Mechanisms of Blood–Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids
Abstract
:1. Introduction
2. Regulation of the BBB Paracellular Permeability in Normal and Pathological Conditions
3. SCFAs and Their Impact on Brain Health
4. Effect of SCFA-Produced Microbiota on BBB Integrity
5. Protective Effect of SCFAs on BBB Integrity in In Vitro BBB Models and in In Vivo Animal Models of CNS Pathologies
6. SCFAs as Ligands of G Protein-Coupled Receptors
7. SCFAs as Histone Deacetylase Inhibitors
8. Revealed and Putative Mechanisms Underlying SCFAs Protective Effect on the BBB
8.1. SCFAs/HDAC/NF-κB
8.2. SCFAs/GPR43/β-Arrestin-2/NF-κB
8.3. SCFAs/NF-κB/NLRP3 Inflammasome
8.4. SCFAs/NF-κB/MMP-9
8.5. SCFA/Keap-1/Nrf2 Signaling Pathway
8.6. SCFAs/HDAC/FoxO1/Claudin-5
8.7. SCFAs / HDAC / PPAR γ
8.8. SCFAs/Myosin Light Chain Kinase
8.9. SCFAs/Wnt/β-Catenin
9. Indirect Effects of SCFA on BBB Integrity
9.1. Systemic Inflammation
9.2. Microglia and Astrocytes
10. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
AJ | adherens junction |
AMPK | AMP-activated protein kinase |
ApoE | apolipoprotein E |
ARE | antioxidant response element |
BBB | blood–brain barrier |
bEnd.3 | murine brain endothelial cell line |
COX2 | cyclooxygenase 2 |
CNS | central nervous system |
ERK1/2 | extracellular-signal-regulated kinase 1/2 |
GCLC | glutamate-cysteine ligase catalytic subunit |
GCLM | glutamate-cysteine ligase regulatory subunit |
GJ | gap junctions |
GPR | G protein-coupled receptor |
HAT | histone acetylase |
HDAC | histone deacetylase |
hCMEC/D3 | human cerebral microvascular endothelial cell line |
H3K9/14 | histone |
H3 | acetylated in lysine 9/14 |
HO-1 | heme oxygenase 1 |
i.g. | intragastrically |
IL-1β | interleukin 1-β |
iNOS | inducible NO-synthase |
i.p. | intraperitoneally |
JAM | junction adhesion molecule |
JNK1/2 | C-Jun N-terminal kinase |
Keap1 | Kelch-like ECH-associated protein 1 |
LPS | lipopolysaccharide |
NF-κB | nuclear factor kappa B |
Nlrp3 | NOD-like receptor family pyrin domain containing 3 |
NQO1 | NAD(P)H:quinone oxidoreductase-1 |
Nrf2 | nuclear erythroid 2-related factor 2 |
NVU | neurovascular unit |
MCAO | middle cerebral artery occlusion |
MCT | monocarboxylate transporter |
MMP | metalloproteinase |
PPARγ | peroxisome proliferator-activated receptor gamma |
SB | sodium butyrate |
SCFA | short-chain fatty acid |
SOD1 | superoxide dismutase 1 |
T2DM | type 2 diabetes mellitus |
TEER | transendothelial electrical resistance |
TJ | tight junction |
TNFα | tumor necrosis factor alpha |
ZO | zonula occludens |
References
- Sweeney, M.D.; Ayyadurai, S.; Zlokovic, B.V. Pericytes of the neurovascular unit: Key functions and signaling pathways. Nat. Neurosci. 2016, 19, 771–783. [Google Scholar] [CrossRef]
- Muoio, V.; Persson, P.B.; Sendeski, M.M. The neurovascular unit—Concept review. Acta Physiol. 2014, 210, 790–798. [Google Scholar] [CrossRef]
- Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef]
- Nehra, G.; Bauer, B.; Hartz, A.M. Blood-brain barrier leakage in Alzheimer’s disease: From discovery to clinical relevance. Pharmacol. Ther. 2022, 234, 108119. [Google Scholar] [CrossRef]
- Takata, F.; Nakagawa, S.; Matsumoto, J.; Dohgu, S. Blood-Brain Barrier Dysfunction Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction. Front. Cell. Neurosci. 2021, 15, 661838. [Google Scholar] [CrossRef]
- Varatharaj, A.; Galea, I. The blood-brain barrier in systemic inflammation. Brain Behav. Immun. 2017, 60, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Tang, C.F.; Wang, C.Y.; Wang, J.H.; Wang, Q.N.; Li, S.J.; Wang, H.O.; Zhou, F.; Li, J.M. Short-Chain Fatty Acids Ameliorate Depressive-like Behaviors of High Fructose-Fed Mice by Rescuing Hippocampal Neurogenesis Decline and Blood–Brain Barrier Damage. Nutrients 2022, 14, 1882. [Google Scholar] [CrossRef]
