Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies
Abstract
:1. Introduction
2. Microglia as a Main Target of Maternal Obesity-Derived Inflammation
3. Obesity, Maternal Microbiota Dysbiosis and Its Sequelae on the Neurodevelopment of the Offspring
3.1. Role of the Microbiota in Human Health: The Forgotten Organ
3.2. Microbiota Dysbiosis in Maternal Obesity
4. Nutritional Strategies to Counteract Maternal Obesity
4.1. LC-PUFA Supplementation as Anti-Inflammatory and Protective Strategy
4.2. Milk Fat Globule Membrane
4.3. The Antioxidant N-Acetyl-Cysteine as an Anti-Inflammatory and Protective Strategy in the Maternal-Fetal Crosstalk
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
AA | Arachidonic Acid |
ARC | Arcuate Nucleus |
BMI | Body Mass Index |
CNS | Central Nervous System |
CD11b | cluster of differentiation 11b |
CA1, 2, 3 | Cornu Ammonis 1, 2, 3 |
CORT | Corticosterone |
CX3CR1 | CX3C chemokine receptor 1 |
CRHR2 | corticosteroid releasing hormone receptor 2 |
DHA | docosahexaenoic acid |
DOHaD | Developmental Origin of Health and Disease |
EPA | Eicosapentaenoic Acid |
ERK1/2 | Extracellular Signal-Regulated Kinases 1/2 |
Fas | Fatty Acids |
GF | Germ Free |
GPR 41, 43, 120 | G-protein-coupled receptors 41,43, 120 |
GSH | Glutathione |
HFD | High Fat Diet |
Iba1 | Ionized calcium-binding adaptor molecule 1 |
IFN-gamma | Interferon-gamma |
IL-1beta | Interleukin -1beta, 6, 10 |
JAK/STAT3 | Janus kinases/Signal Transducer and Activator of Transcription Proteins 3 |
KAT1 | kynurenine aminotransferase 1 |
KMO | kynurenine mono oxygenase |
LA | Linoleic Acid |
LC-PUFAs | Long Chain Polyunsaturated Fatty Acids |
LPS | Lipopolysaccharides |
MCP-1 | Monocyte chemoattractant protein-1 |
MBP | Myelin Basic Protein |
MFGM | Milk Fat Globule Membrane |
NAC | N-Acetyl-Cysteine |
NF-kB | Nuclear Factor kappa-light-chain-enhancer of activated B cells |
NLRP3 | NLR family pyrin domain containing 3 |
OS | Oxidative Stress |
PPAR-gamma | Peroxisome Proliferator-activated Receptor-gamma |
ROS | Reactive Oxygen Species |
RXR | Retinoid X Receptors |
SCFAs | Short Chain Fatty Acids |
SON | Supraoptic Nucleus |
TGF-beta 1 | Transforming growth factor-beta 1 |
TLRs | Toll-like Receptors |
Tmem119 | Transmembrane Protein 119 |
TNF-alpha | Tumor Necrosis Factor alpha |
TREM2 | Triggering receptor expressed on myeloid cells 2 |
VEGF | Vascular Endothelial Growth Factor |
References
- Barker, D.J.P.; Osmond, C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and wales. Lancet 1986, 327, 1077–1081. [Google Scholar] [CrossRef]
- Spencer, S.J.; Korosi, A.; Layé, S.; Shukitt-Hale, B.; Barrientos, R.M. Food for thought: How nutrition impacts cognition and emotion. NPJ Sci. Food 2017, 1, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reemst, K.; Tims, S.; Yam, K.-Y.; Mischke, M.; Knol, J.; Brul, S.; Schipper, L.; Korosi, A. The Role of the Gut Microbiota in the Effects of Early-Life Stress and Dietary Fatty Acids on Later-Life Central and Metabolic Outcomes in Mice. mSystems 2022, 7, e00180-22. [Google Scholar] [CrossRef] [PubMed]
- Gomes, D.; Le, L.; Perschbacher, S.; Haas, N.A.; Netz, H.; Hasbargen, U.; Delius, M.; Lange, K.; Nennstiel, U.; Roscher, A.A.; et al. Predicting the earliest deviation in weight gain in the course towards manifest overweight in offspring exposed to obesity in pregnancy: A longitudinal cohort study. BMC Med. 2022, 20, 1–18. [Google Scholar] [CrossRef]
- Kereliuk, S.M.; Dolinsky, V.W. Recent Experimental Studies of Maternal Obesity, Diabetes during Pregnancy and the Developmental Origins of Cardiovascular Disease. Int. J. Mol. Sci. 2022, 23, 4467. [Google Scholar] [CrossRef]
- Cirulli, F.; Musillo, C.; Berry, A. Maternal Obesity as a Risk Factor for Brain Development and Mental Health in the Offspring. Neuroscience 2020, 447, 122–135. [Google Scholar] [CrossRef]
- Congdon, P. Obesity and Urban Environments. Int. J. Environ. Res. Public Health 2019, 16, 464. [Google Scholar] [CrossRef] [Green Version]
- Musillo, C.; Berry, A.; Cirulli, F. Prenatal psychological or metabolic stress increases the risk for psychiatric disorders: The “funnel effect” model. Neurosci. Biobehav. Rev. 2022, 136, 104624. [Google Scholar] [CrossRef]
- Sullivan, E.L.; Nousen, E.K.; Chamlou, K.A. Maternal high fat diet consumption during the perinatal period programs offspring behavior. Physiol. Behav. 2012, 123, 236–242. [Google Scholar] [CrossRef] [Green Version]
- Rivera, H.M.; Christiansen, K.J.; Sullivan, E.L. The role of maternal obesity in the risk of neuropsychiatric disorders. Front. Neurosci. 2015, 9, 194. [Google Scholar] [CrossRef]
- Bellisario, V.; Panetta, P.; Balsevich, G.; Baumann, V.; Noble, J.; Raggi, C.; Nathan, O.; Berry, A.; Seckl, J.; Schmidt, M.; et al. Maternal high-fat diet acts as a stressor increasing maternal glucocorticoids’ signaling to the fetus and disrupting maternal behavior and brain activation in C57BL/6J mice. Psychoneuroendocrinology 2015, 60, 138–150. [Google Scholar] [CrossRef]
- Iozzo, P.; Holmes, M.; Schmidt, M.V.; Cirulli, F.; Guzzardi, M.A.; Berry, A.; Balsevich, G.; Andreassi, M.G.; Wesselink, J.-J.; Liistro, T.; et al. Developmental ORIgins of Healthy and Unhealthy AgeiNg: The Role of Maternal Obesity-Introduction to DORIAN. Obes. Facts 2014, 7, 130–151. [Google Scholar] [CrossRef]
- Davis, J.; Mire, E. Maternal obesity and developmental programming of neuropsychiatric disorders: An inflammatory hypothesis. Brain Neurosci. Adv. 2021, 5, 23982128211003484. [Google Scholar] [CrossRef]
- Berry, A.; Mazzelli, M.; Musillo, C.; Riva, M.A.; Cattaneo, A.; Cirulli, F. High-fat diet during adulthood interacts with prenatal stress, affecting both brain inflammatory and neuroendocrine markers in male rats. Eur. J. Neurosci. 2021, 55, 2326–2340. [Google Scholar] [CrossRef]
- 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] [Green Version]
- 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-1β Modulate Synaptic Plasticity during Neuroinflammation. Neural Plast. 2018, 2018, 8430123. [Google Scholar] [CrossRef] [Green Version]
