Nati, M.; Gebler, J.; Ziemssen, T.; Goelz, S.E.; et al. A self-sustained loop of inflammation-driven inhibition of beige adipogenesis in obesity. Nat. Immunol. 2017, 18, 654–664. [Google Scholar] [CrossRef] [PubMed]
- Lumeng, C.N.; Bodzin, J.L.; Saltiel, A.R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Investig. 2007, 117, 175–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chatzigeorgiou, A.; Karalis, K.P.; Bornstein, S.R.; Chavakis, T. Lymphocytes in obesity-related adipose tissue inflammation. Diabetologia 2012, 55, 2583–2592. [Google Scholar] [CrossRef]
- Wu, H.; Ballantyne, C.M. Skeletal muscle inflammation and insulin resistance in obesity. J. Clin. Investig. 2017, 127, 43–54. [Google Scholar] [CrossRef]
- Czech, M.P. Insulin action and resistance in obesity and type 2 diabetes. Nat. Med. 2017, 23, 804–814. [Google Scholar] [CrossRef]
- Samuel, V.T.; Shulman, G.I. The pathogenesis of insulin resistance: Integrating signaling pathways and substrate flux. J. Clin. Investig. 2016, 126, 12–22. [Google Scholar] [CrossRef] [Green Version]
- Sabio, G.; Das, M.; Mora, A.; Zhang, Z.; Jun, J.Y.; Ko, H.J.; Barrett, T.; Kim, J.K.; Davis, R.J. A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Science 2008, 322, 1539–1543. [Google Scholar] [CrossRef] [Green Version]
- Hirosumi, J.; Tuncman, G.; Chang, L.; Gorgun, C.Z.; Uysal, K.T.; Maeda, K.; Karin, M.; Hotamisligil, G.S. A central role for JNK in obesity and insulin resistance. Nature 2002, 420, 333–336. [Google Scholar] [CrossRef]
- Uysal, K.T.; Wiesbrock, S.M.; Marino, M.W.; Hotamisligil, G.S. Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature 1997, 389, 610–614. [Google Scholar] [CrossRef]
- Xu, H.; Barnes, G.T.; Yang, Q.; Tan, G.; Yang, D.; Chou, C.J.; Sole, J.; Nichols, A.; Ross, J.S.; Tartaglia, L.A.; et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Investig. 2003, 112, 1821–1830. [Google Scholar] [CrossRef]
- McLaughlin, T.; Ackerman, S.E.; Shen, L.; Engleman, E. Role of innate and adaptive immunity in obesity-associated metabolic disease. J. Clin. Investig. 2017, 127, 5–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De la Monte, S.M.; Longato, L.; Tong, M.; Wands, J.R. Insulin resistance and neurodegeneration: Roles of obesity, type 2 diabetes mellitus and non-alcoholic steatohepatitis. Curr. Opin. Investig. Drugs 2009, 10, 1049–1060. [Google Scholar]
- Procaccini, C.; Santopaolo, M.; Faicchia, D.; Colamatteo, A.; Formisano, L.; de Candia, P.; Galgani, M.; de Rosa, V.; Matarese, G. Role of metabolism in neurodegenerative disorders. Metabolism 2016, 65, 1376–1390. [Google Scholar] [CrossRef]
- Balasubramanian, P.; Kiss, T.; Tarantini, S.; Nyul-Toth, A.; Ahire, C.; Yabluchanskiy, A.; Csipo, T.; Lipecz, A.; Tabak, A.; Institoris, A.; et al. Obesity-induced cognitive impairment in older adults: A microvascular perspective. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H740–H761. [Google Scholar] [CrossRef] [PubMed]
- Milaneschi, Y.; Simmons, W.K.; van Rossum, E.F.C.; Penninx, B.W. Depression and obesity: Evidence of shared biological mechanisms. Mol. Psychiatry 2019, 24, 18–33. [Google Scholar] [CrossRef]
- Anstey, K.J.; Cherbuin, N.; Budge, M.; Young, J. Body mass index in midlife and late-life as a risk factor for dementia: A meta-analysis of prospective studies. Obes. Rev. 2011, 12, e426–e437. [Google Scholar] [CrossRef]
- Gudala, K.; Bansal, D.; Schifano, F.; Bhansali, A. Diabetes mellitus and risk of dementia: A meta-analysis of prospective observational studies. J. Diabetes Investig. 2013, 4, 640–650. [Google Scholar] [CrossRef] [Green Version]
- Kanaya, A.M.; Lindquist, K.; Harris, T.B.; Launer, L.; Rosano, C.; Satterfield, S.; Yaffe, K.; Health, A.B.C.S. Total and regional adiposity and cognitive change in older adults: The Health, Aging and Body Composition (ABC) study. Arch. Neurol. 2009, 66, 329–335. [Google Scholar] [CrossRef]
- Gunstad, J.; Lhotsky, A.; Wendell, C.R.; Ferrucci, L.; Zonderman, A.B. Longitudinal examination of obesity and cognitive function: Results from the Baltimore longitudinal study of aging. Neuroepidemiology 2010, 34, 222–229. [Google Scholar] [CrossRef] [Green Version]
- Benito-Leon, J.; Mitchell, A.J.; Hernandez-Gallego, J.; Bermejo-Pareja, F. Obesity and impaired cognitive functioning in the elderly: A population-based cross-sectional study (NEDICES). Eur. J. Neurol. 2013, 20, 899-e77. [Google Scholar] [CrossRef]
- Munger, K.L.; Chitnis, T.; Ascherio, A. Body size and risk of MS in two cohorts of US women. Neurology 2009, 73, 1543–1550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hedstrom, A.K.; Olsson, T.; Alfredsson, L. High body mass index before age 20 is associated with increased risk for multiple sclerosis in both men and women. Mult. Scler. 2012, 18, 1334–1336. [Google Scholar] [CrossRef] [PubMed]
- Langer-Gould, A.; Brara, S.M.; Beaber, B.E.; Koebnick, C. Childhood obesity and risk of pediatric multiple sclerosis and clinically isolated syndrome. Neurology 2013, 80, 548–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mokry, L.E.; Ross, S.; Timpson, N.J.; Sawcer, S.; Davey Smith, G.; Richards, J.B. Obesity and multiple sclerosis: A mendelian randomization study. PLoS Med. 2016, 13, e1002053. [Google Scholar] [CrossRef] [Green Version]
- Schachter, J.; Martel, J.; Lin, C.S.; Chang, C.J.; Wu, T.R.; Lu, C.C.; Ko, Y.F.; Lai, H.C.; Ojcius, D.M.; Young, J.D. Effects of obesity on depression: A role for inflammation and the gut microbiota. Brain Behav. Immun. 2018, 69, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Quek, Y.H.; Tam, W.W.S.; Zhang, M.W.B.; Ho, R.C.M. Exploring the association between childhood and adolescent obesity and depression: A meta-analysis. Obes. Rev. 2017, 18, 742–754. [Google Scholar] [CrossRef]
- Jokela, M.; Hamer, M.; Singh-Manoux, A.; Batty, G.D.; Kivimaki, M. Association of metabolically healthy obesity with depressive symptoms: Pooled analysis of eight studies. Mol. Psychiatry 2014, 19, 910–914. [Google Scholar] [CrossRef]
- Kullmann, S.; Schweizer, F.; Veit, R.; Fritsche, A.; Preissl, H. Compromised white matter integrity in obesity. Obes. Rev. 2015, 16, 273–281. [Google Scholar] [CrossRef]
- Kullmann, S.; Heni, M.; Hallschmid, M.; Fritsche, A.; Preissl, H.; Haring, H.U. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol. Rev. 2016, 96, 1169–1209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ward, M.A.; Carlsson, C.M.; Trivedi, M.A.; Sager, M.A.; Johnson, S.C. The effect of body mass index on global brain volume in middle-aged adults: A cross sectional study. BMC Neurol. 2005, 5, 23. [Google Scholar] [CrossRef] [Green Version]
- Cazettes, F.; Cohen, J.I.; Yau, P.L.; Talbot, H.; Convit, A. Obesity-mediated inflammation may damage the brain circuit that regulates food intake. Brain Res. 2011, 1373, 101–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samara, A.; Murphy, T.; Strain, J.; Rutlin, J.; Sun, P.; Neyman, O.; Sreevalsan, N.; Shimony, J.S.; Ances, B.M.; Song, S.K.; et al. Neuroinflammation and white matter alterations in obesity assessed by diffusion basis spectrum imaging. Front. Hum. Neurosci. 2019, 13, 464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Strooper, B.; Karran, E. The cellular phase of Alzheimer’s disease. Cell 2016, 164, 603–615. [Google Scholar] [CrossRef] [Green Version]
- Bartels, T.; de Schepper, S.; Hong, S. Microglia modulate neurodegeneration in Alzheimer’s and Parkinson’s diseases. Science 2020, 370, 66–69. [Google Scholar] [CrossRef] [PubMed]
- Voet, S.; Prinz, M.; van Loo, G. Microglia in central nervous system inflammation and multiple sclerosis pathology. Trends Mol. Med. 2019, 25, 112–123. [Google Scholar] [CrossRef]
- Troubat, R.; Barone, P.; Leman, S.; Desmidt, T.; Cressant, A.; Atanasova, B.; Brizard, B.; El Hage, W.; Surget, A.; Belzung, C.; et al. Neuroinflammation and depression: A review. Eur. J. Neurosci. 2021, 53, 151–171. [Google Scholar] [CrossRef]
- Frohnert, B.I.; Jacobs, D.R., Jr.; Steinberger, J.; Moran, A.; Steffen, L.M.; Sinaiko, A.R. Relation between serum free fatty acids and adiposity, insulin resistance, and cardiovascular risk factors from adolescence to adulthood. Diabetes 2013, 62, 3163–3169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levy, M.; Kolodziejczyk, A.A.; Thaiss, C.A.; Elinav, E. Dysbiosis and the immune system. Nat. Rev. Immunol. 2017, 17, 219–232. [Google Scholar] [CrossRef]
- Erny, D.; Hrabe de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
- Swierczynska, M.M.; Mateska, I.; Peitzsch, M.; Bornstein, S.R.; Chavakis, T.; Eisenhofer, G.; Lamounier-Zepter, V.; Eaton, S. Changes in morphology and function of adrenal cortex in mice fed a high-fat diet. Int. J. Obes. 2015, 39, 321–330. [Google Scholar] [CrossRef] [PubMed]
- Sorrells, S.F.; Caso, J.R.; Munhoz, C.D.; Sapolsky, R.M. The stressed CNS: When glucocorticoids aggravate inflammation. Neuron 2009, 64, 33–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sorrells, S.F.; Munhoz, C.D.; Manley, N.C.; Yen, S.; Sapolsky, R.M. Glucocorticoids increase excitotoxic injury and inflammation in the hippocampus of adult male rats. Neuroendocrinology 2014, 100, 129–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hao, S.; Dey, A.; Yu, X.; Stranahan, A.M. Dietary obesity reversibly induces synaptic stripping by microglia and impairs hippocampal plasticity. Brain Behav. Immun. 2016, 51, 230–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cope, E.C.; LaMarca, E.A.; Monari, P.K.; Olson, L.B.; Martinez, S.; Zych, A.D.; Katchur, N.J.; Gould, E. Microglia play an active role in obesity-associated cognitive decline. J. Neurosci. 2018, 38, 8889–8904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Butler, M.J.; Cole, R.M.; Deems, N.P.; Belury, M.A.; Barrientos, R.M. Fatty food, fatty acids, and microglial priming in the adult and aged hippocampus and amygdala. Brain Behav. Immun. 2020, 89, 145–158. [Google Scholar] [CrossRef]