- Hu, L.; Zhu, S.; Peng, X.; Li, K.; Peng, W.; Zhong, Y.; Kang, C.; Cao, X.; Liu, Z.; Zhao, B. High Salt Elicits Brain Inflammation and Cognitive Dysfunction, Accompanied by Alternations in the Gut Microbiota and Decreased SCFA Production. J. Alzheimer’s Dis. 2020, 77, 629–640. [Google Scholar] [CrossRef]
- Wu, Q.; Zhang, Y.; Zhang, Y.; Xia, C.; Lai, Q.; Dong, Z.; Kuang, W.; Yang, C.; Su, D.; Li, H.; et al. Potential effects of antibiotic-induced gut microbiome alteration on blood–brain barrier permeability compromise in rhesus monkeys. Ann. N. Y. Acad. Sci. 2020, 1470, 14–24. [Google Scholar] [CrossRef]
- Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol 2020, 11, 25. [Google Scholar] [CrossRef] [Green Version]
- Cryan, J.F.; O’Riordan, K.J.; Sandhu, K.; Peterson, V.; Dinan, T.G. The gut microbiome in neurological disorders. Lancet Neurol. 2020, 19, 179–194. [Google Scholar] [CrossRef]
- Dinan, T.G.; Cryan, J.F. Gut instincts: Microbiota as a key regulator of brain development, ageing and neurodegeneration. J. Physiol. 2017, 595, 489–503. [Google Scholar] [CrossRef] [Green Version]
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [Green Version]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef]
- O’Riordan, K.J.; Collins, M.K.; Moloney, G.M.; Knox, E.G.; Aburto, M.R.; Fülling, C.; Morley, S.J.; Clarke, G.; Schellekens, H.; Cryan, J.F. Short chain fatty acids: Microbial metabolites for gut-brain axis signalling. Mol. Cell. Endocrinol. 2022, 546, 111572. [Google Scholar] [CrossRef]
- Brown, A.J.; Goldsworthy, S.M.; Barnes, A.A.; Eilert, M.M.; Tcheang, L.; Daniels, D.; Muir, A.I.; Wigglesworth, M.J.; Kinghorn, I.; Fraser, N.J.; et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J. Biol. Chem. 2003, 278, 11312–11319. [Google Scholar] [CrossRef] [Green Version]
- Li, M.; Van Esch, B.C.; Henricks, P.A.; Folkerts, G.; Garssen, J. The Anti-inflammatory Effects of Short Chain Fatty Acids on Lipopolysaccharide- or Tumor Necrosis Factor α-Stimulated Endothelial Cells via Activation of GPR41/43 and Inhibition of HDACs. Front. Pharmacol. 2018, 9, 533. [Google Scholar] [CrossRef] [Green Version]
- González-Bosch, C.; Boorman, E.; Zunszain, P.A.; Mann, G.E. Short-chain fatty acids as modulators of redox signaling in health and disease. Redox Biol. 2021, 47, 102165. [Google Scholar] [CrossRef]
- Erny, D.; Hrabě 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]
- Ahmed, H.; Leyrolle, Q.; Koistinen, V.; Kärkkäinen, O.; Layé, S.; Delzenne, N.; Hanhineva, K. Microbiota-derived metabolites as drivers of gut–brain communication. Gut Microbes 2022, 14, 2102878. [Google Scholar] [CrossRef]
- Dong, Y.; Cui, C. The role of short-chain fatty acids in central nervous system diseases. Mol. Cell. Biochem. 2022, 477, 2595–2607. [Google Scholar] [CrossRef]
- Li, H.; Sun, J.; Du, J.; Wang, F.; Fang, R.; Yu, C.; Xiong, J.; Chen, W.; Lu, Z.; Liu, J. Clostridium butyricum exerts a neuroprotective effect in a mouse model of traumatic brain injury via the gut-brain axis. Neurogastroenterol. Motil. 2018, 30, e13260. [Google Scholar] [CrossRef]
- Sun, N.; Hu, H.; Wang, F.; Li, L.; Zhu, W.; Shen, Y.; Xiu, J.; Xu, Q. Antibiotic-induced microbiome depletion in adult mice disrupts blood-brain barrier and facilitates brain infiltration of monocytes after bone-marrow transplantation. Brain Behav. Immun. 2021, 92, 102–114. [Google Scholar] [CrossRef]