- Sun, K.; Kusminski, C.M.; Scherer, P.E. Adipose tissue remodeling and obesity. J. Clin. Investig. 2011, 121, 2094–2101. [Google Scholar] [CrossRef] [Green Version]
- Longo, M.; Zatterale, F.; Naderi, J.; Parrillo, L.; Formisano, P.; Raciti, G.A.; Beguinot, F.; Miele, C. Adipose Tissue Dysfunction as Determinant of Obesity-Associated Metabolic Complications. Int. J. Mol. Sci. 2019, 20, 2358. [Google Scholar] [CrossRef] [Green Version]
- Fuster, J.J.; Ouchi, N.; Gokce, N.; Walsh, K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ. Res. 2016, 118, 1786–1807. [Google Scholar] [CrossRef] [Green Version]
- Challier, J.; Basu, S.; Bintein, T.; Minium, J.; Hotmire, K.; Catalano, P.; Mouzon, S.H.-D. Obesity in Pregnancy Stimulates Macrophage Accumulation and Inflammation in the Placenta. Placenta 2008, 29, 274–281. [Google Scholar] [CrossRef] [Green Version]
- Basu, S.; Haghiac, M.; Surace, P.; Challier, J.-C.; Guerre-Millo, M.; Singh, K.; Waters, T.; Minium, J.; Presley, L.; Catalano, P.M.; et al. Pregravid Obesity Associates with Increased Maternal Endotoxemia and Metabolic Inflammation. Obesity 2011, 19, 476–482. [Google Scholar] [CrossRef] [Green Version]
- Parisi, F.; Milazzo, R.; Savasi, V.; Cetin, I. Maternal Low-Grade Chronic Inflammation and Intrauterine Programming of Health and Disease. Int. J. Mol. Sci. 2021, 22, 1732. [Google Scholar] [CrossRef]
- Edwards, S.M.; Cunningham, S.A.; Dunlop, A.L.; Corwin, E.J. The Maternal Gut Microbiome During Pregnancy. MCN Am. J. Matern. Nurs. 2017, 42, 310–317. [Google Scholar] [CrossRef]
- Bolton, J.L.; Bilbo, S.D. Developmental programming of brain and behavior by perinatal diet: Focus on inflammatory mechanisms. Dialog-Clin. Neurosci. 2014, 16, 307–320. [Google Scholar] [CrossRef]
- Segovia, S.A.; Vickers, M.H.; Gray, C.; Reynolds, C.M. Maternal obesity, inflammation, and developmental programming. Biomed Res. Int. 2014, 2014. [Google Scholar] [CrossRef]
- Salans, L.B.; Cushman, S.W.; Weismann, R.E. Studies of Human Adipose Tissue adipose cell size and number in nonobese and obese patients. J. Clin. Investig. 1973, 52, 929–941. [Google Scholar] [CrossRef] [Green Version]
- Aye, I.L.; Lager, S.; Ramirez, V.I.; Gaccioli, F.; Dudley, D.J.; Jansson, T.; Powell, T. Increasing Maternal Body Mass Index Is Associated with Systemic Inflammation in the Mother and the Activation of Distinct Placental Inflammatory Pathways1. Biol. Reprod. 2014, 90, 129. [Google Scholar] [CrossRef] [PubMed]
- Edlow, A.G.; Glass, R.M.; Smith, C.J.; Tran, P.K.; James, K.; Bilbo, S. Placental Macrophages: A Window Into Fetal Microglial Function in Maternal Obesity. Int. J. Dev. Neurosci. 2018, 77, 60–68. [Google Scholar] [CrossRef] [PubMed]
- Melekoğlu, R.; Çiftçi, O.; Eraslan, S.; Başak, N.; Celik, E. The Effects of Body Mass Index on Second-Trimester Amniotic Fluid Cytokine and Matrix Metalloproteinase Levels. Gynecol. Obstet. Investig. 2017, 83, 70–75. [Google Scholar] [CrossRef] [PubMed]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
- Álvarez, D.; Muñoz, Y.; Ortiz, M.; Maliqueo, M.; Chouinard-Watkins, R.; Valenzuela, R. Impact of Maternal Obesity on the Metabolism and Bioavailability of Polyunsaturated Fatty Acids during Pregnancy and Breastfeeding. Nutrients 2020, 13, 19. [Google Scholar] [CrossRef]
- Al Bander, Z.; Nitert, M.D.; Mousa, A.; Naderpoor, N. The Gut Microbiota and Inflammation: An Overview. Int. J. Environ. Res. Public Health 2020, 17, 7618. [Google Scholar] [CrossRef]
- Agustí, A.; García-Pardo, M.P.; López-Almela, I.; Campillo, I.; Maes, M.; Romani-Pérez, M.; Sanz, Y. Interplay Between the Gut-Brain Axis, Obesity and Cognitive Function. Front. Neurosci. 2018, 12, 155. [Google Scholar] [CrossRef]
- Carlessi, A.S.; Borba, L.A.; Zugno, A.I.; Quevedo, J.; Réus, G.Z. Gut microbiota–brain axis in depression: The role of neuroinflammation. Eur. J. Neurosci. 2019, 53, 222–235. [Google Scholar] [CrossRef]
- Cirulli, F. Interactions between early life stress and metabolic stress in programming of mental and metabolic health. Curr. Opin. Behav. Sci. 2017, 14, 65–71. [Google Scholar] [CrossRef]
- Gohir, W.; Ratcliffe, E.M.; Sloboda, D.M. Of the bugs that shape us: Maternal obesity, the gut microbiome, and long-term disease risk. Pediatr. Res. 2014, 77, 196–204. [Google Scholar] [CrossRef]
- Bardos, J.; Fiorentino, D.; Longman, R.E.; Paidas, M. Immunological Role of the Maternal Uterine Microbiome in Pregnancy: Pregnancies Pathologies and Alterated Microbiota. Front. Immunol. 2020, 10, 2823. [Google Scholar] [CrossRef]
- Lenz, K.M.; Nelson, L. Microglia and Beyond: Innate Immune Cells As Regulators of Brain Development and Behavioral Function. Front. Immunol. 2018, 9, 698. [Google Scholar] [CrossRef] [Green Version]
- Hammond, B.P.; Manek, R.; Kerr, B.J.; Macauley, M.S.; Plemel, J.R. Regulation of microglia population dynamics throughout development, health, and disease. Glia 2021, 69, 2771–2797. [Google Scholar] [CrossRef]
- Bilbo, S.D.; Tsang, V. Enduring consequences of maternal obesity for brain inflammation and behavior of offspring. FASEB J. 2010, 24, 2104–2115. [Google Scholar] [CrossRef]
- Bordeleau, M.; de Cossío, L.F.; Chakravarty, M.M.; Tremblay, M. From Maternal Diet to Neurodevelopmental Disorders: A Story of Neuroinflammation. Front. Cell. Neurosci. 2021, 14, 612705. [Google Scholar] [CrossRef]
- Bordeleau, M.; Lacabanne, C.; de Cossío, L.F.; Vernoux, N.; Savage, J.C.; González-Ibáñez, F.; Tremblay, M. Microglial and peripheral immune priming is partially sexually dimorphic in adolescent mouse offspring exposed to maternal high-fat diet. J. Neuroinflamm. 2020, 17, 1–28. [Google Scholar] [CrossRef]
- Butovsky, O.; Weiner, H.L. Microglial signatures and their role in health and disease. Nat. Rev. Neurosci. 2018, 19, 622–635. [Google Scholar] [CrossRef]
- Zhang, X.; Kracht, L.; Lerario, A.M.; Dubbelaar, M.L.; Brouwer, N.; Wesseling, E.M.; Boddeke, E.W.G.M.; Eggen, B.J.L.; Kooistra, S.M. Epigenetic regulation of innate immune memory in microglia. J. Neuroinflamm. 2022, 19, 1–19. [Google Scholar] [CrossRef]