- Kawamura, N.; Katsuura, G.; Yamada-Goto, N.; Novianti, E.; Inui, A.; Asakawa, A. Impaired brain fractalkine-CX3CR1 signaling is implicated in cognitive dysfunction in diet-induced obese mice. BMJ Open Diabetes Res. Care 2021, 9, 1. [Google Scholar] [CrossRef]
- Cai, M.; Wang, H.; Li, J.J.; Zhang, Y.L.; Xin, L.; Li, F.; Lou, S.J. The signaling mechanisms of hippocampal endoplasmic reticulum stress affecting neuronal plasticity-related protein levels in high fat diet-induced obese rats and the regulation of aerobic exercise. Brain Behav. Immun. 2016, 57, 347–359. [Google Scholar] [CrossRef] [PubMed]
- Nakandakari, S.; Munoz, V.R.; Kuga, G.K.; Gaspar, R.C.; Sant’Ana, M.R.; Pavan, I.C.B.; da Silva, L.G.S.; Morelli, A.P.; Simabuco, F.M.; da Silva, A.S.R.; et al. Short-term high-fat diet modulates several inflammatory, ER stress, and apoptosis markers in the hippocampus of young mice. Brain Behav. Immun. 2019, 79, 284–293. [Google Scholar] [CrossRef]
- Scheiblich, H.; Schlutter, A.; Golenbock, D.T.; Latz, E.; Martinez-Martinez, P.; Heneka, M.T. Activation of the NLRP3 inflammasome in microglia: The role of ceramide. J. Neurochem. 2017, 143, 534–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feng, X.; Zhao, Y.; Yang, T.; Song, M.; Wang, C.; Yao, Y.; Fan, H. Glucocorticoid-driven NLRP3 inflammasome activation in hippocampal microglia mediates chronic stress-induced depressive-like behaviors. Front. Mol. Neurosci. 2019, 12, 210. [Google Scholar] [CrossRef]
- Jeon, B.T.; Jeong, E.A.; Shin, H.J.; Lee, Y.; Lee, D.H.; Kim, H.J.; Kang, S.S.; Cho, G.J.; Choi, W.S.; Roh, G.S. Resveratrol attenuates obesity-associated peripheral and central inflammation and improves memory deficit in mice fed a high-fat diet. Diabetes 2012, 61, 1444–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sims-Robinson, C.; Bakeman, A.; Glasser, R.; Boggs, J.; Pacut, C.; Feldman, E.L. The role of endoplasmic reticulum stress in hippocampal insulin resistance. Exp. Neurol. 2016, 277, 261–267. [Google Scholar] [CrossRef] [Green Version]
- Stranahan, A.M.; Norman, E.D.; Lee, K.; Cutler, R.G.; Telljohann, R.S.; Egan, J.M.; Mattson, M.P. Diet-induced insulin resistance impairs hippocampal synaptic plasticity and cognition in middle-aged rats. Hippocampus 2008, 18, 1085–1088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kroner, A.; Greenhalgh, A.D.; Zarruk, J.G.; Passos Dos Santos, R.; Gaestel, M.; David, S. TNF and increased intracellular iron alter macrophage polarization to a detrimental M1 phenotype in the injured spinal cord. Neuron 2014, 83, 1098–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Siddiqui, T.A.; Lively, S.; Schlichter, L.C. Complex molecular and functional outcomes of single versus sequential cytokine stimulation of rat microglia. J. Neuroinflamm. 2016, 13, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, Y.; Vidal-Itriago, A.; Milanova, I.; Korpel, N.L.; Kalsbeek, M.J.; Tom, R.Z.; Kalsbeek, A.; Hofmann, S.M.; Yi, C.X. Deficiency of leptin receptor in myeloid cells disrupts hypothalamic metabolic circuits and causes body weight increase. Mol. Metab. 2018, 7, 155–160. [Google Scholar] [CrossRef] [PubMed]
- Tang, C.H.; Lu, D.Y.; Yang, R.S.; Tsai, H.Y.; Kao, M.C.; Fu, W.M.; Chen, Y.F. Leptin-induced IL-6 production is mediated by leptin receptor, insulin receptor substrate-1, phosphatidylinositol 3-kinase, Akt, NF-kappaB, and p300 pathway in microglia. J. Immunol. 2007, 179, 1292–1302. [Google Scholar] [CrossRef] [Green Version]
- Lafrance, V.; Inoue, W.; Kan, B.; Luheshi, G.N. Leptin modulates cell morphology and cytokine release in microglia. Brain Behav. Immun. 2010, 24, 358–365. [Google Scholar] [CrossRef] [PubMed]
- Fernandez-Martos, C.M.; Gonzalez, P.; Rodriguez, F.J. Acute leptin treatment enhances functional recovery after spinal cord injury. PLoS ONE 2012, 7, e35594. [Google Scholar] [CrossRef]
- Vinuesa, A.; Bentivegna, M.; Calfa, G.; Filipello, F.; Pomilio, C.; Bonaventura, M.M.; Lux-Lantos, V.; Matzkin, M.E.; Gregosa, A.; Presa, J.; et al. Early exposure to a high-fat diet impacts on hippocampal plasticity: Implication of microglia-derived exosome-like extracellular vesicles. Mol. Neurobiol. 2019, 56, 5075–5094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Milanski, M.; Degasperi, G.; Coope, A.; Morari, J.; Denis, R.; Cintra, D.E.; Tsukumo, D.M.; Anhe, G.; Amaral, M.E.; Takahashi, H.K.; et al. Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus: Implications for the pathogenesis of obesity. J. Neurosci. 2009, 29, 359–370. [Google Scholar] [CrossRef]
- Yanguas-Casas, N.; Crespo-Castrillo, A.; de Ceballos, M.L.; Chowen, J.A.; Azcoitia, I.; Arevalo, M.A.; Garcia-Segura, L.M. Sex differences in the phagocytic and migratory activity of microglia and their impairment by palmitic acid. Glia 2018, 66, 522–537. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Liu, D.; Wang, F.; Liu, S.; Zhao, S.; Ling, E.A.; Hao, A. Saturated fatty acids activate microglia via Toll-like receptor 4/NF-kappaB signalling. Br. J. Nutr. 2012, 107, 229–241. [Google Scholar] [CrossRef] [Green Version]
- Bazinet, R.P.; Laye, S. Polyunsaturated fatty acids and their metabolites in brain function and disease. Nat. Rev. Neurosci. 2014, 15, 771–785. [Google Scholar] [CrossRef] [PubMed]
- Laye, S.; Nadjar, A.; Joffre, C.; Bazinet, R.P. Anti-inflammatory effects of omega-3 fatty acids in the brain: Physiological mechanisms and relevance to pharmacology. Pharmacol. Rev. 2018, 70, 12–38. [Google Scholar] [CrossRef]
- Oksman, M.; Iivonen, H.; Hogyes, E.; Amtul, Z.; Penke, B.; Leenders, I.; Broersen, L.; Lutjohann, D.; Hartmann, T.; Tanila, H. Impact of different saturated fatty acid, polyunsaturated fatty acid and cholesterol containing diets on beta-amyloid accumulation in APP/PS1 transgenic mice. Neurobiol. Dis. 2006, 23, 563–572. [Google Scholar] [CrossRef] [PubMed]
- Ma, Q.L.; Yang, F.; Rosario, E.R.; Ubeda, O.J.; Beech, W.; Gant, D.J.; Chen, P.P.; Hudspeth, B.; Chen, C.; Zhao, Y.; et al. Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: Suppression by omega-3 fatty acids and curcumin. J. Neurosci. 2009, 29, 9078–9089. [Google Scholar] [CrossRef]
- De Smedt-Peyrusse, V.; Sargueil, F.; Moranis, A.; Harizi, H.; Mongrand, S.; Laye, S. Docosahexaenoic acid prevents lipopolysaccharide-induced cytokine production in microglial cells by inhibiting lipopolysaccharide receptor presentation but not its membrane subdomain localization. J. Neurochem. 2008, 105, 296–307. [Google Scholar] [CrossRef] [PubMed]
- Antonietta Ajmone-Cat, M.; Lavinia Salvatori, M.; de Simone, R.; Mancini, M.; Biagioni, S.; Bernardo, A.; Cacci, E.; Minghetti, L. Docosahexaenoic acid modulates inflammatory and antineurogenic functions of activated microglial cells. J. Neurosci. Res. 2012, 90, 575–587. [Google Scholar] [CrossRef]
- Mulders, R.J.; de Git, K.C.G.; Schele, E.; Dickson, S.L.; Sanz, Y.; Adan, R.A.H. Microbiota in obesity: Interactions with enteroendocrine, immune and central nervous systems. Obes. Rev. 2018, 19, 435–451. [Google Scholar] [CrossRef]
- Soto, M.; Herzog, C.; Pacheco, J.A.; Fujisaka, S.; Bullock, K.; Clish, C.B.; Kahn, C.R. Gut microbiota modulate neurobehavior through changes in brain insulin sensitivity and metabolism. Mol. Psychiatry 2018, 23, 2287–2301. [Google Scholar] [CrossRef] [PubMed]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell 2016, 167, 1469–1480.e12. [Google Scholar] [CrossRef] [Green Version]
- Witt, A.; Mirtschink, P.; Palladini, A.; Mateska, I.; Abdelmegeed, H.; Grzybek, M.; Wielockx, B.; Peitzsch, M.; Coskun, Ü.; Chavakis, T.; et al. Obesity-associated lipidomic remodeling of the adrenal gland indicates an important role of the FADS2-arachidonic acid axis in adrenocortical hormone production. bioRxiv 2020. [Google Scholar] [CrossRef]
- Stranahan, A.M.; Arumugam, T.V.; Cutler, R.G.; Lee, K.; Egan, J.M.; Mattson, M.P. Diabetes impairs hippocampal function through glucocorticoid-mediated effects on new and mature neurons. Nat. Neurosci. 2008, 11, 309–317. [Google Scholar] [CrossRef] [Green Version]
- Hofmann, A.; Peitzsch, M.; Brunssen, C.; Mittag, J.; Jannasch, A.; Frenzel, A.; Brown, N.; Weldon, S.M.; Eisenhofer, G.; Bornstein, S.R.; et al. Elevated steroid hormone production in the db/db mouse model of obesity and type 2 diabetes. Horm. Metab. Res. 2017, 49, 43–49. [Google Scholar] [CrossRef]
- Sobesky, J.L.; D’Angelo, H.M.; Weber, M.D.; Anderson, N.D.; Frank, M.G.; Watkins, L.R.; Maier, S.F.; Barrientos, R.M. Glucocorticoids mediate short-term high-fat diet induction of neuroinflammatory priming, the NLRP3 inflammasome, and the danger signal HMGB1. eNeuro 2016, 3. [Google Scholar] [CrossRef] [Green Version]
- Van Olst, L.; Bielefeld, P.; Fitzsimons, C.P.; de Vries, H.E.; Schouten, M. Glucocorticoid-mediated modulation of morphological changes associated with aging in microglia. Aging Cell 2018, 17, e12790. [Google Scholar] [CrossRef] [PubMed]
- Ros-Bernal, F.; Hunot, S.; Herrero, M.T.; Parnadeau, S.; Corvol, J.C.; Lu, L.; Alvarez-Fischer, D.; Carrillo-de Sauvage, M.A.; Saurini, F.; Coussieu, C.; et al. Microglial glucocorticoid receptors play a pivotal role in regulating dopaminergic neurodegeneration in parkinsonism. Proc. Natl. Acad. Sci. USA 2011, 108, 6632–6637. [Google Scholar] [CrossRef] [Green Version]
- Maatouk, L.; Compagnion, A.C.; Sauvage, M.C.; Bemelmans, A.P.; Leclere-Turbant, S.; Cirotteau, V.; Tohme, M.; Beke, A.; Trichet, M.; Bazin, V.; et al. TLR9 activation via microglial glucocorticoid receptors contributes to degeneration of midbrain dopamine neurons. Nat. Commun. 2018, 9, 2450. [Google Scholar] [CrossRef] [PubMed]
- Valladolid-Acebes, I.; Fole, A.; Martin, M.; Morales, L.; Cano, M.V.; Ruiz-Gayo, M.; Del Olmo, N. Spatial memory impairment and changes in hippocampal morphology are triggered by high-fat diets in adolescent mice. Is there a role of leptin? Neurobiol. Learn. Mem. 2013, 106, 18–25. [Google Scholar] [CrossRef] [PubMed]
- Heyward, F.D.; Walton, R.G.; Carle, M.S.; Coleman, M.A.; Garvey, W.T.; Sweatt, J.D. Adult mice maintained on a high-fat diet exhibit object location memory deficits and reduced hippocampal SIRT1 gene expression. Neurobiol. Learn. Mem. 2012, 98, 25–32. [Google Scholar] [CrossRef] [Green Version]