- Liu, J.; Sun, J.; Wang, F.; Yu, X.; Ling, Z.; Li, H.; Zhang, H.; Jin, J.; Chen, W.; Pang, M.; et al. Neuroprotective Effects of Clostridium butyricum against Vascular Dementia in Mice via Metabolic Butyrate. BioMed Res. Int. 2015, 2015, 512946. [Google Scholar] [CrossRef] [Green Version]
- Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [Green Version]
- Fröhlich, E.E.; Farzi, A.; Mayerhofer, R.; Reichmann, F.; Jačan, A.; Wagner, B.; Zinser, E.; Bordag, N.; Magnes, C.; Fröhlich, E.; et al. Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain Behav. Immun. 2016, 56, 140–155. [Google Scholar] [CrossRef] [Green Version]
- Hoyles, L.; Pontifex, M.G.; Rodriguez-Ramiro, I.; Anis-Alavi, M.A.; Jelane, K.S.; Snelling, T.; Solito, E.; Fonseca, S.; Carvalho, A.L.; Carding, S.R.; et al. Regulation of blood–brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. Microbiome 2021, 9, 235. [Google Scholar] [CrossRef]
- Parker, A.; Fonseca, S.; Carding, S.R. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes 2020, 11, 135–157. [Google Scholar] [CrossRef] [Green Version]
- Park, M.J.; Sohrabji, F. The histone deacetylase inhibitor, sodium butyrate, exhibits neuroprotective effects for ischemic stroke in middle-aged female rats. J. Neuroinflamm. 2016, 13, 300. [Google Scholar] [CrossRef] [Green Version]
- Li, H.; Sun, J.; Wang, F.; Ding, G.; Chen, W.; Fang, R.; Yao, Y.; Pang, M.; Lu, Z.-Q.; Liu, J. Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain Res. 2016, 1642, 70–78. [Google Scholar] [CrossRef]
- Liu, J.; Jin, Y.; Ye, Y.; Tang, Y.; Dai, S.; Li, M.; Zhao, G.; Hong, G.; Lu, Z.-Q. The Neuroprotective Effect of Short Chain Fatty Acids against Sepsis-Associated Encephalopathy in Mice. Front. Immunol. 2021, 12, 626894. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Wang, F.; Liu, S.; Du, J.; Hu, X.; Xiong, J.; Fang, R.; Chen, W.; Sun, J. Sodium butyrate exerts protective effect against Parkinson’s disease in mice via stimulation of glucagon like peptide-1. J. Neurol. Sci. 2017, 381, 176–181. [Google Scholar] [CrossRef]
- Hoyles, L.; Snelling, T.; Umlai, U.-K.; Nicholson, J.K.; Carding, S.R.; Glen, R.C.; McArthur, S. Microbiome–host systems interactions: Protective effects of propionate upon the blood–brain barrier. Microbiome 2018, 6, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Villabona-Rueda, A.; Erice, C.; Pardo, C.A.; Stins, M.F. The Evolving Concept of the Blood Brain Barrier (BBB): From a Single Static Barrier to a Heterogeneous and Dynamic Relay Center. Front. Cell. Neurosci. 2019, 13, 405. [Google Scholar] [CrossRef]
- Andreone, B.J.; Chow, B.W.; Tata, A.; Lacoste, B.; Ben-Zvi, A.; Bullock, K.; Deik, A.A.; Ginty, D.D.; Clish, C.B.; Gu, C. Blood-Brain Barrier Permeability Is Regulated by Lipid Transport-Dependent Suppression of Caveolae-Mediated Transcytosis. Neuron 2017, 94, 581–594.e5. [Google Scholar] [CrossRef] [Green Version]
- Kadry, H.; Noorani, B.; Cucullo, L. A blood–brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef]
- Galea, I. The blood–brain barrier in systemic infection and inflammation. Cell. Mol. Immunol. 2021, 18, 2489–2501. [Google Scholar] [CrossRef]
- Cong, X.; Kong, W. Endothelial tight junctions and their regulatory signaling pathways in vascular homeostasis and disease. Cell. Signal. 2020, 66, 109485. [Google Scholar] [CrossRef]
- Parnova, R.G. Critical Role of Endothelial Lysophosphatidylcholine Transporter Mfsd2a in Maintaining Blood–Brain Barrier Integrity and Delivering Omega 3 PUFA to the Brain. J. Evol. Biochem. Physiol. 2022, 58, 742–754. [Google Scholar] [CrossRef]