- Schaafsma, W.; Basterra, L.B.; Jacobs, S.; Brouwer, N.; Meerlo, P.; Schaafsma, A.; Boddeke, E.W.; Eggen, B.J. Maternal inflammation induces immune activation of fetal microglia and leads to disrupted microglia immune responses, behavior, and learning performance in adulthood. Neurobiol. Dis. 2017, 106, 291–300. [Google Scholar] [CrossRef]
- Thion, M.S.; Low, D.; Silvin, A.; Chen, J.; Grisel, P.; Schulte-Schrepping, J.; Blecher, R.; Ulas, T.; Squarzoni, P.; Hoeffel, G.; et al. Microbiome Influences Prenatal and Adult Microglia in a Sex-Specific Manner. Cell 2017, 172, 500–516.e16. [Google Scholar] [CrossRef] [Green Version]
- Williamson, L.L.; Sholar, P.W.; Mistry, R.S.; Smith, S.H.; Bilbo, S.D. Microglia and Memory: Modulation by Early-Life Infection. J. Neurosci. 2011, 31, 15511–15521. [Google Scholar] [CrossRef]
- Han, V.X.; Patel, S.; Jones, H.F.; Dale, R.C. Maternal immune activation and neuroinflammation in human neurodevelopmental disorders. Nat. Rev. Neurol. 2021, 17, 564–579. [Google Scholar] [CrossRef] [PubMed]
- Hunter, S.K.; Hoffman, M.C.; D’Alessandro, A.; Noonan, K.; Wyrwa, A.; Freedman, R.; Law, A.J. Male fetus susceptibility to maternal inflammation: C-reactive protein and brain development. Psychol. Med. 2019, 51, 450–459. [Google Scholar] [CrossRef] [PubMed]
- Monaco-Brown, M.; Lawrence, D.A. Obesity and Maternal-Placental-Fetal Immunology and Health. Front. Pediatr. 2022, 10, 859885. [Google Scholar] [CrossRef] [PubMed]
- Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [Green Version]
- Rezaie, P.; Male, D. Colonisation of the developing human brain and spinal cord by microglia: A review. Microsc. Res. Tech. 1999, 45, 359–382. [Google Scholar] [CrossRef]
- Neher, J.J.; Cunningham, C. Priming Microglia for Innate Immune Memory in the Brain. Trends Immunol. 2019, 40, 358–374. [Google Scholar] [CrossRef]
- Kang, S.S.; Kurti, A.; Fair, D.A.; Fryer, J.D. Dietary intervention rescues maternal obesity induced behavior deficits and neuroinflammation in offspring. J. Neuroinflamm. 2014, 11, 156. [Google Scholar] [CrossRef]
- Sasaki, A.; de Vega, W.; St-Cyr, S.; Pan, P.; McGowan, P. Perinatal high fat diet alters glucocorticoid signaling and anxiety behavior in adulthood. Neuroscience 2013, 240, 1–12. [Google Scholar] [CrossRef]
- Sasaki, A.; de Vega, W.; Sivanathan, S.; St-Cyr, S.; McGowan, P. Maternal high-fat diet alters anxiety behavior and glucocorticoid signaling in adolescent offspring. Neuroscience 2014, 272, 92–101. [Google Scholar] [CrossRef]
- Winther, G.; Elfving, B.; Müller, H.K.; Lund, S.; Wegener, G. Maternal High-fat Diet Programs Offspring Emotional Behavior in Adulthood. Neuroscience 2018, 388, 87–101. [Google Scholar] [CrossRef] [PubMed]
- Wijenayake, S.; Rahman, M.F.; Lum, C.M.W.; De Vega, W.C.; Sasaki, A.; McGowan, P.O. Maternal high-fat diet induces sex-specific changes to glucocorticoid and inflammatory signaling in response to corticosterone and lipopolysaccharide challenge in adult rat offspring. J. Neuroinflamm. 2020, 17, 1–16. [Google Scholar] [CrossRef] [Green Version]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [Green Version]
- Rocha-Gomes, A.; Teixeira, A.E.; de Oliveira, D.G.; Santiago, C.M.O.; da Silva, A.A.; Riul, T.R.; Lacerda, A.C.R.; Mendonça, V.A.; Rocha-Vieira, E.; Leite, H.R. LPS tolerance prevents anxiety-like behavior and amygdala inflammation of high-fat-fed dams’ adolescent offspring. Behav. Brain Res. 2021, 411, 113371. [Google Scholar] [CrossRef]
- Huang, C.-F.; Du, J.-X.; Deng, W.; Cheng, X.-C.; Zhang, S.-Y.; Zhao, S.-J.; Tao, M.-J.; Chen, G.-Z.; Hao, X.-Q. Effect of prenatal exposure to LPS combined with pre- and post-natal high-fat diet on hippocampus in rat offspring. Neuroscience 2015, 286, 364–370. [Google Scholar] [CrossRef] [PubMed]
- Ajmone-Cat, M.A.; D’Urso, M.C.; di Blasio, G.; Brignone, M.S.; De Simone, R.; Minghetti, L. Glycogen synthase kinase 3 is part of the molecular machinery regulating the adaptive response to LPS stimulation in microglial cells. Brain Behav. Immun. 2016, 55, 225–235. [Google Scholar] [CrossRef] [PubMed]
- Graf, A.E.; Lallier, S.W.; Waidyaratne, G.; Thompson, M.D.; Tipple, T.E.; Hester, M.E.; Trask, A.J.; Rogers, L.K. Maternal high fat diet exposure is associated with increased hepcidin levels, decreased myelination, and neurobehavioral changes in male offspring. Brain, Behav. Immun. 2016, 58, 369–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bordeleau, M.; de Cossío, L.F.; Lacabanne, C.; Savage, J.C.; Vernoux, N.; Chakravarty, M.; Tremblay, M. Maternal high-fat diet modifies myelin organization, microglial interactions, and results in social memory and sensorimotor gating deficits in adolescent mouse offspring. Brain Behav. Immun. Health 2021, 15, 100281. [Google Scholar] [CrossRef]
- Hasebe, K.; Kendig, M.D.; Morris, M.J. Mechanisms Underlying the Cognitive and Behavioural Effects of Maternal Obesity. Nutrients 2021, 13, 240. [Google Scholar] [CrossRef]
- Maldonado-Ruiz, R.; Garza-Ocañas, L.; Camacho, A. Inflammatory domains modulate autism spectrum disorder susceptibility during maternal nutritional programming. Neurochem. Int. 2019, 126, 109–117. [Google Scholar] [CrossRef]
- Kim, D.W.; Glendining, K.A.; Grattan, D.R.; Jasoni, C.L.; Grattan, D. Maternal obesity leads to increased proliferation and numbers of astrocytes in the developing fetal and neonatal mouse hypothalamus. Int. J. Dev. Neurosci. 2016, 53, 18–25. [Google Scholar] [CrossRef]
- Hughes, H.K.; Rose, D.; Ashwood, P. The Gut Microbiota and Dysbiosis in Autism Spectrum Disorders. Curr. Neurol. Neurosci. Rep. 2018, 18, 81. [Google Scholar] [CrossRef]
- Yu, L.W.; Agirman, G.; Hsiao, E.Y. The Gut Microbiome as a Regulator of the Neuroimmune Landscape. Annu. Rev. Immunol. 2022, 40, 143–167. [Google Scholar] [CrossRef]
- Angelucci, F.; Cechova, K.; Amlerova, J.; Hort, J. Antibiotics, gut microbiota, and Alzheimer’s disease. J. Neuroinflamm. 2019, 16, 1–10. [Google Scholar] [CrossRef]