- Hwang, L.L.; Wang, C.H.; Li, T.L.; Chang, S.D.; Lin, L.C.; Chen, C.P.; Chen, C.T.; Liang, K.C.; Ho, I.K.; Yang, W.S.; et al. Sex differences in high-fat diet-induced obesity, metabolic alterations and learning, and synaptic plasticity deficits in mice. Obesity 2010, 18, 463–469. [Google Scholar] [CrossRef] [PubMed]
- Woo, J.; Shin, K.O.; Park, S.Y.; Jang, K.S.; Kang, S. Effects of exercise and diet change on cognition function and synaptic plasticity in high fat diet induced obese rats. Lipids Health Dis. 2013, 12, 144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Granholm, A.C.; Bimonte-Nelson, H.A.; Moore, A.B.; Nelson, M.E.; Freeman, L.R.; Sambamurti, K. Effects of a saturated fat and high cholesterol diet on memory and hippocampal morphology in the middle-aged rat. J. Alzheimers Dis. 2008, 14, 133–145. [Google Scholar] [CrossRef] [Green Version]
- Erion, J.R.; Wosiski-Kuhn, M.; Dey, A.; Hao, S.; Davis, C.L.; Pollock, N.K.; Stranahan, A.M. Obesity elicits interleukin 1-mediated deficits in hippocampal synaptic plasticity. J. Neurosci. 2014, 34, 2618–2631. [Google Scholar] [CrossRef] [Green Version]
- Bocarsly, M.E.; Fasolino, M.; Kane, G.A.; LaMarca, E.A.; Kirschen, G.W.; Karatsoreos, I.N.; McEwen, B.S.; Gould, E. Obesity diminishes synaptic markers, alters microglial morphology, and impairs cognitive function. Proc. Natl. Acad. Sci. USA 2015, 112, 15731–15736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shinohara, M.; Tashiro, Y.; Shinohara, M.; Hirokawa, J.; Suzuki, K.; Onishi-Takeya, M.; Mukouzono, M.; Takeda, S.; Saito, T.; Fukumori, A.; et al. Increased levels of Abeta42 decrease the lifespan of ob/ob mice with dysregulation of microglia and astrocytes. FASEB J. 2020, 34, 2425–2435. [Google Scholar] [CrossRef] [Green Version]
- Sobesky, J.L.; Barrientos, R.M.; de May, H.S.; Thompson, B.M.; Weber, M.D.; Watkins, L.R.; Maier, S.F. High-fat diet consumption disrupts memory and primes elevations in hippocampal IL-1beta, an effect that can be prevented with dietary reversal or IL-1 receptor antagonism. Brain Behav. Immun. 2014, 42, 22–32. [Google Scholar] [CrossRef] [Green Version]
- Brykczynska, U.; Geigges, M.; Wiedemann, S.J.; Dror, E.; Boni-Schnetzler, M.; Hess, C.; Donath, M.Y.; Paro, R. Distinct transcriptional responses across tissue-resident macrophages to short-term and long-term metabolic challenge. Cell Rep. 2020, 30, 1627–1643.e7. [Google Scholar] [CrossRef]
- Dror, E.; Dalmas, E.; Meier, D.T.; Wueest, S.; Thevenet, J.; Thienel, C.; Timper, K.; Nordmann, T.M.; Traub, S.; Schulze, F.; et al. Postprandial macrophage-derived IL-1beta stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat. Immunol. 2017, 18, 283–292. [Google Scholar] [CrossRef] [Green Version]
- Cavaliere, G.; Trinchese, G.; Penna, E.; Cimmino, F.; Pirozzi, C.; Lama, A.; Annunziata, C.; Catapano, A.; Mattace Raso, G.; Meli, R.; et al. High-fat diet induces neuroinflammation and mitochondrial impairment in mice cerebral cortex and synaptic fraction. Front. Cell Neurosci. 2019, 13, 509. [Google Scholar] [CrossRef] [PubMed]
- Cai, M.; Hu, J.Y.; Liu, B.B.; Li, J.J.; Li, F.; Lou, S. The molecular mechanisms of excessive hippocampal endoplasmic reticulum stress depressing cognition-related proteins expression and the regulatory effects of Nrf2. Neuroscience 2020, 431, 152–165. [Google Scholar] [CrossRef] [PubMed]
- Tucsek, Z.; Toth, P.; Sosnowska, D.; Gautam, T.; Mitschelen, M.; Koller, A.; Szalai, G.; Sonntag, W.E.; Ungvari, Z.; Csiszar, A. Obesity in aging exacerbates blood-brain barrier disruption, neuroinflammation, and oxidative stress in the mouse hippocampus: Effects on expression of genes involved in beta-amyloid generation and Alzheimer’s disease. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 1212–1226. [Google Scholar] [CrossRef] [PubMed]
- Graham, L.C.; Harder, J.M.; Soto, I.; de Vries, W.N.; John, S.W.; Howell, G.R. Chronic consumption of a western diet induces robust glial activation in aging mice and in a mouse model of Alzheimer’s disease. Sci. Rep. 2016, 6, 21568. [Google Scholar] [CrossRef] [PubMed]
- Valcarcel-Ares, M.N.; Tucsek, Z.; Kiss, T.; Giles, C.B.; Tarantini, S.; Yabluchanskiy, A.; Balasubramanian, P.; Gautam, T.; Galvan, V.; Ballabh, P.; et al. Obesity in aging exacerbates neuroinflammation, dysregulating synaptic function-related genes and altering eicosanoid synthesis in the mouse hippocampus: Potential role in impaired synaptic plasticity and cognitive decline. J. Gerontol. A Biol. Sci. Med. Sci. 2019, 74, 290–298. [Google Scholar] [CrossRef] [PubMed]
- Yin, Z.; Raj, D.D.; Schaafsma, W.; van der Heijden, R.A.; Kooistra, S.M.; Reijne, A.C.; Zhang, X.; Moser, J.; Brouwer, N.; Heeringa, P.; et al. Low-fat diet with caloric restriction reduces white matter microglia activation during aging. Front. Mol. Neurosci. 2018, 11, 65. [Google Scholar] [CrossRef] [PubMed]
- Julien, C.; Tremblay, C.; Phivilay, A.; Berthiaume, L.; Emond, V.; Julien, P.; Calon, F. High-fat diet aggravates amyloid-beta and tau pathologies in the 3xTg-AD mouse model. Neurobiol. Aging 2010, 31, 1516–1531. [Google Scholar] [CrossRef] [PubMed]
- Leboucher, A.; Laurent, C.; Fernandez-Gomez, F.J.; Burnouf, S.; Troquier, L.; Eddarkaoui, S.; Demeyer, D.; Caillierez, R.; Zommer, N.; Vallez, E.; et al. Detrimental effects of diet-induced obesity on tau pathology are independent of insulin resistance in tau transgenic mice. Diabetes 2013, 62, 1681–1688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maesako, M.; Uemura, K.; Kubota, M.; Kuzuya, A.; Sasaki, K.; Hayashida, N.; Asada-Utsugi, M.; Watanabe, K.; Uemura, M.; Kihara, T.; et al. Exercise is more effective than diet control in preventing high fat diet-induced beta-amyloid deposition and memory deficit in amyloid precursor protein transgenic mice. J. Biol. Chem. 2012, 287, 23024–23033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ivanova, N.; Liu, Q.; Agca, C.; Agca, Y.; Noble, E.G.; Whitehead, S.N.; Cechetto, D.F. White matter inflammation and cognitive function in a co-morbid metabolic syndrome and prodromal Alzheimer’s disease rat model. J. Neuroinflamm. 2020, 17, 29. [Google Scholar] [CrossRef] [Green Version]
- Choi, J.Y.; Jang, E.H.; Park, C.S.; Kang, J.H. Enhanced susceptibility to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity in high-fat diet-induced obesity. Free Radic. Biol. Med. 2005, 38, 806–816. [Google Scholar] [CrossRef] [PubMed]
- Morris, J.K.; Bomhoff, G.L.; Stanford, J.A.; Geiger, P.C. Neurodegeneration in an animal model of Parkinson’s disease is exacerbated by a high-fat diet. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 299, R1082–R1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boitard, C.; Cavaroc, A.; Sauvant, J.; Aubert, A.; Castanon, N.; Laye, S.; Ferreira, G. Impairment of hippocampal-dependent memory induced by juvenile high-fat diet intake is associated with enhanced hippocampal inflammation in rats. Brain Behav. Immun. 2014, 40, 9–17. [Google Scholar] [CrossRef]
- Andre, C.; Dinel, A.L.; Ferreira, G.; Laye, S.; Castanon, N. Diet-induced obesity progressively alters cognition, anxiety-like behavior and lipopolysaccharide-induced depressive-like behavior: Focus on brain indoleamine 2,3-dioxygenase activation. Brain Behav. Immun. 2014, 41, 10–21. [Google Scholar] [CrossRef]
- Gao, Y.; Ottaway, N.; Schriever, S.C.; Legutko, B.; Garcia-Caceres, C.; de la Fuente, E.; Mergen, C.; Bour, S.; Thaler, J.P.; Seeley, R.J.; et al. Hormones and diet, but not body weight, control hypothalamic microglial activity. Glia 2014, 62, 17–25. [Google Scholar] [CrossRef] [Green Version]
- Thaler, J.P.; Yi, C.X.; Schur, E.A.; Guyenet, S.J.; Hwang, B.H.; Dietrich, M.O.; Zhao, X.; Sarruf, D.A.; Izgur, V.; Maravilla, K.R.; et al. Obesity is associated with hypothalamic injury in rodents and humans. J. Clin. Investig. 2012, 122, 153–162. [Google Scholar] [CrossRef] [Green Version]
- Baufeld, C.; Osterloh, A.; Prokop, S.; Miller, K.R.; Heppner, F.L. High-fat diet-induced brain region-specific phenotypic spectrum of CNS resident microglia. Acta Neuropathol. 2016, 132, 361–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.; Zhang, G.; Zhang, H.; Karin, M.; Bai, H.; Cai, D. Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity. Cell 2008, 135, 61–73. [Google Scholar] [CrossRef] [Green Version]
- Horvath, T.L.; Sarman, B.; Garcia-Caceres, C.; Enriori, P.J.; Sotonyi, P.; Shanabrough, M.; Borok, E.; Argente, J.; Chowen, J.A.; Perez-Tilve, D.; et al. Synaptic input organization of the melanocortin system predicts diet-induced hypothalamic reactive gliosis and obesity. Proc. Natl. Acad. Sci. USA 2010, 107, 14875–14880. [Google Scholar] [CrossRef] [Green Version]
- Butler, M.J.; Perrini, A.A.; Eckel, L.A. Estradiol treatment attenuates high fat diet-induced microgliosis in ovariectomized rats. Horm. Behav. 2020, 120, 104675. [Google Scholar] [CrossRef]
- De Souza, C.T.; Araujo, E.P.; Bordin, S.; Ashimine, R.; Zollner, R.L.; Boschero, A.C.; Saad, M.J.; Velloso, L.A. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 2005, 146, 4192–4199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Posey, K.A.; Clegg, D.J.; Printz, R.L.; Byun, J.; Morton, G.J.; Vivekanandan-Giri, A.; Pennathur, S.; Baskin, D.G.; Heinecke, J.W.; Woods, S.C.; et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet. Am. J. Physiol. Endocrinol. Metab. 2009, 296, E1003–E1012. [Google Scholar] [CrossRef] [PubMed]
- Cavaliere, G.; Viggiano, E.; Trinchese, G.; de Filippo, C.; Messina, A.; Monda, V.; Valenzano, A.; Cincione, R.I.; Zammit, C.; Cimmino, F.; et al. Long feeding high-fat diet induces hypothalamic oxidative stress and inflammation, and prolonged hypothalamic AMPK activation in rat animal model. Front. Physiol. 2018, 9, 818. [Google Scholar] [CrossRef] [PubMed]
- Ozcan, L.; Ergin, A.S.; Lu, A.; Chung, J.; Sarkar, S.; Nie, D.; Myers, M.G., Jr.; Ozcan, U. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009, 9, 35–51. [Google Scholar] [CrossRef] [Green Version]