- Tietz, S.; Engelhardt, B. Brain barriers: Crosstalk between complex tight junctions and adherens junctions. J. Cell Biol. 2015, 209, 493–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keep, R.F.; Andjelkovic, A.V.; Xiang, J.; Stamatovic, S.M.; Antonetti, D.; Hua, Y.; Xi, G. Brain endothelial cell junctions after cerebral hemorrhage: Changes, mechanisms and therapeutic targets. J. Cereb. Blood Flow Metab. 2018, 38, 1255–1275. [Google Scholar] [CrossRef] [PubMed]
- Goncalves, A.; Antonetti, D.A. Transgenic animal models to explore and modulate the blood brain and blood retinal barriers of the CNS. Fluids Barriers CNS 2022, 19, 86. [Google Scholar] [CrossRef]
- Stamatovic, S.M.; Johnson, A.M.; Sladojevic, N.; Keep, R.F.; Andjelkovic, A.V. Endocytosis of tight junction proteins and the regulation of degradation and recycling. Ann. N. Y. Acad. Sci. 2017, 1397, 54–65. [Google Scholar] [CrossRef] [PubMed]
- Stamatovic, S.M.; Johnson, A.M.; Keep, R.F.; Andjelkovic, A.V. Junctional proteins of the blood-brain barrier: New insights into function and dysfunction. Tissue Barriers 2016, 4, e1154641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Itallie, C.M.; Anderson, J.M. Phosphorylation of tight junction transmembrane proteins: Many sites, much to do. Tissue Barriers 2018, 6, e1382671. [Google Scholar] [CrossRef] [Green Version]
- Stamatovic, S.M.; Keep, R.F.; Andjelkovic, A.V. Brain endothelial cell-cell junctions: How to “open” the blood brain barrier. Curr. Neuropharmacol. 2008, 6, 179–192. [Google Scholar] [CrossRef] [Green Version]
- Mondal, N.K.; Behera, J.; Kelly, K.E.; George, A.K.; Tyagi, P.K.; Tyagi, N. Tetrahydrocurcumin epigenetically mitigates mitochondrial dysfunction in brain vasculature during ischemic stroke. Neurochem. Int. 2019, 122, 120–138. [Google Scholar] [CrossRef]
- Stamatovic, S.M.; Phillips, C.M.; Martinez-Revollar, G.; Keep, R.F.; Andjelkovic, A.V. Involvement of Epigenetic Mechanisms and Non-coding RNAs in Blood-Brain Barrier and Neurovascular Unit Injury and Recovery after Stroke. Front. Neurosci. 2019, 13, 864. [Google Scholar] [CrossRef]
- Zhuang, Y.; Peng, H.; Mastej, V.; Chen, W. MicroRNA Regulation of Endothelial Junction Proteins and Clinical Consequence. Mediat. Inflamm. 2016, 2016, 5078627. [Google Scholar] [CrossRef] [Green Version]
- Niccolai, E.; Baldi, S.; Ricci, F.; Russo, E.; Nannini, G.; Menicatti, M.; Poli, G.; Taddei, A.; Bartolucci, G.; Calabrò, A.S.; et al. Evaluation and comparison of short chain fatty acids composition in gut diseases. World J. Gastroenterol. 2019, 25, 5543–5558. [Google Scholar] [CrossRef]
- Tarini, J.; Wolever, T.M.S. The fermentable fibre inulin increases postprandial serum short-chain fatty acids and reduces free-fatty acids and ghrelin in healthy subjects. Appl. Physiol. Nutr. Metab. 2010, 35, 9–16. [Google Scholar] [CrossRef]
- Opeyemi, M.O.M.; Rogers, M.B.; Firek, B.A.; Janesko-Feldman, M.K.; Vagni, V.; Mullett, S.J.; Wendell, S.G.; Nelson, B.P.; New, L.A.; Mariño, E.; et al. Sustained Dysbiosis and Decreased Fecal Short-Chain Fatty Acids after Traumatic Brain Injury and Impact on Neurologic Outcome. J. Neurotrauma 2021, 38, 2610–2621. [Google Scholar] [CrossRef] [PubMed]
- Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vogt, J.A.; Pencharz, P.B.; Wolever, T.M. l-Rhamnose increases serum propionate in humans. Am. J. Clin. Nutr. 2004, 80, 89–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nilsson, A.C.; Östman, E.M.; Knudsen, K.E.B.; Holst, J.J.; Björck, I.M.E. A cereal-based evening meal rich in indigestible carbohydrates increases plasma butyrate the next morning. J. Nutr. 2010, 140, 1932–1936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, A.; Li, S.; Wang, X.; Xie, Z.; Li, S.; Hua, D. Cefazolin Improves Anesthesia and Surgery-Induced Cognitive Impairments by Modulating Blood-Brain Barrier Function, Gut Bacteria and Short Chain Fatty Acids. Front. Aging Neurosci. 2021, 13, 748637. [Google Scholar] [CrossRef] [PubMed]