- O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 2006, 7, 688–693. [Google Scholar] [CrossRef] [Green Version]
- Vuong, H.E.; Pronovost, G.N.; Williams, D.W.; Coley, E.J.L.; Siegler, E.L.; Qiu, A.; Kazantsev, M.; Wilson, C.J.; Rendon, T.; Hsiao, E.Y. The maternal microbiome modulates fetal neurodevelopment in mice. Nature 2020, 586, 281–286. [Google Scholar] [CrossRef]
- Codagnone, M.G.; Spichak, S.; O’Mahony, S.M.; O’Leary, O.F.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Programming Bugs: Microbiota and the Developmental Origins of Brain Health and Disease. Biol. Psychiatry 2018, 85, 150–163. [Google Scholar] [CrossRef]
- Daliry, A.; Pereira, E.N.G.D.S. Role of Maternal Microbiota and Nutrition in Early-Life Neurodevelopmental Disorders. Nutrients 2021, 13, 3533. [Google Scholar] [CrossRef]
- Aatsinki, A.-K.; Uusitupa, H.-M.; Munukka, E.; Pesonen, H.; Rintala, A.; Pietilä, S.; Lahti, L.; Eerola, E.; Karlsson, L.; Karlsson, H. Gut Microbiota Composition in Mid-Pregnancy Is Associated with Gestational Weight Gain but Not Prepregnancy Body Mass Index. J. Women’s Health 2018, 27, 1293–1301. [Google Scholar] [CrossRef]
- Turjeman, S.; Collado, M.C.; Koren, O. The gut microbiome in pregnancy and pregnancy complications. Curr. Opin. Endocr. Metab. Res. 2021, 18, 133–138. [Google Scholar] [CrossRef]
- D’Argenio, V. The Prenatal Microbiome: A New Player for Human Health. High-Throughput 2018, 7, 38. [Google Scholar] [CrossRef] [Green Version]
- Miko, E.; Csaszar, A.; Bodis, J.; Kovacs, K. The Maternal–Fetal Gut Microbiota Axis: Physiological Changes, Dietary Influence, and Modulation Possibilities. Life 2022, 12, 424. [Google Scholar] [CrossRef]
- Bodden, C.; Hannan, A.J.; Reichelt, A.C. Of ‘junk food’ and ‘brain food’: How parental diet influences offspring neurobiology and behaviour. Trends Endocrinol. Metab. 2021, 32, 566–578. [Google Scholar] [CrossRef]
- Buffington, S.A.; Di Prisco, G.V.; Auchtung, T.A.; Ajami, N.J.; Petrosino, J.F.; Costa-Mattioli, M. Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell 2016, 165, 1762–1775. [Google Scholar] [CrossRef] [Green Version]
- Alsharairi, N.A. The Role of Short-Chain Fatty Acids in the Interplay between a Very Low-Calorie Ketogenic Diet and the Infant Gut Microbiota and Its Therapeutic Implications for Reducing Asthma. Int. J. Mol. Sci. 2020, 21, 9580. [Google Scholar] [CrossRef]
- Alsharairi, N.A. The Role of Short-Chain Fatty Acids in Mediating Very Low-Calorie Ketogenic Diet-Infant Gut Microbiota Relationships and Its Therapeutic Potential in Obesity. Nutrients 2021, 13, 3702. [Google Scholar] [CrossRef]
- Al Rubaye, H.; Adamson, C.C.; Jadavji, N.M. The role of maternal diet on offspring gut microbiota development: A review. J. Neurosci. Res. 2020, 99, 284–293. [Google Scholar] [CrossRef]
- Ferguson, J. Maternal microbial molecules affect offspring health. Science 2020, 367, 978–979. [Google Scholar] [CrossRef]
- Kimura, I.; Miyamoto, J.; Ohue-Kitano, R.; Watanabe, K.; Yamada, T.; Onuki, M.; Aoki, R.; Isobe, Y.; Kashihara, D.; Inoue, D.; et al. Maternal gut microbiota in pregnancy influences offspring metabolic phenotype in mice. Science 2020, 367, eaaw8429. [Google Scholar] [CrossRef]
- Cowan, M.; Petri, W.A.J. Microglia: Immune Regulators of Neurodevelopment. Front. Immunol. 2018, 9, 2576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abdel-Haq, R.; Schlachetzki, J.C.; Glass, C.K.; Mazmanian, S.K. Microbiome–microglia connections via the gut–brain axis. J. Exp. Med. 2018, 216, 41–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castillo-Ruiz, A.; Mosley, M.; George, A.; Mussaji, L.F.; Fullerton, E.F.; Ruszkowski, E.M.; Jacobs, A.J.; Gewirtz, A.T.; Chassaing, B.; Forger, N.G. The microbiota influences cell death and microglial colonization in the perinatal mouse brain. Brain Behav. Immun. 2017, 67, 218–229. [Google Scholar] [CrossRef] [PubMed]
- Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tenenbaum-Gavish, K.; Hod, M. Impact of Maternal Obesity on Fetal Health. Fetal Diagn. Ther. 2013, 34, 1–7. [Google Scholar] [CrossRef]
- Jeyabalan, A. Epidemiology of preeclampsia: Impact of obesity. Nutr. Rev. 2013, 71, S18–S25. [Google Scholar] [CrossRef] [Green Version]
- Berry, A.; Cirulli, F. High-Fat Diet and Foetal Programming: Use of P66Shc Knockouts and Implications for Human Kind. In Diet, Nutrition, and Fetal Programming; Humana Press: Cham, Switzerland, 2017; pp. 557–568. [Google Scholar] [CrossRef]
- Thangaratinam, S.; Rogozinska, E.; Jolly, K.; Glinkowski, S.; Roseboom, T.; Tomlinson, J.W.; Kunz, R.; Mol, B.W.; Coomarasamy, A.; Khan, K.S. Effects of interventions in pregnancy on maternal weight and obstetric outcomes: Meta-analysis of randomised evidence. BMJ 2012, 344, e2088. [Google Scholar] [CrossRef] [Green Version]
- Bruno, R.; Petrella, E.; Bertarini, V.; Pedrielli, G.; Neri, I.; Facchinetti, F. Adherence to a lifestyle programme in overweight/obese pregnant women and effect on gestational diabetes mellitus: A randomized controlled trial. Matern. Child Nutr. 2016, 13, e12333. [Google Scholar] [CrossRef] [Green Version]
- Shrestha, A.; Prowak, M.; Berlandi-Short, V.-M.; Garay, J.; Ramalingam, L. Maternal Obesity: A Focus on Maternal Interventions to Improve Health of Offspring. Front. Cardiovasc. Med. 2021, 8, 696812. [Google Scholar] [CrossRef]
- Vidakovic, A.J.; Jaddoe, V.W.V.; Gishti, O.; Felix, J.F.; Williams, M.A.; Hofman, A.; Demmelmair, H.; Koletzko, B.; Tiemeier, H.; Gaillard, R. Body mass index, gestational weight gain and fatty acid concentrations during pregnancy: The Generation R Study. Eur. J. Epidemiol. 2015, 30, 1175–1185. [Google Scholar] [CrossRef] [Green Version]
- Benaim, C.; Freitas-Vilela, A.A.; Pinto, T.D.J.P.; Lepsch, J.; Farias, D.R.; Dos Santos Vaz, J.; El-Bacha, T.; Kac, G. Early pregnancy body mass index modifies the association of pre-pregnancy dietary patterns with serum polyunsaturated fatty acid concentrations throughout pregnancy in Brazilian women. Matern. Child Nutr. 2018, 14, e12480. [Google Scholar] [CrossRef] [Green Version]