- Weissmann, L.; Quaresma, P.G.; Santos, A.C.; de Matos, A.H.; Pascoal, V.D.; Zanotto, T.M.; Castro, G.; Guadagnini, D.; da Silva, J.M.; Velloso, L.A.; et al. IKKepsilon is key to induction of insulin resistance in the hypothalamus, and its inhibition reverses obesity. Diabetes 2014, 63, 3334–3345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andre, C.; Guzman-Quevedo, O.; Rey, C.; Remus-Borel, J.; Clark, S.; Castellanos-Jankiewicz, A.; Ladeveze, E.; Leste-Lasserre, T.; Nadjar, A.; Abrous, D.N.; et al. Inhibiting microglia expansion prevents diet-induced hypothalamic and peripheral inflammation. Diabetes 2017, 66, 908–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valdearcos, M.; Robblee, M.M.; Benjamin, D.I.; Nomura, D.K.; Xu, A.W.; Koliwad, S.K. Microglia dictate the impact of saturated fat consumption on hypothalamic inflammation and neuronal function. Cell Rep. 2014, 9, 2124–2138. [Google Scholar] [CrossRef] [Green Version]
- Valdearcos, M.; Douglass, J.D.; Robblee, M.M.; Dorfman, M.D.; Stifler, D.R.; Bennett, M.L.; Gerritse, I.; Fasnacht, R.; Barres, B.A.; Thaler, J.P.; et al. Microglial inflammatory signaling orchestrates the hypothalamic immune response to dietary excess and mediates obesity susceptibility. Cell Metab. 2017, 26, 185–197.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Tang, Y.; Cai, D. IKKbeta/NF-kappaB disrupts adult hypothalamic neural stem cells to mediate a neurodegenerative mechanism of dietary obesity and pre-diabetes. Nat. Cell Biol. 2012, 14, 999–1012. [Google Scholar] [CrossRef] [Green Version]
- McNay, D.E.; Briancon, N.; Kokoeva, M.V.; Maratos-Flier, E.; Flier, J.S. Remodeling of the arcuate nucleus energy-balance circuit is inhibited in obese mice. J. Clin. Investig. 2012, 122, 142–152. [Google Scholar] [CrossRef] [Green Version]
- Jais, A.; Bruning, J.C. Hypothalamic inflammation in obesity and metabolic disease. J. Clin. Investig. 2017, 127, 24–32. [Google Scholar] [CrossRef]
- Dionysopoulou, S.; Charmandari, E.; Bargiota, A.; Vlahos, N.; Mastorakos, G.; Valsamakis, G. The role of hypothalamic inflammation in diet-induced obesity and its association with cognitive and mood disorders. Nutrients 2021, 13, 498. [Google Scholar] [CrossRef]
- Dorfman, M.D.; Thaler, J.P. Hypothalamic inflammation and gliosis in obesity. Curr. Opin. Endocrinol. Diabetes Obes. 2015, 22, 325–330. [Google Scholar] [CrossRef] [Green Version]
- Schur, E.A.; Melhorn, S.J.; Oh, S.K.; Lacy, J.M.; Berkseth, K.E.; Guyenet, S.J.; Sonnen, J.A.; Tyagi, V.; Rosalynn, M.; de Leon, B.; et al. Radiologic evidence that hypothalamic gliosis is associated with obesity and insulin resistance in humans. Obesity 2015, 23, 2142–2148. [Google Scholar] [CrossRef]
- Puig, J.; Blasco, G.; Daunis, I.E.J.; Molina, X.; Xifra, G.; Ricart, W.; Pedraza, S.; Fernandez-Aranda, F.; Fernandez-Real, J.M. Hypothalamic damage is associated with inflammatory markers and worse cognitive performance in obese subjects. J. Clin. Endocrinol. Metab. 2015, 100, E276–E281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Contreras, C.; Gonzalez-Garcia, I.; Martinez-Sanchez, N.; Seoane-Collazo, P.; Jacas, J.; Morgan, D.A.; Serra, D.; Gallego, R.; Gonzalez, F.; Casals, N.; et al. Central ceramide-induced hypothalamic lipotoxicity and ER stress regulate energy balance. Cell Rep. 2014, 9, 366–377. [Google Scholar] [CrossRef] [Green Version]
- Kleinridders, A.; Schenten, D.; Konner, A.C.; Belgardt, B.F.; Mauer, J.; Okamura, T.; Wunderlich, F.T.; Medzhitov, R.; Bruning, J.C. MyD88 signaling in the CNS is required for development of fatty acid-induced leptin resistance and diet-induced obesity. Cell Metab. 2009, 10, 249–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, H.; Graham, L.C.; Reagan, A.M.; Grabowska, W.A.; Schott, W.H.; Howell, G.R. Transcriptome profiling of brain myeloid cells revealed activation of Itgal, Trem1, and Spp1 in western diet-induced obesity. J. Neuroinflamm. 2019, 16, 169. [Google Scholar] [CrossRef] [PubMed]
- Shigemoto-Mogami, Y.; Hoshikawa, K.; Sato, K. Activated Microglia disrupt the blood-brain barrier and induce chemokines and cytokines in a rat in vitro model. Front. Cell Neurosci. 2018, 12, 494. [Google Scholar] [CrossRef] [PubMed]
- Bar-Or, A.; Nuttall, R.K.; Duddy, M.; Alter, A.; Kim, H.J.; Ifergan, I.; Pennington, C.J.; Bourgoin, P.; Edwards, D.R.; Yong, V.W. Analyses of all matrix metalloproteinase members in leukocytes emphasize monocytes as major inflammatory mediators in multiple sclerosis. Brain 2003, 126 Pt 12, 2738–2749. [Google Scholar] [CrossRef] [Green Version]
- Muri, L.; Leppert, D.; Grandgirard, D.; Leib, S.L. MMPs and ADAMs in neurological infectious diseases and multiple sclerosis. Cell. Mol. Life Sci. 2019, 76, 3097–3116. [Google Scholar] [CrossRef] [PubMed]
- Amaya, J.M.; Suidgeest, E.; Sahut-Barnola, I.; Dumontet, T.; Montanier, N.; Pages, G.; Keller, C.; van der Weerd, L.; Pereira, A.M.; Martinez, A.; et al. Effects of long-term endogenous corticosteroid exposure on brain volume and glial cells in the AdKO mouse. Front. Neurosci. 2021, 15, 604103. [Google Scholar] [CrossRef] [PubMed]
- Van der Werff, S.J.; Andela, C.D.; Nienke Pannekoek, J.; Meijer, O.C.; van Buchem, M.A.; Rombouts, S.A.; van der Mast, R.C.; Biermasz, N.R.; Pereira, A.M.; van der Wee, N.J. Widespread reductions of white matter integrity in patients with long-term remission of Cushing’s disease. Neuroimage Clin. 2014, 4, 659–667. [Google Scholar] [CrossRef] [Green Version]
- Tucsek, Z.; Toth, P.; Tarantini, S.; Sosnowska, D.; Gautam, T.; Warrington, J.P.; Giles, C.B.; Wren, J.D.; Koller, A.; Ballabh, P.; et al. Aging exacerbates obesity-induced cerebromicrovascular rarefaction, neurovascular uncoupling, and cognitive decline in mice. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 1339–1352. [Google Scholar] [CrossRef] [PubMed]
- Profaci, C.P.; Munji, R.N.; Pulido, R.S.; Daneman, R. The blood-brain barrier in health and disease: Important unanswered questions. J. Exp. Med. 2020, 217, e20190062. [Google Scholar] [CrossRef] [PubMed]
- Jolivel, V.; Bicker, F.; Biname, F.; Ploen, R.; Keller, S.; Gollan, R.; Jurek, B.; Birkenstock, J.; Poisa-Beiro, L.; Bruttger, J.; et al. Perivascular microglia promote blood vessel disintegration in the ischemic penumbra. Acta Neuropathol. 2015, 129, 279–295. [Google Scholar] [CrossRef]
- Haruwaka, K.; Ikegami, A.; Tachibana, Y.; Ohno, N.; Konishi, H.; Hashimoto, A.; Matsumoto, M.; Kato, D.; Ono, R.; Kiyama, H.; et al. Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat. Commun. 2019, 10, 5816. [Google Scholar] [CrossRef] [Green Version]
- Sumi, N.; Nishioku, T.; Takata, F.; Matsumoto, J.; Watanabe, T.; Shuto, H.; Yamauchi, A.; Dohgu, S.; Kataoka, Y. Lipopolysaccharide-activated microglia induce dysfunction of the blood-brain barrier in rat microvascular endothelial cells co-cultured with microglia. Cell Mol. Neurobiol. 2010, 30, 247–253. [Google Scholar] [CrossRef] [PubMed]
- Kanoski, S.E.; Zhang, Y.; Zheng, W.; Davidson, T.L. The effects of a high-energy diet on hippocampal function and blood-brain barrier integrity in the rat. J. Alzheimers Dis. 2010, 21, 207–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davidson, T.L.; Monnot, A.; Neal, A.U.; Martin, A.A.; Horton, J.J.; Zheng, W. The effects of a high-energy diet on hippocampal-dependent discrimination performance and blood-brain barrier integrity differ for diet-induced obese and diet-resistant rats. Physiol. Behav. 2012, 107, 26–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Acharya, N.K.; Levin, E.C.; Clifford, P.M.; Han, M.; Tourtellotte, R.; Chamberlain, D.; Pollaro, M.; Coretti, N.J.; Kosciuk, M.C.; Nagele, E.P.; et al. Diabetes and hypercholesterolemia increase blood-brain barrier permeability and brain amyloid deposition: Beneficial effects of the LpPLA2 inhibitor darapladib. J. Alzheimers Dis. 2013, 35, 179–198. [Google Scholar] [CrossRef] [Green Version]
- Hargrave, S.L.; Davidson, T.L.; Zheng, W.; Kinzig, K.P. Western diets induce blood-brain barrier leakage and alter spatial strategies in rats. Behav. Neurosci. 2016, 130, 123–135. [Google Scholar] [CrossRef] [PubMed]
- Fuller, J.P.; Stavenhagen, J.B.; Teeling, J.L. New roles for Fc receptors in neurodegeneration-the impact on Immunotherapy for Alzheimer’s disease. Front. Neurosci. 2014, 8, 235. [Google Scholar] [CrossRef] [Green Version]
- Davalos, D.; Ryu, J.K.; Merlini, M.; Baeten, K.M.; Le Moan, N.; Petersen, M.A.; Deerinck, T.J.; Smirnoff, D.S.; Bedard, C.; Hakozaki, H.; et al. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat. Commun. 2012, 3, 1227. [Google Scholar] [CrossRef] [Green Version]
- Merlini, M.; Rafalski, V.A.; Rios Coronado, P.E.; Gill, T.M.; Ellisman, M.; Muthukumar, G.; Subramanian, K.S.; Ryu, J.K.; Syme, C.A.; Davalos, D.; et al. Fibrinogen induces microglia-mediated spine elimination and cognitive impairment in an Alzheimer’s disease model. Neuron 2019, 101, 1099–1108.e6. [Google Scholar] [CrossRef] [Green Version]
- Nagareddy, P.R.; Kraakman, M.; Masters, S.L.; Stirzaker, R.A.; Gorman, D.J.; Grant, R.W.; Dragoljevic, D.; Hong, E.S.; Abdel-Latif, A.; Smyth, S.S.; et al. Adipose tissue macrophages promote myelopoiesis and monocytosis in obesity. Cell Metab. 2014, 19, 821–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christou, K.A.; Christou, G.A.; Karamoutsios, A.; Vartholomatos, G.; Gartzonika, K.; Tsatsoulis, A.; Tigas, S. Metabolically Healthy Obesity is Characterized by a Proinflammatory Phenotype of Circulating Monocyte Subsets. Metab. Syndr. Relat. Disord. 2019, 17, 259–265. [Google Scholar] [CrossRef]
- Friedrich, K.; Sommer, M.; Strobel, S.; Thrum, S.; Bluher, M.; Wagner, U.; Rossol, M. Perturbation of the monocyte compartment in human obesity. Front. Immunol. 2019, 10, 1874. [Google Scholar] [CrossRef] [Green Version]
- Satoh, N.; Shimatsu, A.; Himeno, A.; Sasaki, Y.; Yamakage, H.; Yamada, K.; Suganami, T.; Ogawa, Y. Unbalanced M1/M2 phenotype of peripheral blood monocytes in obese diabetic patients: Effect of pioglitazone. Diabetes Care 2010, 33, e7. [Google Scholar] [CrossRef] [Green Version]