- Joffre, C.; Grégoire, S.; De Smedt, V.; Acar, N.; Bretillon, L.; Nadjar, A.; Layé, S. Modulation of brain PUFA content in different experimental models of mice. Prostaglandins Leukot. Essent. Fat. Acids 2016, 114, 1–10. [Google Scholar] [CrossRef]
- Sakayori, N.; Kikkawa, T.; Tokuda, H.; Kiryu, E.; Yoshizaki, K.; Kawashima, H.; Yamada, T.; Arai, H.; Kang, J.X.; Katagiri, H.; et al. Maternal dietary imbalance between omega-6 and omega-3 polyunsaturated fatty acids impairs neocortical development via epoxy metabolites. Stem Cells 2015, 34, 470–482. [Google Scholar] [CrossRef] [Green Version]
- Powell, T.L.; Barner, K.; Madi, L.; Armstrong, M.; Manke, J.; Uhlson, C.; Jansson, T.; Ferchaud-Roucher, V. Sex-specific responses in placental fatty acid oxidation, esterification and transfer capacity to maternal obesity. Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2020, 1866, 158861. [Google Scholar] [CrossRef]
- Mao, J.; Zhang, X.; Sieli, P.T.; Falduto, M.T.; Torres, K.E.; Rosenfeld, C.S. Contrasting effects of different maternal diets on sexually dimorphic gene expression in the murine placenta. Proc. Natl. Acad. Sci. USA 2010, 107, 5557–5562. [Google Scholar] [CrossRef] [Green Version]
- Penfield-Cyr, A.; Monthe-Dreze, C.; Smid, M.C.; Sen, S. Maternal BMI, Mid-pregnancy Fatty Acid Concentrations, and Perinatal Outcomes. Clin. Ther. 2018, 40, 1659–1667.e1. [Google Scholar] [CrossRef] [Green Version]
- Tylek, K.; Trojan, E.; Leśkiewicz, M.; Regulska, M.; Bryniarska, N.; Curzytek, K.; Lacivita, E.; Leopoldo, M.; Basta-Kaim, A. Time-Dependent Protective and Pro-Resolving Effects of FPR2 Agonists on Lipopolysaccharide-Exposed Microglia Cells Involve Inhibition of NF-κB and MAPKs Pathways. Cells 2021, 10, 2373. [Google Scholar] [CrossRef]
- Gorica, E.; Calderone, V. Arachidonic Acid Derivatives and Neuroinflammation. CNS Neurol. Disord.-Drug Targets 2022, 21, 118–129. [Google Scholar] [CrossRef]
- Wang, B.; Wu, L.; Chen, J.; Dong, L.; Chen, C.; Wen, Z.; Hu, J.; Fleming, I.; Wang, D.W. Metabolism pathways of arachidonic acids: Mechanisms and potential therapeutic targets. Signal Transduct. Target. Ther. 2021, 6, 1–30. [Google Scholar] [CrossRef] [PubMed]
- Ahme, O.S.; Galano, J.M.; Pavlickova, T.; Revol-Cavalier, J.; Vigor, C.; Lee, J.C.Y.; Oger, C.; Durand, T. Moving forward with isoprostanes, neuroprostanes and phytoprostanes: Where are we now? Essays Biochem. 2020, 64, 463–484. [Google Scholar]
- Belcastro, L.; Ferreira, C.S.; Saraiva, M.A.; Mucci, D.B.; Murgia, A.; Lai, C.; Vigor, C.; Oger, C.; Galano, J.-M.; Pinto, G.D.A.; et al. Decreased Fatty Acid Transporter FABP1 and Increased Isoprostanes and Neuroprostanes in the Human Term Placenta: Implications for Inflammation and Birth Weight in Maternal Pre-Gestational Obesity. Nutrients 2021, 13, 2768. [Google Scholar] [CrossRef] [PubMed]
- Heerwagen, M.J.R.; Stewart, M.S.; De La Houssaye, B.A.; Janssen, R.C.; Friedman, J.E. Transgenic Increase in N-3/N-6 Fatty Acid Ratio Reduces Maternal Obesity-Associated Inflammation and Limits Adverse Developmental Programming in Mice. PLoS ONE 2013, 8, e67791. [Google Scholar] [CrossRef]
- Alvarado, F.L.; Calabuig-Navarro, V.; Haghiac, M.; Puchowicz, M.; Tsai, P.-J.S.; O’Tierney-Ginn, P. Maternal obesity is not associated with placental lipid accumulation in women with high omega-3 fatty acid levels. Placenta 2018, 69, 96–101. [Google Scholar] [CrossRef]
- Lager, S.; Ramirez, V.I.; Acosta, O.; Meireles, C.; Miller, E.; Gaccioli, F.; Rosario, F.J.; Gelfond, J.A.L.; Hakala, K.; Weintraub, S.T.; et al. Docosahexaenoic Acid Supplementation in Pregnancy Modulates Placental Cellular Signaling and Nutrient Transport Capacity in Obese Women. J. Clin. Endocrinol. Metab. 2017, 102, 4557–4567. [Google Scholar] [CrossRef] [Green Version]
- Labrousse, V.; Leyrolle, Q.; Amadieu, C.; Aubert, A.; Sere, A.; Coutureau, E.; Grégoire, S.; Bretillon, L.; Pallet, V.; Gressens, P.; et al. Dietary omega-3 deficiency exacerbates inflammation and reveals spatial memory deficits in mice exposed to lipopolysaccharide during gestation. Brain Behav. Immun. 2018, 73, 427–440. [Google Scholar] [CrossRef] [Green Version]
- Madore, C.; Nadjar, A.; Delpech, J.-C.; Sere, A.; Aubert, A.; Portal, C.; Joffre, C.; Layé, S. Nutritional n-3 PUFAs deficiency during perinatal periods alters brain innate immune system and neuronal plasticity-associated genes. Brain Behav. Immun. 2014, 41, 22–31. [Google Scholar] [CrossRef]
- Rey, C.; Delpech, J.; Madore, C.; Nadjar, A.; Greenhalgh, A.; Amadieu, C.; Aubert, A.; Pallet, V.; Vaysse, C.; Layé, S.; et al. Dietary n-3 long chain PUFA supplementation promotes a pro-resolving oxylipin profile in the brain. Brain Behav. Immun. 2018, 76, 17–27. [Google Scholar] [CrossRef]
- Yang, B.; Yang, B.; Li, R.; Liu, P.N.; Geng, X.; Mooney, B.P.; Mooney, B.P.; Chen, C.; Cheng, J.; Fritsche, K.L.; et al. Quantitative Proteomics Reveals Docosahexaenoic Acid-Mediated Neuroprotective Effects in Lipopolysaccharide-Stimulated Microglial Cells. J. Proteome Res. 2020, 19, 2236–2246. [Google Scholar] [CrossRef]
- Layé, 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. 2017, 70, 12–38. [Google Scholar] [CrossRef]
- Fourrier, C.; Remus-Borel, J.; Greenhalgh, A.D.; Guichardant, M.; Bernoud-Hubac, N.; Lagarde, M.; Joffre, C.; Layé, S. Docosahexaenoic acid-containing choline phospholipid modulates LPS-induced neuroinflammation in vivo and in microglia in vitro. J. Neuroinflamm. 2017, 14, 1–13. [Google Scholar] [CrossRef]
- Hao, S.; Dey, A.; Yu, X.; Stranahan, A.M. Dietary obesity reversibly induces synaptic stripping by microglia and impairs hippocampal plasticity. Brain Behav. Immun. 2015, 51, 230–239. [Google Scholar] [CrossRef] [Green Version]
- Tremblay, M.-E.; Zhang, I.; Bisht, K.; Savage, J.C.; Lecours, C.; Parent, M.; Titorenko, V.; Maysinger, D. Remodeling of lipid bodies by docosahexaenoic acid in activated microglial cells. J. Neuroinflamm. 2016, 13, 1–18. [Google Scholar] [CrossRef] [Green Version]
- Chang, P.K.-Y.; Khatchadourian, A.; McKinney, R.A.; Maysinger, D. Docosahexaenoic acid (DHA): A modulator of microglia activity and dendritic spine morphology. J. Neuroinflamm. 2015, 12, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Ajmone-Cat, M.A.; Salvatori, M.L.; 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. 2011, 90, 575–587. [Google Scholar] [CrossRef]
- De Smedt-Peyrusse, V.; Sargueil, F.; Moranis, A.; Harizi, H.; Mongrand, S.; Layé, 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]
- Madore, C.; Leyrolle, Q.; Morel, L.; Rossitto, M.; Greenhalgh, A.D.; Delpech, J.C.; Martinat, M.; Bosch-Bouju, C.; Bourel, J.; Rani, B.; et al. Essential omega-3 fatty acids tune microglial phagocytosis of synaptic elements in the mouse developing brain. Nat. Commun. 2020, 11, 6133. [Google Scholar] [CrossRef]
- Basak, S.; Mallick, R.; Banerjee, A.; Pathak, S.; Duttaroy, A. Maternal Supply of Both Arachidonic and Docosahexaenoic Acids Is Required for Optimal Neurodevelopment. Nutrients 2021, 13, 2061. [Google Scholar] [CrossRef]
- Lauritzen, L.; Brambilla, P.; Mazzocchi, A.; Harsløf, L.B.S.; Ciappolino, V.; Agostoni, C. DHA Effects in Brain Development and Function. Nutrients 2016, 8, 6. [Google Scholar] [CrossRef] [Green Version]
- Hadley, K.B.; Ryan, A.S.; Forsyth, S.; Gautier, S.; Salem, N., Jr. The Essentiality of Arachidonic Acid in Infant Development. Nutrients 2016, 8, 216. [Google Scholar] [CrossRef] [Green Version]
- Swanson, D.; Block, R.; Mousa, S.A. Omega-3 Fatty Acids EPA and DHA: Health Benefits Throughout Life. Adv. Nutr. 2012, 3, 1–7. [Google Scholar] [CrossRef]
- Bernardo, A.; Giammarco, M.L.; De Nuccio, C.; Ajmone-Cat, M.A.; Visentin, S.; De Simone, R.; Minghetti, L. Docosahexaenoic acid promotes oligodendrocyte differentiation via PPAR-γ signalling and prevents tumor necrosis factor-α-dependent maturational arrest. Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2017, 1862, 1013–1023. [Google Scholar] [CrossRef]
- Belayev, L.; Hong, S.; Freitas, R.S.; Menghani, H.; Marcell, S.J.; Khoutorova, L.; Mukherjee, P.K.; Reid, M.M.; Oria, R.B.; Bazan, N.G. DHA modulates MANF and TREM2 abundance, enhances neurogenesis, reduces infarct size, and improves neurological function after experimental ischemic stroke. CNS Neurosci. Ther. 2020, 26, 1155–1167. [Google Scholar] [CrossRef]
- Darios, F.; Davletov, B. Omega-3 and omega-6 fatty acids stimulate cell membrane expansion by acting on syntaxin 3. Nature 2006, 440, 813–817. [Google Scholar] [CrossRef] [PubMed]
- Das, M.; Das, S. Docosahexaenoic Acid (DHA) Induced Morphological Differentiation of Astrocytes Is Associated with Transcriptional Upregulation and Endocytosis of β2-AR. Mol. Neurobiol. 2018, 56, 2685–2702. [Google Scholar] [CrossRef] [PubMed]
- Hamazaki, K.; Itomura, M.; Huan, M.; Nishizawa, H.; Sawazaki, S.; Tanouchi, M.; Watanabe, S.; Hamazaki, T.; Terasawa, K.; Yazawa, K. Effect of ω-3 fatty acid-containing phospholipids on blood catecholamine concentrations in healthy volunteers: A randomized, placebo-controlled, double-blind trial. Nutrition 2005, 21, 705–710. [Google Scholar] [CrossRef] [PubMed]
- Kawakita, E.; Hashimoto, M.; Shido, O. Docosahexaenoic acid promotes neurogenesis in vitro and in vivo. Neuroscience 2006, 139, 991–997. [Google Scholar] [CrossRef]
- Tokuda, H.; Kontani, M.; Kawashima, H.; Kiso, Y.; Shibata, H.; Osumi, N. Differential effect of arachidonic acid and docosahexaenoic acid on age-related decreases in hippocampal neurogenesis. Neurosci. Res. 2014, 88, 58–66. [Google Scholar] [CrossRef]
- Zhang, J.; Yi, C.; Han, J.; Ming, T.; Zhou, J.; Lu, C.; Li, Y.; Su, X. Novel high-docosahexaenoic-acid tuna oil supplementation modulates gut microbiota and alleviates obesity in high-fat diet mice. Food Sci. Nutr. 2020, 8, 6513–6527. [Google Scholar] [CrossRef]
- Noriega, B.S.; Sanchez-Gonzalez, M.A.; Salyakina, D.; Coffman, J. Understanding the Impact of Omega-3 Rich Diet on the Gut Microbiota. Case Rep. Med. 2016, 2016, 1–6. [Google Scholar] [CrossRef] [Green Version]
- Kaliannan, K.; Wang, B.; Li, X.-Y.; Kim, K.-J.; Kang, J.X. A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia. Sci. Rep. 2015, 5, 11276. [Google Scholar] [CrossRef] [Green Version]
- Yu, H.-N.; Zhu, J.; Pan, W.-S.; Shen, S.-R.; Shan, W.-G.; Das, U.N. Effects of Fish Oil with a High Content of n-3 Polyunsaturated Fatty Acids on Mouse Gut Microbiota. Arch. Med Res. 2014, 45, 195–202. [Google Scholar] [CrossRef]
- Yu, H.; Fang, C.; Li, P.; Wu, M.; Shen, S. The relevance of DHA with modulating of host-gut microbiome signatures alterations and repairing of lipids metabolism shifts. Eur. J. Pharmacol. 2021, 895, 173885. [Google Scholar] [CrossRef]
- Tang, Q.; Li, S.; Fang, C.; Yu, H. Evaluating the reparative effects and the mechanism of action of docosahexaenoic acid on azithromycin-induced lipid metabolism dysfunction. Food Chem. Toxicol. 2021, 159, 112699. [Google Scholar] [CrossRef]
- Vahdaninia, M.; Mackenzie, H.; Dean, T.; Helps, S. The effectiveness of ω-3 polyunsaturated fatty acid interventions during pregnancy on obesity measures in the offspring: An up-to-date systematic review and meta-analysis. Eur. J. Nutr. 2018, 58, 2597–2613. [Google Scholar] [CrossRef] [Green Version]
- Monthé-Drèze, C.; Sen, S.; Mouzon, S.H.-D.; Catalano, P.M. Effect of Omega-3 Supplementation in Pregnant Women with Obesity on Newborn Body Composition, Growth and Length of Gestation: A Randomized Controlled Pilot Study. Nutrients 2021, 13, 578. [Google Scholar] [CrossRef]
- Brink, L.R.; Lönnerdal, B. Milk fat globule membrane: The role of its various components in infant health and development. J. Nutr. Biochem. 2020, 85, 108465. [Google Scholar] [CrossRef]
- Kosmerl, E.; Rocha-Mendoza, D.; Ortega-Anaya, J.; Jiménez-Flores, R.; García-Cano, I. Improving Human Health with Milk Fat Globule Membrane, Lactic Acid Bacteria, and Bifidobacteria. Microorganisms 2021, 9, 341. [Google Scholar] [CrossRef]
- Mudd, A.T.; Alexander, L.S.; Berding, K.; Waworuntu, R.V.; Berg, B.M.; Donovan, S.M.; Dilger, R.N. Dietary Prebiotics, Milk Fat Globule Membrane, and Lactoferrin Affects Structural Neurodevelopment in the Young Piglet. Front. Pediatr. 2016, 4, 4. [Google Scholar] [CrossRef] [Green Version]