- Devevre, E.F.; Renovato-Martins, M.; Clement, K.; Sautes-Fridman, C.; Cremer, I.; Poitou, C. Profiling of the three circulating monocyte subpopulations in human obesity. J. Immunol. 2015, 194, 3917–3923. [Google Scholar] [CrossRef]
- Alfakry, H.; Malle, E.; Koyani, C.N.; Pussinen, P.J.; Sorsa, T. Neutrophil proteolytic activation cascades: A possible mechanistic link between chronic periodontitis and coronary heart disease. Innate Immun. 2016, 22, 85–99. [Google Scholar] [CrossRef] [Green Version]
- Niu, H.; Li, Y.; Li, H.; Chi, Y.; Zhuang, M.; Zhang, T.; Liu, M.; Nie, L. Matrix metalloproteinase 12 modulates high-fat-diet induced glomerular fibrogenesis and inflammation in a mouse model of obesity. Sci. Rep. 2016, 6, 20171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fischoeder, A.; Meyborg, H.; Stibenz, D.; Fleck, E.; Graf, K.; Stawowy, P. Insulin augments matrix metalloproteinase-9 expression in monocytes. Cardiovasc. Res. 2007, 73, 841–848. [Google Scholar] [CrossRef] [Green Version]
- Sindhu, S.; Al-Roub, A.; Koshy, M.; Thomas, R.; Ahmad, R. Palmitate-induced MMP-9 expression in the human monocytic cells is mediated through the TLR4-MyD88 dependent mechanism. Cell Physiol. Biochem. 2016, 39, 889–900. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.T.; Pamir, N.; Liu, N.C.; Kirk, E.A.; Averill, M.M.; Becker, L.; Larson, I.; Hagman, D.K.; Foster-Schubert, K.E.; van Yserloo, B.; et al. Macrophage metalloelastase (MMP12) regulates adipose tissue expansion, insulin sensitivity, and expression of inducible nitric oxide synthase. Endocrinology 2014, 155, 3409–3420. [Google Scholar] [CrossRef] [Green Version]
- Behl, T.; Kaur, G.; Sehgal, A.; Bhardwaj, S.; Singh, S.; Buhas, C.; Judea-Pusta, C.; Uivarosan, D.; Munteanu, M.A.; Bungau, S. Multifaceted role of matrix metalloproteinases in neurodegenerative diseases: Pathophysiological and therapeutic perspectives. Int. J. Mol. Sci. 2021, 22, 1413. [Google Scholar] [CrossRef]
- Kouwenhoven, M.; Ozenci, V.; Gomes, A.; Yarilin, D.; Giedraitis, V.; Press, R.; Link, H. Multiple sclerosis: Elevated expression of matrix metalloproteinases in blood monocytes. J. Autoimmun. 2001, 16, 463–470. [Google Scholar] [CrossRef]
- Ong, C.W.; Pabisiak, P.J.; Brilha, S.; Singh, P.; Roncaroli, F.; Elkington, P.T.; Friedland, J.S. Complex regulation of neutrophil-derived MMP-9 secretion in central nervous system tuberculosis. J. Neuroinflamm. 2017, 14, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feng, S.; Cen, J.; Huang, Y.; Shen, H.; Yao, L.; Wang, Y.; Chen, Z. Matrix metalloproteinase-2 and -9 secreted by leukemic cells increase the permeability of blood-brain barrier by disrupting tight junction proteins. PLoS ONE 2011, 6, e20599. [Google Scholar] [CrossRef]
- Goyal, R.; Faizy, A.F.; Siddiqui, S.S.; Singhai, M. Evaluation of TNF-alpha and IL-6 levels in obese and non-obese diabetics: Pre-and postinsulin effects. N. Am. J. Med. Sci. 2012, 4, 180–184. [Google Scholar]
- Alzamil, H. Elevated serum TNF-alpha is related to obesity in type 2 diabetes mellitus and is associated with glycemic control and insulin resistance. J. Obes. 2020, 2020, 5076858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, C.; Gerdes, N.; Fritzenwanger, M.; Figulla, H.R. Circulating levels of interleukin-1 family cytokines in overweight adolescents. Mediat. Inflamm. 2010, 2010, 958403. [Google Scholar] [CrossRef] [PubMed]
- Shoda, H.; Nagafuchi, Y.; Tsuchida, Y.; Sakurai, K.; Sumitomo, S.; Fujio, K.; Yamamoto, K. Increased serum concentrations of IL-1 beta, IL-21 and Th17 cells in overweight patients with rheumatoid arthritis. Arthritis Res. Ther. 2017, 19, 111. [Google Scholar] [CrossRef] [Green Version]
- Knoll, J.G.; Krasnow, S.M.; Marks, D.L. Interleukin-1beta signaling in fenestrated capillaries is sufficient to trigger sickness responses in mice. J. Neuroinflamm. 2017, 14, 219. [Google Scholar] [CrossRef] [Green Version]
- Lively, S.; Schlichter, L.C. Microglia responses to pro-inflammatory stimuli (LPS, IFNgamma+TNFalpha) and reprogramming by resolving cytokines (IL-4, IL-10). Front. Cell Neurosci. 2018, 12, 215. [Google Scholar] [CrossRef] [Green Version]
- Chen, A.Q.; Fang, Z.; Chen, X.L.; Yang, S.; Zhou, Y.F.; Mao, L.; Xia, Y.P.; Jin, H.J.; Li, Y.N.; You, M.F.; et al. Microglia-derived TNF-alpha mediates endothelial necroptosis aggravating blood brain-barrier disruption after ischemic stroke. Cell Death Dis. 2019, 10, 487. [Google Scholar] [CrossRef] [PubMed]
- Guadagno, J.; Xu, X.; Karajgikar, M.; Brown, A.; Cregan, S.P. Microglia-derived TNFalpha induces apoptosis in neural precursor cells via transcriptional activation of the Bcl-2 family member Puma. Cell Death Dis. 2013, 4, e538. [Google Scholar] [CrossRef]
- Rothaug, M.; Becker-Pauly, C.; Rose-John, S. The role of interleukin-6 signaling in nervous tissue. Biochim. Biophys. Acta 2016, 1863 (Pt 6), 1218–1227. [Google Scholar] [CrossRef]
- Hunter, C.A.; Jones, S.A. IL-6 as a keystone cytokine in health and disease. Nat. Immunol. 2015, 16, 448–457. [Google Scholar] [CrossRef]
- Tapia-Gonzalez, S.; Garcia-Segura, L.M.; Tena-Sempere, M.; Frago, L.M.; Castellano, J.M.; Fuente-Martin, E.; Garcia-Caceres, C.; Argente, J.; Chowen, J.A. Activation of microglia in specific hypothalamic nuclei and the cerebellum of adult rats exposed to neonatal overnutrition. J. Neuroendocrinol. 2011, 23, 365–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Recasens, M.; Almolda, B.; Perez-Clausell, J.; Campbell, I.L.; Gonzalez, B.; Castellano, B. Chronic exposure to IL-6 induces a desensitized phenotype of the microglia. J. Neuroinflamm. 2021, 18, 31. [Google Scholar] [CrossRef] [PubMed]
- Gyengesi, E.; Rangel, A.; Ullah, F.; Liang, H.; Niedermayer, G.; Asgarov, R.; Venigalla, M.; Gunawardena, D.; Karl, T.; Munch, G. Chronic microglial activation in the GFAP-IL6 mouse contributes to age-dependent cerebellar volume loss and impairment in motor function. Front. Neurosci. 2019, 13, 303. [Google Scholar] [CrossRef] [Green Version]
- Wallenius, V.; Wallenius, K.; Ahren, B.; Rudling, M.; Carlsten, H.; Dickson, S.L.; Ohlsson, C.; Jansson, J.O. Interleukin-6-deficient mice develop mature-onset obesity. Nat. Med. 2002, 8, 75–79. [Google Scholar] [CrossRef] [PubMed]
- Mishra, D.; Richard, J.E.; Maric, I.; Porteiro, B.; Haring, M.; Kooijman, S.; Musovic, S.; Eerola, K.; Lopez-Ferreras, L.; Peris, E.; et al. Parabrachial interleukin-6 reduces body weight and food intake and increases thermogenesis to regulate energy metabolism. Cell Rep. 2019, 26, 3011–3026.e5. [Google Scholar] [CrossRef] [Green Version]
- Ropelle, E.R.; Flores, M.B.; Cintra, D.E.; Rocha, G.Z.; Pauli, J.R.; Morari, J.; de Souza, C.T.; Moraes, J.C.; Prada, P.O.; Guadagnini, D.; et al. IL-6 and IL-10 anti-inflammatory activity links exercise to hypothalamic insulin and leptin sensitivity through IKKbeta and ER stress inhibition. PLoS Biol. 2010, 8, e1000465. [Google Scholar] [CrossRef]
- Funk, J.A.; Gohlke, J.; Kraft, A.D.; McPherson, C.A.; Collins, J.B.; Jean Harry, G. Voluntary exercise protects hippocampal neurons from trimethyltin injury: Possible role of interleukin-6 to modulate tumor necrosis factor receptor-mediated neurotoxicity. Brain Behav. Immun. 2011, 25, 1063–1077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Willis, E.F.; MacDonald, K.P.A.; Nguyen, Q.H.; Garrido, A.L.; Gillespie, E.R.; Harley, S.B.R.; Bartlett, P.F.; Schroder, W.A.; Yates, A.G.; Anthony, D.C.; et al. Repopulating microglia promote brain repair in an IL-6-dependent manner. Cell 2020, 180, 833–846.e16. [Google Scholar] [CrossRef]
- Ahima, R.S.; Flier, J.S. Leptin. Annu. Rev. Physiol. 2000, 62, 413–437. [Google Scholar] [CrossRef] [Green Version]
- Zhao, S.; Kusminski, C.M.; Elmquist, J.K.; Scherer, P.E. Leptin: Less is more. Diabetes 2020, 69, 823–829. [Google Scholar] [CrossRef]
- Caron, A.; Lee, S.; Elmquist, J.K.; Gautron, L. Leptin and brain-adipose crosstalks. Nat. Rev. Neurosci. 2018, 19, 153–165. [Google Scholar] [CrossRef] [PubMed]
- Fujita, Y.; Yamashita, T. The effects of leptin on glial cells in neurological diseases. Front. Neurosci. 2019, 13, 828. [Google Scholar] [CrossRef] [PubMed]
- McGregor, G.; Harvey, J. Leptin regulation of synaptic function at hippocampal TA-CA1 and SC-CA1 synapses: Implications for health and disease. Neurochem. Res. 2019, 44, 650–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Achari, A.E.; Jain, S.K. Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int. J. Mol. Sci. 2017, 18, 1321. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Palanivel, R.; Rai, E.; Park, M.; Gabor, T.V.; Scheid, M.P.; Xu, A.; Sweeney, G. Adiponectin stimulates autophagy and reduces oxidative stress to enhance insulin sensitivity during high-fat diet feeding in mice. Diabetes 2015, 64, 36–48. [Google Scholar] [CrossRef] [Green Version]
- Kusminski, C.M.; McTernan, P.G.; Schraw, T.; Kos, K.; O’Hare, J.P.; Ahima, R.; Kumar, S.; Scherer, P.E. Adiponectin complexes in human cerebrospinal fluid: Distinct complex distribution from serum. Diabetologia 2007, 50, 634–642. [Google Scholar] [CrossRef]
- Liu, J.; Guo, M.; Zhang, D.; Cheng, S.Y.; Liu, M.; Ding, J.; Scherer, P.E.; Liu, F.; Lu, X.Y. Adiponectin is critical in determining susceptibility to depressive behaviors and has antidepressant-like activity. Proc. Natl. Acad. Sci. USA 2012, 109, 12248–12253. [Google Scholar] [CrossRef] [Green Version]
- Qi, Y.; Takahashi, N.; Hileman, S.M.; Patel, H.R.; Berg, A.H.; Pajvani, U.B.; Scherer, P.E.; Ahima, R.S. Adiponectin acts in the brain to decrease body weight. Nat. Med. 2004, 10, 524–529. [Google Scholar] [CrossRef]
- Yau, S.Y.; Li, A.; Hoo, R.L.; Ching, Y.P.; Christie, B.R.; Lee, T.M.; Xu, A.; So, K.F. Physical exercise-induced hippocampal neurogenesis and antidepressant effects are mediated by the adipocyte hormone adiponectin. Proc. Natl. Acad. Sci. USA 2014, 111, 15810–15815. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.; Tu, T.H.; Park, B.S.; Yang, S.; Kim, J.G. Adiponectin reverses the hypothalamic microglial inflammation during short-term exposure to fat-rich diet. Int. J. Mol. Sci. 2019, 20, 5738. [Google Scholar] [CrossRef] [Green Version]
- Ng, R.C.; Cheng, O.Y.; Jian, M.; Kwan, J.S.; Ho, P.W.; Cheng, K.K.; Yeung, P.K.; Zhou, L.L.; Hoo, R.L.; Chung, S.K.; et al. Chronic adiponectin deficiency leads to Alzheimer’s disease-like cognitive impairments and pathologies through AMPK inactivation and cerebral insulin resistance in aged mice. Mol. Neurodegener. 2016, 11, 71. [Google Scholar] [CrossRef] [Green Version]
- Ng, R.C.; Jian, M.; Ma, O.K.; Bunting, M.; Kwan, J.S.; Zhou, G.J.; Senthilkumar, K.; Iyaswamy, A.; Chan, P.K.; Li, M.; et al. Chronic oral administration of adipoRon reverses cognitive impairments and ameliorates neuropathology in an Alzheimer’s disease mouse model. Mol. Psychiatry 2020. [Google Scholar] [CrossRef] [PubMed]
- Chabry, J.; Nicolas, S.; Cazareth, J.; Murris, E.; Guyon, A.; Glaichenhaus, N.; Heurteaux, C.; Petit-Paitel, A. Enriched environment decreases microglia and brain macrophages inflammatory phenotypes through adiponectin-dependent mechanisms: Relevance to depressive-like behavior. Brain Behav. Immun. 2015, 50, 275–287. [Google Scholar] [CrossRef] [PubMed]
- Nicolas, S.; Veyssiere, J.; Gandin, C.; Zsurger, N.; Pietri, M.; Heurteaux, C.; Glaichenhaus, N.; Petit-Paitel, A.; Chabry, J. Neurogenesis-independent antidepressant-like effects of enriched environment is dependent on adiponectin. Psychoneuroendocrinology 2015, 57, 72–83. [Google Scholar] [CrossRef]
- Nicolas, S.; Cazareth, J.; Zarif, H.; Guyon, A.; Heurteaux, C.; Chabry, J.; Petit-Paitel, A. Globular adiponectin limits microglia pro-inflammatory phenotype through an AdipoR1/NF-kappaB signaling pathway. Front. Cell Neurosci. 2017, 11, 352. [Google Scholar] [CrossRef] [Green Version]
- Song, J.; Choi, S.M.; Kim, B.C. Adiponectin regulates the polarization and function of microglia via PPAR-gamma signaling under amyloid beta toxicity. Front. Cell Neurosci. 2017, 11, 64. [Google Scholar] [CrossRef] [Green Version]
- Jian, M.; Kwan, J.S.; Bunting, M.; Ng, R.C.; Chan, K.H. Adiponectin suppresses amyloid-beta oligomer (AbetaO)-induced inflammatory response of microglia via AdipoR1-AMPK-NF-kappaB signaling pathway. J. Neuroinflamm. 2019, 16, 110. [Google Scholar] [CrossRef] [PubMed]
- Zheng, J.; Sun, Z.; Liang, F.; Xu, W.; Lu, J.; Shi, L.; Shao, A.; Yu, J.; Zhang, J. AdipoRon attenuates neuroinflammation after intracerebral hemorrhage through AdipoR1-AMPK pathway. Neuroscience 2019, 412, 116–130. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Chen, S.; Sherchan, P.; Ding, Y.; Zhao, W.; Guo, Z.; Yu, J.; Tang, J.; Zhang, J.H. Recombinant CTRP9 administration attenuates neuroinflammation via activating adiponectin receptor 1 after intracerebral hemorrhage in mice. J. Neuroinflamm. 2018, 15, 215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hahm, J.R.; Jo, M.H.; Ullah, R.; Kim, M.W.; Kim, M.O. Metabolic stress alters antioxidant systems, suppresses the adiponectin receptor 1 and induces Alzheimer’s like pathology in mice brain. Cells 2020, 9, 249. [Google Scholar] [CrossRef] [Green Version]
- Bays, H.E.; Toth, P.P.; Kris-Etherton, P.M.; Abate, N.; Aronne, L.J.; Brown, W.V.; Gonzalez-Campoy, J.M.; Jones, S.R.; Kumar, R.; La Forge, R.; et al. Obesity, adiposity, and dyslipidemia: A consensus statement from the National Lipid Association. J. Clin. Lipidol. 2013, 7, 304–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, H.; Urso, C.J.; Jadeja, V. Saturated fatty acids in obesity-associated inflammation. J. Inflamm. Res. 2020, 13, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Zhao, L.; Ni, Y.; Ma, X.; Zhao, A.; Bao, Y.; Liu, J.; Chen, T.; Xie, G.; Panee, J.; Su, M.; et al. A panel of free fatty acid ratios to predict the development of metabolic abnormalities in healthy obese individuals. Sci. Rep. 2016, 6, 28418. [Google Scholar] [CrossRef] [Green Version]
- Dumas, J.A.; Bunn, J.Y.; Nickerson, J.; Crain, K.I.; Ebenstein, D.B.; Tarleton, E.K.; Makarewicz, J.; Poynter, M.E.; Kien, C.L. Dietary saturated fat and monounsaturated fat have reversible effects on brain function and the secretion of pro-inflammatory cytokines in young women. Metabolism 2016, 65, 1582–1588. [Google Scholar] [CrossRef] [Green Version]
- Pakiet, A.; Jakubiak, A.; Mierzejewska, P.; Zwara, A.; Liakh, I.; Sledzinski, T.; Mika, A. The effect of a high-fat diet on the fatty acid composition in the hearts of mice. Nutrients 2020, 12, 824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tu, T.H.; Kim, H.; Yang, S.; Kim, J.K.; Kim, J.G. Linoleic acid rescues microglia inflammation triggered by saturated fatty acid. Biochem. Biophys. Res. Commun. 2019, 513, 201–206. [Google Scholar] [CrossRef] [PubMed]
- Karmi, A.; Iozzo, P.; Viljanen, A.; Hirvonen, J.; Fielding, B.A.; Virtanen, K.; Oikonen, V.; Kemppainen, J.; Viljanen, T.; Guiducci, L.; et al. Increased brain fatty acid uptake in metabolic syndrome. Diabetes 2010, 59, 2171–2177. [Google Scholar] [CrossRef] [Green Version]
- Borg, M.L.; Omran, S.F.; Weir, J.; Meikle, P.J.; Watt, M.J. Consumption of a high-fat diet, but not regular endurance exercise training, regulates hypothalamic lipid accumulation in mice. J. Physiol. 2012, 590, 4377–4389. [Google Scholar] [CrossRef] [Green Version]
- Melo, H.M.; Seixas da Silva, G.D.S.; Sant’Ana, M.R.; Teixeira, C.V.L.; Clarke, J.R.; Miya Coreixas, V.S.; de Melo, B.C.; Fortuna, J.T.S.; Forny-Germano, L.; Ledo, J.H.; et al. Palmitate is increased in the cerebrospinal fluid of humans with obesity and induces memory impairment in mice via pro-inflammatory TNF-alpha. Cell Rep. 2020, 30, 2180–2194.e8. [Google Scholar] [CrossRef] [Green Version]
- Spinelli, M.; Fusco, S.; Mainardi, M.; Scala, F.; Natale, F.; Lapenta, R.; Mattera, A.; Rinaudo, M.; Li Puma, D.D.; Ripoli, C.; et al. Brain insulin resistance impairs hippocampal synaptic plasticity and memory by increasing GluA1 palmitoylation through FoxO3a. Nat. Commun. 2017, 8, 2009. [Google Scholar] [CrossRef]
- Morselli, E.; Fuente-Martin, E.; Finan, B.; Kim, M.; Frank, A.; Garcia-Caceres, C.; Navas, C.R.; Gordillo, R.; Neinast, M.; Kalainayakan, S.P.; et al. Hypothalamic PGC-1alpha protects against high-fat diet exposure by regulating ERalpha. Cell Rep. 2014, 9, 633–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marwarha, G.; Claycombe, K.; Schommer, J.; Collins, D.; Ghribi, O. Palmitate-induced endoplasmic reticulum stress and subsequent C/EBPalpha Homologous Protein activation attenuates leptin and Insulin-like growth factor 1 expression in the brain. Cell Sign. 2016, 28, 1789–1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Plociennikowska, A.; Hromada-Judycka, A.; Borzecka, K.; Kwiatkowska, K. Co-operation of TLR4 and raft proteins in LPS-induced pro-inflammatory signaling. Cell. Mol. Life Sci. 2015, 72, 557–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bosetti, F. Arachidonic acid metabolism in brain physiology and pathology: Lessons from genetically altered mouse models. J. Neurochem. 2007, 102, 577–586. [Google Scholar] [CrossRef] [Green Version]
- Simopoulos, A.P. Evolutionary aspects of diet: The omega-6/omega-3 ratio and the brain. Mol. Neurobiol. 2011, 44, 203–215. [Google Scholar] [CrossRef]
- Serhan, C.N.; Chiang, N. Resolution phase lipid mediators of inflammation: Agonists of resolution. Curr. Opin. Pharmacol. 2013, 13, 632–640. [Google Scholar] [CrossRef] [Green Version]
- Hjorth, E.; Zhu, M.; Toro, V.C.; Vedin, I.; Palmblad, J.; Cederholm, T.; Freund-Levi, Y.; Faxen-Irving, G.; Wahlund, L.O.; Basun, H.; et al. Omega-3 fatty acids enhance phagocytosis of Alzheimer’s disease-related amyloid-beta42 by human microglia and decrease inflammatory markers. J. Alzheimers Dis. 2013, 35, 697–713. [Google Scholar] [CrossRef] [Green Version]
- Ma, Q.L.; Zhu, C.; Morselli, M.; Su, T.; Pelligrini, M.; Lu, Z.; Jones, M.; Denver, P.; Castro, D.; Gu, X.; et al. The novel omega-6 fatty acid docosapentaenoic acid positively modulates brain innate immune response for resolving neuroinflammation at early and late stages of humanized APOE-based Alzheimer’s disease Models. Front. Immunol. 2020, 11, 558036. [Google Scholar] [CrossRef]
- Chen, X.; Wu, S.; Chen, C.; Xie, B.; Fang, Z.; Hu, W.; Chen, J.; Fu, H.; He, H. Omega-3 polyunsaturated fatty acid supplementation attenuates microglial-induced inflammation by inhibiting the HMGB1/TLR4/NF-kappaB pathway following experimental traumatic brain injury. J. Neuroinflamm. 2017, 14, 143. [Google Scholar] [CrossRef]
- Chen, X.; Chen, C.; Fan, S.; Wu, S.; Yang, F.; Fang, Z.; Fu, H.; Li, Y. Omega-3 polyunsaturated fatty acid attenuates the inflammatory response by modulating microglia polarization through SIRT1-mediated deacetylation of the HMGB1/NF-kappaB pathway following experimental traumatic brain injury. J. Neuroinflamm. 2018, 15, 116. [Google Scholar] [CrossRef]
- Chen, S.; Zhang, H.; Pu, H.; Wang, G.; Li, W.; Leak, R.K.; Chen, J.; Liou, A.K.; Hu, X. n-3 PUFA supplementation benefits microglial responses to myelin pathology. Sci. Rep. 2014, 4, 7458. [Google Scholar] [CrossRef]
- Chen, W.; Balland, E.; Cowley, M.A. Hypothalamic insulin resistance in obesity: Effects on glucose homeostasis. Neuroendocrinology 2017, 104, 364–381. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.; Joo, Y.J.; Kim, R.Y.; Hwang, J.; Lim, S.M.; Yoon, S.; Kim, J. Obesity may connect insulin resistance to decreased neuronal viability in human diabetic brain. Obesity 2020, 28, 1626–1630. [Google Scholar] [CrossRef] [PubMed]
- Kullmann, S.; Valenta, V.; Wagner, R.; Tschritter, O.; Machann, J.; Haring, H.U.; Preissl, H.; Fritsche, A.; Heni, M. Brain insulin sensitivity is linked to adiposity and body fat distribution. Nat. Commun. 2020, 11, 1841. [Google Scholar] [CrossRef] [Green Version]
- Maher, P.A.; Schubert, D.R. Metabolic links between diabetes and Alzheimer’s disease. Expert Rev. Neurother. 2009, 9, 617–630. [Google Scholar] [CrossRef]