- Vickers, M.H.; Guan, J.; Gustavsson, M.; Krägeloh, C.U.; Breier, B.H.; Davison, M.; Fong, B.; Norris, C.; McJarrow, P.; Hodgkinson, S.C. Supplementation with a mixture of complex lipids derived from milk to growing rats results in improvements in parameters related to growth and cognition. Nutr. Res. 2009, 29, 426–435. [Google Scholar] [CrossRef]
- Moukarzel, S.; Dyer, R.A.; Garcia, C.; Wiedeman, A.M.; Boyce, G.; Weinberg, J.; Keller, B.O.; Elango, R.; Innis, S.M. Milk Fat Globule Membrane Supplementation in Formula-fed Rat Pups Improves Reflex Development and May Alter Brain Lipid Composition. Sci. Rep. 2018, 8, 1–9. [Google Scholar] [CrossRef] [Green Version]
- O’Mahony, S.M.; Neufeld, K.-A.M.; Waworuntu, R.V.; Pusceddu, M.M.; Manurung, S.; Murphy, K.; Strain, C.; Laguna, M.C.; Peterson, V.L.; Stanton, C.; et al. The enduring effects of early-life stress on the microbiota–gut–brain axis are buffered by dietary supplementation with milk fat globule membrane and a prebiotic blend. Eur. J. Neurosci. 2020, 51, 1042–1058. [Google Scholar] [CrossRef]
- Collins, J.M.; Caputi, V.; Manurung, S.; Gross, G.; Fitzgerald, P.; Golubeva, A.V.; Popov, J.; Deady, C.; Dinan, T.G.; Cryan, J.F.; et al. Supplementation with milk fat globule membrane from early life reduces maternal separation-induced visceral pain independent of enteric nervous system or intestinal permeability changes in the rat. Neuropharmacology 2022, 210, 109026. [Google Scholar] [CrossRef]
- Snow, D.R.; Ward, R.E.; Olsen, A.; Jimenez-Flores, R.; Hintze, K.J. Membrane-rich milk fat diet provides protection against gastrointestinal leakiness in mice treated with lipopolysaccharide. J. Dairy Sci. 2011, 94, 2201–2212. [Google Scholar] [CrossRef] [Green Version]
- Huang, S.; Wu, Z.; Liu, C.; Han, D.; Feng, C.; Wang, S.; Wang, J. Milk Fat Globule Membrane Supplementation Promotes Neonatal Growth and Alleviates Inflammation in Low-Birth-Weight Mice Treated with Lipopolysaccharide. BioMed Res. Int. 2019, 2019, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arnoldussen, I.A.C.; Morrison, M.C.; Wiesmann, M.; van Diepen, J.A.; Worms, N.; Voskuilen, M.; Verweij, V.; Geenen, B.; Gualdo, N.P.; van der Logt, L.; et al. Milk fat globule membrane attenuates high fat diet-induced neuropathological changes in obese Ldlr−/−.Leiden mice. Int. J. Obes. 2021, 46, 342–349. [Google Scholar] [CrossRef]
- Gustavsson, M.; Hodgkinson, S.C.; Fong, B.; Norris, C.; Guan, J.; Krageloh, C.U.; Breier, B.H.; Davison, M.; McJarrow, P.; Vickers, M.H. Maternal supplementation with a complex milk lipid mixture during pregnancy and lactation alters neonatal brain lipid composition but lacks effect on cognitive function in rats. Nutr. Res. 2010, 30, 279–289. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, M.; Henare, K.; Balakrishnan, B.; Lowe, E.; Fong, B.; McJarrow, P. Transfer of gangliosides across the human placenta. Placenta 2012, 33, 312–316. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.; Mo, T.-T.; Norris, T.; Sun, S.; Zhang, T.; Han, T.-L.; Rowan, A.; Xia, Y.-Y.; Zhang, H.; Qi, H.-B.; et al. The CLIMB (Complex Lipids In Mothers and Babies) study: Protocol for a multicentre, three-group, parallel randomised controlled trial to investigate the effect of supplementation of complex lipids in pregnancy, on maternal ganglioside status and subsequent. BMJ Open 2017, 7, 16637. [Google Scholar] [CrossRef]
- Norris, T.; Souza, R.; Xia, Y.; Zhang, T.; Rowan, A.; Gallier, S.; Zhang, H.; Qi, H.; Baker, P. Effect of supplementation of complex milk lipids in pregnancy on fetal growth: Results from the Complex Lipids in Mothers and Babies (CLIMB) randomized controlled trial. J. Matern. Neonatal Med. 2019, 34, 3313–3322. [Google Scholar] [CrossRef]
- Yuan, Q.; Gong, H.; Du, M.; Mao, X. Supplementation of milk polar lipids to obese dams improves neurodevelopment and cognitive function in male offspring. FASEB J. 2021, 35, e21454. [Google Scholar] [CrossRef]
- Li, T.; Gong, H.; Yuan, Q.; Du, M.; Ren, F.; Mao, X. Supplementation of polar lipids-enriched milk fat globule membrane in high-fat diet-fed rats during pregnancy and lactation promotes brown/beige adipocyte development and prevents obesity in male offspring. FASEB J. 2020, 34, 4619–4634. [Google Scholar] [CrossRef]
- Li, T.; Yuan, Q.; Gong, H.; Du, M.; Mao, X. Gut microbiota mediates the alleviative effect of polar lipids-enriched milk fat globule membrane on obesity-induced glucose metabolism disorders in peripheral tissues in rat dams. Int. J. Obes. 2022, 46, 793–801. [Google Scholar] [CrossRef]
- Milard, M.; Laugerette, F.; Durand, A.; Buisson, C.; Meugnier, E.; Loizon, E.; Louche-Pelissier, C.; Sauvinet, V.; Garnier, L.; Viel, S.; et al. Milk Polar Lipids in a High-Fat Diet Can Prevent Body Weight Gain: Modulated Abundance of Gut Bacteria in Relation with Fecal Loss of Specific Fatty Acids. Mol. Nutr. Food Res. 2019, 63, e1801078. [Google Scholar] [CrossRef] [Green Version]
- Demmer, E.; Van Loan, M.D.; Rivera, N.; Rogers, T.S.; Gertz, E.R.; German, J.B.; Smilowitz, J.T.; Zivkovic, A.M. Addition of a dairy fraction rich in milk fat globule membrane to a high-saturated fat meal reduces the postprandial insulinaemic and inflammatory response in overweight and obese adults. J. Nutr. Sci. 2016, 5, e14. [Google Scholar] [CrossRef] [Green Version]
- Cirulli, F.; Berry, A. Early Developmental Trajectories of Brain Development: New Directions in the Search for Early Determinants of Health and Longevity. In Adaptive and Maladaptive Aspects of Developmental Stress; Springer: New York, NY, USA, 2012; pp. 211–227. [Google Scholar] [CrossRef]
- Berry, A.; Bellisario, V.; Panetta, P.; Raggi, C.; Magnifico, M.C.; Arese, M.; Cirulli, F. Administration of the Antioxidant N-Acetyl-Cysteine in Pregnant Mice Has Long-Term Positive Effects on Metabolic and Behavioral Endpoints of Male and Female Offspring Prenatally Exposed to a High-Fat Diet. Front. Behav. Neurosci. 2018, 12, 48. [Google Scholar] [CrossRef] [Green Version]