- Fishel, M.A.; Watson, G.S.; Montine, T.J.; Wang, Q.; Green, P.S.; Kulstad, J.J.; Cook, D.G.; Peskind, E.R.; Baker, L.D.; Goldgaber, D.; et al. Hyperinsulinemia provokes synchronous increases in central inflammation and beta-amyloid in normal adults. Arch. Neurol. 2005, 62, 1539–1544. [Google Scholar] [CrossRef] [PubMed]
- Ho, L.; Qin, W.; Pompl, P.N.; Xiang, Z.; Wang, J.; Zhao, Z.; Peng, Y.; Cambareri, G.; Rocher, A.; Mobbs, C.V.; et al. Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. FASEB J. 2004, 18, 902–904. [Google Scholar] [CrossRef]
- Takeda, S.; Sato, N.; Uchio-Yamada, K.; Sawada, K.; Kunieda, T.; Takeuchi, D.; Kurinami, H.; Shinohara, M.; Rakugi, H.; Morishita, R. Diabetes-accelerated memory dysfunction via cerebrovascular inflammation and Abeta deposition in an Alzheimer mouse model with diabetes. Proc. Natl. Acad. Sci. USA 2010, 107, 7036–7041. [Google Scholar] [CrossRef] [Green Version]
- Dinel, A.L.; Andre, C.; Aubert, A.; Ferreira, G.; Laye, S.; Castanon, N. Cognitive and emotional alterations are related to hippocampal inflammation in a mouse model of metabolic syndrome. PLoS ONE 2011, 6, e24325. [Google Scholar] [CrossRef]
- Pistell, P.J.; Morrison, C.D.; Gupta, S.; Knight, A.G.; Keller, J.N.; Ingram, D.K.; Bruce-Keller, A.J. Cognitive impairment following high fat diet consumption is associated with brain inflammation. J. Neuroimmunol. 2010, 219, 25–32. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Zhai, Y.Q.; Xu, L.L.; Qiao, C.; Sun, X.L.; Ding, J.H.; Lu, M.; Hu, G. Metabolic inflammation exacerbates dopaminergic neuronal degeneration in response to acute MPTP challenge in type 2 diabetes mice. Exp. Neurol. 2014, 251, 22–29. [Google Scholar] [CrossRef] [PubMed]
- Haas, C.B.; de Carvalho, A.K.; Muller, A.P.; Eggen, B.J.L.; Portela, L.V. Insulin activates microglia and increases COX-2/IL-1beta expression in young but not in aged hippocampus. Brain Res. 2020, 1741, 146884. [Google Scholar] [CrossRef]
- Ieronymaki, E.; Theodorakis, E.M.; Lyroni, K.; Vergadi, E.; Lagoudaki, E.; Al-Qahtani, A.; Aznaourova, M.; Neofotistou-Themeli, E.; Eliopoulos, A.G.; Vaporidi, K.; et al. Insulin resistance in macrophages alters their metabolism and promotes an M2-like phenotype. J. Immunol. 2019, 202, 1786–1797. [Google Scholar] [CrossRef] [PubMed]
- Galitzky, J.; Bouloumie, A. Human visceral-fat-specific glucocorticoid tuning of adipogenesis. Cell Metab. 2013, 18, 3–5. [Google Scholar] [CrossRef] [Green Version]
- Mateska, I.; Nanda, K.; Dye, N.A.; Alexaki, V.I.; Eaton, S. Range of SHH signaling in adrenal gland is limited by membrane contact to cells with primary cilia. J. Cell Biol. 2020, 219, e201910087. [Google Scholar] [CrossRef]
- Steffensen, C.; Dekkers, O.M.; Lyhne, J.; Pedersen, B.G.; Rasmussen, F.; Rungby, J.; Poulsen, P.L.; Jorgensen, J.O.L. Hypercortisolism in Newly Diagnosed Type 2 Diabetes: A Prospective Study of 384 Newly Diagnosed Patients. Horm. Metab. Res. 2019, 51, 62–68. [Google Scholar] [CrossRef] [PubMed]
- Yau, S.Y.; Lau, B.W.; Tong, J.B.; Wong, R.; Ching, Y.P.; Qiu, G.; Tang, S.W.; Lee, T.M.; So, K.F. Hippocampal neurogenesis and dendritic plasticity support running-improved spatial learning and depression-like behaviour in stressed rats. PLoS ONE 2011, 6, e24263. [Google Scholar] [CrossRef] [Green Version]
- Brummelte, S.; Galea, L.A. Chronic high corticosterone reduces neurogenesis in the dentate gyrus of adult male and female rats. Neuroscience 2010, 168, 680–690. [Google Scholar] [CrossRef] [Green Version]
- Park, H.J.; Lee, S.; Jung, J.W.; Kim, B.C.; Ryu, J.H.; Kim, D.H. Glucocorticoid- and long-term stress-induced aberrant synaptic plasticity are mediated by activation of the glucocorticoid receptor. Arch. Pharm. Res. 2015, 38, 1204–1212. [Google Scholar] [CrossRef]
- Coutinho, A.E.; Chapman, K.E. The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol. Cell. Endocrinol. 2011, 335, 2–13. [Google Scholar] [CrossRef]
- Munhoz, C.D.; Sorrells, S.F.; Caso, J.R.; Scavone, C.; Sapolsky, R.M. Glucocorticoids exacerbate lipopolysaccharide-induced signaling in the frontal cortex and hippocampus in a dose-dependent manner. J. Neurosci. 2010, 30, 13690–13698. [Google Scholar] [CrossRef] [Green Version]
- Munhoz, C.D.; Lepsch, L.B.; Kawamoto, E.M.; Malta, M.B.; Lima Lde, S.; Avellar, M.C.; Sapolsky, R.M.; Scavone, C. Chronic unpredictable stress exacerbates lipopolysaccharide-induced activation of nuclear factor-kappaB in the frontal cortex and hippocampus via glucocorticoid secretion. J. Neurosci. 2006, 26, 3813–3820. [Google Scholar] [CrossRef]
- Garcia-Bueno, B.; Madrigal, J.L.; Perez-Nievas, B.G.; Leza, J.C. Stress mediators regulate brain prostaglandin synthesis and peroxisome proliferator-activated receptor-gamma activation after stress in rats. Endocrinology 2008, 149, 1969–1978. [Google Scholar] [CrossRef] [Green Version]
- Sierra, A.; Gottfried-Blackmore, A.; Milner, T.A.; McEwen, B.S.; Bulloch, K. Steroid hormone receptor expression and function in microglia. Glia 2008, 56, 659–674. [Google Scholar] [CrossRef]
- Carrillo-de Sauvage, M.A.; Maatouk, L.; Arnoux, I.; Pasco, M.; Sanz Diez, A.; Delahaye, M.; Herrero, M.T.; Newman, T.A.; Calvo, C.F.; Audinat, E.; et al. Potent and multiple regulatory actions of microglial glucocorticoid receptors during CNS inflammation. Cell Death Differ. 2013, 20, 1546–1557. [Google Scholar] [CrossRef] [PubMed]
- Frank, M.G.; Fonken, L.K.; Watkins, L.R.; Maier, S.F. Microglia: Neuroimmune-sensors of stress. Semin. Cell Dev. Biol. 2019, 94, 176–185. [Google Scholar] [CrossRef]
- Sanguino-Gomez, J.; Buurstede, J.C.; Abiega, O.; Fitzsimons, C.P.; Lucassen, P.J.; Eggen, B.J.L.; Lesuis, S.L.; Meijer, O.C.; Krugers, H.J. An emerging role for microglia in stress-effects on memory. Eur. J. Neurosci. 2021. [Google Scholar] [CrossRef] [PubMed]
- De Pablos, R.M.; Villaran, R.F.; Arguelles, S.; Herrera, A.J.; Venero, J.L.; Ayala, A.; Cano, J.; Machado, A. Stress increases vulnerability to inflammation in the rat prefrontal cortex. J. Neurosci. 2006, 26, 5709–5719. [Google Scholar] [CrossRef] [PubMed]
- Pedrazzoli, M.; Losurdo, M.; Paolone, G.; Medelin, M.; Jaupaj, L.; Cisterna, B.; Slanzi, A.; Malatesta, M.; Coco, S.; Buffelli, M. Glucocorticoid receptors modulate dendritic spine plasticity and microglia activity in an animal model of Alzheimer’s disease. Neurobiol. Dis. 2019, 132, 104568. [Google Scholar] [CrossRef]
- Hoeijmakers, L.; Ruigrok, S.R.; Amelianchik, A.; Ivan, D.; van Dam, A.M.; Lucassen, P.J.; Korosi, A. Early-life stress lastingly alters the neuroinflammatory response to amyloid pathology in an Alzheimer’s disease mouse model. Brain Behav. Immun. 2017, 63, 160–175. [Google Scholar] [CrossRef]
- Catale, C.; Gironda, S.; Lo Iacono, L.; Carola, V. Microglial function in the effects of early-life stress on brain and behavioral development. J. Clin. Med. 2020, 9, 468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dey, A.; Hao, S.; Erion, J.R.; Wosiski-Kuhn, M.; Stranahan, A.M. Glucocorticoid sensitization of microglia in a genetic mouse model of obesity and diabetes. J. Neuroimmunol. 2014, 269, 20–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stranahan, A.M.; Arumugam, T.V.; Mattson, M.P. Lowering corticosterone levels reinstates hippocampal brain-derived neurotropic factor and Trkb expression without influencing deficits in hypothalamic brain-derived neurotropic factor expression in leptin receptor-deficient mice. Neuroendocrinology 2011, 93, 58–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wosiski-Kuhn, M.; Erion, J.R.; Gomez-Sanchez, E.P.; Gomez-Sanchez, C.E.; Stranahan, A.M. Glucocorticoid receptor activation impairs hippocampal plasticity by suppressing BDNF expression in obese mice. Psychoneuroendocrinology 2014, 42, 165–177. [Google Scholar] [CrossRef] [Green Version]
- Frank, M.G.; Baratta, M.V.; Sprunger, D.B.; Watkins, L.R.; Maier, S.F. Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav. Immun. 2007, 21, 47–59. [Google Scholar] [CrossRef]
- Frank, M.G.; Thompson, B.M.; Watkins, L.R.; Maier, S.F. Glucocorticoids mediate stress-induced priming of microglial pro-inflammatory responses. Brain Behav. Immun. 2012, 26, 337–345. [Google Scholar] [CrossRef] [Green Version]
- Duque Ede, A.; Munhoz, C.D. The pro-inflammatory effects of glucocorticoids in the brain. Front. Endocrinol. 2016, 7, 78. [Google Scholar] [CrossRef] [Green Version]
- Lim, H.Y.; Muller, N.; Herold, M.J.; van den Brandt, J.; Reichardt, H.M. Glucocorticoids exert opposing effects on macrophage function dependent on their concentration. Immunology 2007, 122, 47–53. [Google Scholar] [CrossRef]
- Dhabhar, F.S.; McEwen, B.S. Enhancing versus suppressive effects of stress hormones on skin immune function. Proc. Natl. Acad. Sci. USA 1999, 96, 1059–1064. [Google Scholar] [CrossRef] [Green Version]
- Quatrini, L.; Ugolini, S. New insights into the cell- and tissue-specificity of glucocorticoid actions. Cell. Mol. Immunol. 2020, 18, 269–278. [Google Scholar] [CrossRef]
- Tilg, H.; Zmora, N.; Adolph, T.E.; Elinav, E. The intestinal microbiota fuelling metabolic inflammation. Nat. Rev. Immunol. 2020, 20, 40–54. [Google Scholar] [CrossRef] [PubMed]
- Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef] [PubMed]
- Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Cheng, J.; Duncan, A.E.; Kau, A.L.; Griffin, N.W.; Lombard, V.; Henrissat, B.; Bain, J.R.; et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013, 341, 1241214. [Google Scholar] [CrossRef] [Green Version]
- Backhed, F.; Manchester, J.K.; Semenkovich, C.F.; Gordon, J.I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc. Natl. Acad. Sci. USA 2007, 104, 979–984. [Google Scholar] [CrossRef] [Green Version]
- Rabot, S.; Membrez, M.; Bruneau, A.; Gerard, P.; Harach, T.; Moser, M.; Raymond, F.; Mansourian, R.; Chou, C.J. Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J. 2010, 24, 4948–4959. [Google Scholar]
- Fung, T.C.; Olson, C.A.; Hsiao, E.Y. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017, 20, 145–155. [Google Scholar] [CrossRef]
- Bischoff, S.C.; Barbara, G.; Buurman, W.; Ockhuizen, T.; Schulzke, J.D.; Serino, M.; Tilg, H.; Watson, A.; Wells, J.M. Intestinal permeability--a new target for disease prevention and therapy. BMC Gastroenterol. 2014, 14, 189. [Google Scholar] [CrossRef] [Green Version]
- Shi, H.; Wang, Q.; Zheng, M.; Hao, S.; Lum, J.S.; Chen, X.; Huang, X.F.; Yu, Y.; Zheng, K. Supplement of microbiota-accessible carbohydrates prevents neuroinflammation and cognitive decline by improving the gut microbiota-brain axis in diet-induced obese mice. J. Neuroinflamm. 2020, 17, 77. [Google Scholar] [CrossRef]
- Chen, J.; Haase, N.; Haange, S.B.; Sucher, R.; Munzker, J.; Jager, E.; Schischke, K.; Seyfried, F.; von Bergen, M.; Hankir, M.K.; et al. Roux-en-Y gastric bypass contributes to weight loss-independent improvement in hypothalamic inflammation and leptin sensitivity through gut-microglia-neuron-crosstalk. Mol. Metab. 2021, 48, 101214. [Google Scholar] [CrossRef] [PubMed]
- D’Mello, C.; Ronaghan, N.; Zaheer, R.; Dicay, M.; Le, T.; MacNaughton, W.K.; Surrette, M.G.; Swain, M.G. Probiotics improve inflammation-associated sickness behavior by altering communication between the peripheral immune system and the brain. J. Neurosci. 2015, 35, 10821–10830. [Google Scholar] [CrossRef] [Green Version]
- Shi, H.; Yu, Y.; Lin, D.; Zheng, P.; Zhang, P.; Hu, M.; Wang, Q.; Pan, W.; Yang, X.; Hu, T.; et al. beta-glucan attenuates cognitive impairment via the gut-brain axis in diet-induced obese mice. Microbiome 2020, 8, 143. [Google Scholar] [CrossRef] [PubMed]
- Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; von Glehn, F.; Yan, R.; Patel, B.; Mazzola, M.A.; Liu, S.; Glanz, B.L.; et al. Alterations of the human gut microbiome in multiple sclerosis. Nat. Commun. 2016, 7, 12015. [Google Scholar] [CrossRef]
- Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef] [PubMed]
- Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef] [Green Version]
- Kaiser, T.; Feng, G. Tmem119-EGFP and Tmem119-CreERT2 transgenic mice for labeling and manipulating microglia. eNeuro 2019, 6. [Google Scholar] [CrossRef] [Green Version]
- Goldmann, T.; Wieghofer, P.; Muller, P.F.; Wolf, Y.; Varol, D.; Yona, S.; Brendecke, S.M.; Kierdorf, K.; Staszewski, O.; Datta, M.; et al. A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation. Nat. Neurosci. 2013, 16, 1618–1626. [Google Scholar] [CrossRef]
- McKinsey, G.L.; Lizama, C.O.; Keown-Lang, A.E.; Niu, A.; Santander, N.; Larpthaveesarp, A.; Chee, E.; Gonzalez, F.F.; Arnold, T.D. A new genetic strategy for targeting microglia in development and disease. Elife 2020, 9, e54590. [Google Scholar] [CrossRef]
- Bohlen, C.J.; Bennett, F.C.; Tucker, A.F.; Collins, H.Y.; Mulinyawe, S.B.; Barres, B.A. Diverse requirements for microglial survival, specification, and function revealed by defined-medium cultures. Neuron 2017, 94, 759–773.e8. [Google Scholar] [CrossRef] [Green Version]
- Delbridge, A.R.D.; Huh, D.; Brickelmaier, M.; Burns, J.C.; Roberts, C.; Challa, R.; Raymond, N.; Cullen, P.; Carlile, T.M.; Ennis, K.A.; et al. Organotypic brain slice culture microglia exhibit molecular similarity to acutely-isolated adult microglia and provide a platform to study neuroinflammation. Front. Cell Neurosci. 2020, 14, 592005. [Google Scholar] [CrossRef]
- Han, J.; Zhu, K.; Zhang, X.M.; Harris, R.A. Enforced microglial depletion and repopulation as a promising strategy for the treatment of neurological disorders. Glia 2019, 67, 217–231. [Google Scholar] [CrossRef] [Green Version]
- Nissen, J.C.; Thompson, K.K.; West, B.L.; Tsirka, S.E. Csf1R inhibition attenuates experimental autoimmune encephalomyelitis and promotes recovery. Exp. Neurol. 2018, 307, 24–36. [Google Scholar] [CrossRef]
- Beckmann, N.; Giorgetti, E.; Neuhaus, A.; Zurbruegg, S.; Accart, N.; Smith, P.; Perdoux, J.; Perrot, L.; Nash, M.; Desrayaud, S.; et al. Brain region-specific enhancement of remyelination and prevention of demyelination by the CSF1R kinase inhibitor BLZ945. Acta Neuropathol. Commun. 2018, 6, 9. [Google Scholar] [CrossRef] [Green Version]
- Acharya, M.M.; Green, K.N.; Allen, B.D.; Najafi, A.R.; Syage, A.; Minasyan, H.; Le, M.T.; Kawashita, T.; Giedzinski, E.; Parihar, V.K.; et al. Elimination of microglia improves cognitive function following cranial irradiation. Sci. Rep. 2016, 6, 31545. [Google Scholar] [CrossRef]
- Ali, S.; Mansour, A.G.; Huang, W.; Queen, N.J.; Mo, X.; Anderson, J.M.; Hassan, Q.N., 2nd; Patel, R.S.; Wilkins, R.K.; Caligiuri, M.A.; et al. CSF1R inhibitor PLX5622 and environmental enrichment additively improve metabolic outcomes in middle-aged female mice. Aging 2020, 12, 2101–2122. [Google Scholar] [CrossRef]
- Spangenberg, E.E.; Lee, R.J.; Najafi, A.R.; Rice, R.A.; Elmore, M.R.; Blurton-Jones, M.; West, B.L.; Green, K.N. Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-beta pathology. Brain 2016, 139 Pt 4, 1265–1281. [Google Scholar] [CrossRef] [Green Version]
- Zhao, R.; Hu, W.; Tsai, J.; Li, W.; Gan, W.B. Microglia limit the expansion of beta-amyloid plaques in a mouse model of Alzheimer’s disease. Mol. Neurodegener. 2017, 12, 47. [Google Scholar] [CrossRef]
- Wheeler, D.L.; Sariol, A.; Meyerholz, D.K.; Perlman, S. Microglia are required for protection against lethal coronavirus encephalitis in mice. J. Clin. Investig. 2018, 128, 931–943. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Ren, H.; Wood, K.; Li, M.; Qiu, S.; Shi, F.D.; Ma, C.; Liu, Q. Depletion of microglia augments the dopaminergic neurotoxicity of MPTP. FASEB J. 2018, 32, 3336–3345. [Google Scholar] [CrossRef] [Green Version]
- Varvel, N.H.; Grathwohl, S.A.; Baumann, F.; Liebig, C.; Bosch, A.; Brawek, B.; Thal, D.R.; Charo, I.F.; Heppner, F.L.; Aguzzi, A.; et al. Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells. Proc. Natl. Acad. Sci. USA 2012, 109, 18150–18155. [Google Scholar] [CrossRef] [Green Version]
- Scott, C.L.; Zheng, F.; de Baetselier, P.; Martens, L.; Saeys, Y.; de Prijck, S.; Lippens, S.; Abels, C.; Schoonooghe, S.; Raes, G.; et al. Bone marrow-derived monocytes give rise to self-renewing and fully differentiated Kupffer cells. Nat. Commun. 2016, 7, 10321. [Google Scholar] [CrossRef]
- Rice, R.A.; Pham, J.; Lee, R.J.; Najafi, A.R.; West, B.L.; Green, K.N. Microglial repopulation resolves inflammation and promotes brain recovery after injury. Glia 2017, 65, 931–944. [Google Scholar] [CrossRef]
- Lazaridis, I.; Charalampopoulos, I.; Alexaki, V.I.; Avlonitis, N.; Pediaditakis, I.; Efstathopoulos, P.; Calogeropoulou, T.; Castanas, E.; Gravanis, A. Neurosteroid dehydroepiandrosterone interacts with nerve growth factor (NGF) receptors, preventing neuronal apoptosis. PLoS Biol. 2011, 9, e1001051. [Google Scholar] [CrossRef]
- Pediaditakis, I.; Efstathopoulos, P.; Prousis, K.C.; Zervou, M.; Arevalo, J.C.; Alexaki, V.I.; Nikoletopoulou, V.; Karagianni, E.; Potamitis, C.; Tavernarakis, N.; et al. Selective and differential interactions of BNN27, a novel C17-spiroepoxy steroid derivative, with TrkA receptors, regulating neuronal survival and differentiation. Neuropharmacology 2016, 111, 266–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ziogas, A.; Maekawa, T.; Wiessner, J.R.; Le, T.T.; Sprott, D.; Troullinaki, M.; Neuwirth, A.; Anastasopoulou, V.; Grossklaus, S.; Chung, K.J.; et al. DHEA inhibits leukocyte recruitment through regulation of the integrin antagonist DEL-1. J. Immunol. 2020, 204, 1214–1224. [Google Scholar] [CrossRef]
- Zhao, N.; Francis, N.L.; Calvelli, H.R.; Moghe, P.V. Microglia-targeting nanotherapeutics for neurodegenerative diseases. APL Bioeng. 2020, 4, 030902. [Google Scholar] [CrossRef]
- Kohler, O.; Benros, M.E.; Nordentoft, M.; Farkouh, M.E.; Iyengar, R.L.; Mors, O.; Krogh, J. Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: A systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry 2014, 71, 1381–1391. [Google Scholar] [CrossRef] [PubMed]
- Raison, C.L.; Rutherford, R.E.; Woolwine, B.J.; Shuo, C.; Schettler, P.; Drake, D.F.; Haroon, E.; Miller, A.H. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: The role of baseline inflammatory biomarkers. JAMA Psychiatry 2013, 70, 31–41. [Google Scholar] [CrossRef]
- Kodama, L.; Gan, L. Do microglial sex differences contribute to sex differences in neurodegenerative diseases? Trends Mol. Med. 2019, 25, 741–749. [Google Scholar] [CrossRef]
- Mee-Inta, O.; Zhao, Z.W.; Kuo, Y.M. Physical exercise inhibits inflammation and microglial activation. Cells 2019, 8, 691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madore, C.; Yin, Z.; Leibowitz, J.; Butovsky, O. Microglia, Lifestyle stress, and neurodegeneration. Immunity 2020, 52, 222–240. [Google Scholar] [CrossRef] [PubMed]
Factor | Cause or Source | Effect on Microglia | References |
---|---|---|---|
TNF | Adipose tissue macrophages, adipocytes | Inflammation, reduced phagocytosis | [10,52,92,147,205,206] |
Leptin | Adipocytes | Pro- and anti-inflammatory effects, ramification | [10,147,207,208,209,210] |
Saturated fatty acids | Diet, adipose tissue | Inflammation, inflammasome activation, migration and phagocytosis | [196,200,211,212,213,214] |
Omega-6/omega-3 fatty acids | Diet | Increased inflammation, decreased phagocytosis and reduced resolution of inflammation | [215,216,217,218,219,220] |
Endotoxins | Gut microbiome | Inflammation | [221,222] |
SCFA | Gut microbiome | Proper microglial function and homeostasis | [190,223] |
Glucocorticoids | Adrenal glands | Anti- and pro-inflammatory effects | [191,192,193,224,225,226,227,228,229,230] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Alexaki, V.I. The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives. Cells 2021, 10, 1584. https://doi.org/10.3390/cells10071584
Alexaki VI. The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives. Cells. 2021; 10(7):1584. https://doi.org/10.3390/cells10071584
Chicago/Turabian StyleAlexaki, Vasileia Ismini. 2021. "The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives" Cells 10, no. 7: 1584. https://doi.org/10.3390/cells10071584
APA StyleAlexaki, V. I. (2021). The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives. Cells, 10(7), 1584. https://doi.org/10.3390/cells10071584