- Khullar, S.; Greenwood, S.L.; McCord, N.; Glazier, J.D.; Ayuk, P.T.-Y. Nitric oxide and superoxide impair human placental amino acid uptake and increase Na+ permeability: Implications for fetal growth. Free Radic. Biol. Med. 2004, 36, 271–277. [Google Scholar] [CrossRef]
- Mokhtari, V.; Afsharian, P.; Shahhoseini, M.; Kalantar, S.M.; Moini, A. A Review on Various Uses of N-Acetyl Cysteine. Cell J. 2017, 19, 11–17. [Google Scholar] [CrossRef]
- Oh, S.-I.; Park, S.-K. N-acetyl-l-cysteine mimics the effect of dietary restriction on lifespan and reduces amyloid beta-induced toxicity in Caenorhabditis elegans. Food Sci. Biotechnol. 2017, 26, 783–790. [Google Scholar] [CrossRef]
- Berry, A.; Cirulli, F. The p66Shc gene paves the way for healthspan: Evolutionary and mechanistic perspectives. Neurosci. Biobehav. Rev. 2013, 37, 790–802. [Google Scholar] [CrossRef]
- Masschelin, P.M.; Cox, A.R.; Chernis, N.; Hartig, S.M. The Impact of Oxidative Stress on Adipose Tissue Energy Balance. Front. Physiol. 2020, 10, 1638. [Google Scholar] [CrossRef]
- Berniakovich, I.; Trinei, M.; Stendardo, M.; Migliaccio, E.; Minucci, S.; Bernardi, P.; Pelicci, P.G.; Giorgio, M. p66Shc-generated Oxidative Signal Promotes Fat Accumulation. J. Biol. Chem. 2008, 283, 34283–34293. [Google Scholar] [CrossRef] [Green Version]
- Motawei, S.M.; Attalla, S.; Gouda, H.E.; Harouny, M.A.; Elmansoury, A.M. The effects of N-acetyl cysteine on oxidative stress among patients with pre-eclampsia. Int. J. Gynecol. Obstet. 2016, 135, 226–227. [Google Scholar] [CrossRef]
- Giorgio, M.; Berry, A.; Berniakovich, I.; Poletaeva, I.; Trinei, M.; Stendardo, M.; Hagopian, K.; Ramsey, J.J.; Cortopassi, G.; Migliaccio, E.; et al. The p66Shcknockout mice are short lived under natural condition. Aging Cell 2011, 11, 162–168. [Google Scholar] [CrossRef] [Green Version]
- Kabe, Y.; Ando, K.; Hirao, S.; Yoshida, M.; Handa, H. Redox Regulation of NF-κB Activation: Distinct Redox Regulation Between the Cytoplasm and the Nucleus. Antioxid. Redox Signal. 2005, 7, 395–403. [Google Scholar] [CrossRef]
- Oka, S.-I.; Kamata, H.; Kamata, K.; Yagisawa, H.; Hirata, H. N-Acetylcysteine suppresses TNF-induced NF-κB activation through inhibition of IκB kinases. FEBS Lett. 2000, 472, 196–202. [Google Scholar] [CrossRef] [Green Version]
- Tenório, M.C.d.S.; Graciliano, N.G.; Moura, F.; de Oliveira, A.C.M.; Goulart, M.O.F. N-Acetylcysteine (NAC): Impacts on Human Health. Antioxidants 2021, 10, 967. [Google Scholar] [CrossRef]
- Williams, L.; Burgos, E.S.; Vuguin, P.M.; Manuel, C.R.; Pekson, R.; Munnangi, S.; Reznik, S.E.; Charron, M.J. N-Acetylcysteine Resolves Placental Inflammatory-Vasculopathic Changes in Mice Consuming a High-Fat Diet. Am. J. Pathol. 2019, 189, 2246–2257. [Google Scholar] [CrossRef]
- Paintlia, M.K.; Paintlia, A.S.; Singh, A.K.; Singh, I. Attenuation of Lipopolysaccharide-Induced Inflammatory Response and Phospholipids Metabolism at the Feto-Maternal Interface by N-Acetyl Cysteine. Pediatr. Res. 2008, 64, 334–339. [Google Scholar] [CrossRef] [Green Version]
- Paintlia, M.K.; Paintlia, A.; Barbosa, E.; Singh, I.; Singh, A.K. N-acetylcysteine prevents endotoxin-induced degeneration of oligodendrocyte progenitors and hypomyelination in developing rat brain. J. Neurosci. Res. 2004, 78, 347–361. [Google Scholar] [CrossRef]
- Lopez-Jaramillo, P.; Arenas, W.D.; Garcia, R.G.; Rincon, M.Y.; López, M. Review: The role of the L-arginine-nitric oxide pathway in preeclampsia. Ther. Adv. Cardiovasc. Dis. 2008, 2, 261–275. [Google Scholar] [CrossRef] [Green Version]
- Lopez-Jaramillo, P.; Barajas, J.; Rueda-Quijano, S.M.; Lopez-Lopez, C.; Felix, C. Obesity and Preeclampsia: Common Pathophysiological Mechanisms. Front. Physiol. 2018, 9, 1838. [Google Scholar] [CrossRef] [Green Version]
- Tenório, M.B.; Ferreira, R.C.; Moura, F.A.; Bueno, N.B.; De Oliveira, A.C.M.; Goulart, M.O.F. Cross-Talk between Oxidative Stress and Inflammation in Preeclampsia. Oxidative Med. Cell. Longev. 2019, 2019, 1–26. [Google Scholar] [CrossRef] [Green Version]
- Luo, Z.; Xu, X.; Sho, T.; Luo, W.; Zhang, J.; Xu, W.; Yao, J.; Xu, J. Effects of n-acetyl-cysteine supplementation in late gestational diet on maternal-placental redox status, placental NLRP3 inflammasome, and fecal microbiota in sows1. J. Anim. Sci. 2019, 97, 1757–1771. [Google Scholar] [PubMed]
- Gaccioli, F.; Aye, I.L.M.H.; Roos, S.; Lager, S.; Ramirez, V.I.; Kanai, Y.; Powell, T.L.; Jansson, T. Expression and functional characterisation of System L amino acid transporters in the human term placenta. Reprod. Biol. Endocrinol. 2015, 13, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, H.J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosenblat, J.D.; McIntyre, R.S. Bipolar Disorder and Immune Dysfunction: Epidemiological Findings, Proposed Pathophysiology and Clinical Implications. Brain Sci. 2017, 7, 144. [Google Scholar] [CrossRef] [Green Version]
- Severance, E.G.; Yolken, R.H.; Eaton, W.W. Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: More than a gut feeling. Schizophr. Res. 2014, 176, 23–35. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. 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
Cirulli, F.; De Simone, R.; Musillo, C.; Ajmone-Cat, M.A.; Berry, A. Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies. Nutrients 2022, 14, 3150. https://doi.org/10.3390/nu14153150
Cirulli F, De Simone R, Musillo C, Ajmone-Cat MA, Berry A. Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies. Nutrients. 2022; 14(15):3150. https://doi.org/10.3390/nu14153150
Chicago/Turabian StyleCirulli, Francesca, Roberta De Simone, Chiara Musillo, Maria Antonietta Ajmone-Cat, and Alessandra Berry. 2022. "Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies" Nutrients 14, no. 15: 3150. https://doi.org/10.3390/nu14153150
APA StyleCirulli, F., De Simone, R., Musillo, C., Ajmone-Cat, M. A., & Berry, A. (2022). Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies. Nutrients, 14(15), 3150. https://doi.org/10.3390/nu14153150