- Vijay, N.; Morris, M.E. Role of monocarboxylate transporters in drug delivery to the brain. Curr. Pharm. Des. 2014, 20, 1487–1498. [Google Scholar] [CrossRef] [Green Version]
- Uchida, Y.; Ohtsuki, S.; Katsukura, Y.; Ikeda, C.; Suzuki, T.; Kamiie, J.; Terasaki, T. Quantitative targeted absolute proteomics of human blood-brain barrier transporters and receptors. J. Neurochem. 2011, 117, 333–345. [Google Scholar] [CrossRef]
- Gerhart, D.Z.; Enerson, B.E.; Zhdankina, O.Y.; Leino, R.L.; Drewes, L.R. Expression of monocarboxylate transporter MCT1 by brain endothelium and glia in adult and suckling rats. Am. J. Physiol. Metab. 1997, 273, E207–E213. [Google Scholar] [CrossRef]
- Leino, R.L.; Gerhart, D.Z.; Drewes, L.R. Monocarboxylate transporter (MCT1) abundance in brains of suckling and adult rats: A quantitative electron microscopic immunogold study. Dev. Brain Res. 1999, 113, 47–54. [Google Scholar] [CrossRef]
- Mitchell, R.W.; On, N.H.; Del Bigio, M.R.; Miller, D.W.; Hatch, G.M. Fatty acid transport protein expression in human brain and potential role in fatty acid transport across human brain microvessel endothelial cells. J. Neurochem. 2011, 117, 735–746. [Google Scholar] [CrossRef] [PubMed]
- Oldendorf, W.H. Carrier-mediated blood-brain barrier transport of short-chain monocarboxylic organic acids. Am. J. Physiol. Content 1973, 224, 1450–1453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bachmann, C.; Colombo, J.-P.; Berüter, J. Short chain fatty acids in plasma and brain: Quantitative determination by gas chromatography. Clin. Chim. Acta 1979, 92, 153–159. [Google Scholar] [CrossRef] [PubMed]
- Gershon, M.D.; Margolis, K.G. The gut, its microbiome, and the brain: Connections and communications. J. Clin. Investig. 2021, 131. [Google Scholar] [CrossRef] [PubMed]
- Saresella, M.; Marventano, I.; Barone, M.; La Rosa, F.; Piancone, F.; Mendozzi, L.; D’Arma, A.; Rossi, V.; Pugnetti, L.; Roda, G.; et al. Alterations in Circulating Fatty Acid Are Associated with Gut Microbiota Dysbiosis and Inflammation in Multiple Sclerosis. Front. Immunol. 2020, 11, 1390. [Google Scholar] [CrossRef]
- Stanley, D.; Moore, R.J.; Wong, C.H.Y. An insight into intestinal mucosal microbiota disruption after stroke. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Liao, H.; Li, H.; Bao, H.; Jiang, L.; Du, J.; Guo, Y.; Si, Y. Short Chain Fatty Acids Protect the Cognitive Function of Sepsis Associated Encephalopathy Mice via GPR43. Front. Neurol. 2022, 13, 909439. [Google Scholar] [CrossRef]
- Zhang, L.; Wang, Y.; Xiayu, X.; Shi, C.; Chen, W.; Song, N.; Fu, X.; Zhou, R.; Xu, Y.-F.; Huang, L.; et al. Altered Gut Microbiota in a Mouse Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 60, 1241–1257. [Google Scholar] [CrossRef]
- Wallen, Z.D.; Appah, M.; Dean, M.N.; Sesler, C.L.; Factor, S.A.; Molho, E.; Zabetian, C.P.; Standaert, D.G.; Payami, H. Characterizing dysbiosis of gut microbiome in PD: Evidence for overabundance of opportunistic pathogens. NPJ Park. Dis. 2020, 6, 1–12. [Google Scholar] [CrossRef]
- Chen, R.; Xu, Y.; Wu, P.; Zhou, H.; Lasanajak, Y.; Fang, Y.; Tang, L.; Ye, L.; Li, X.; Cai, Z.; et al. Transplantation of fecal microbiota rich in short chain fatty acids and butyric acid treat cerebral ischemic stroke by regulating gut microbiota. Pharmacol. Res. 2019, 148, 104403. [Google Scholar] [CrossRef]
- Wen, J.; Ding, Y.; Wang, L.; Xiao, Y. Gut microbiome improves postoperative cognitive function by decreasing permeability of the blood-brain barrier in aged mice. Brain Res. Bull. 2020, 164, 249–256. [Google Scholar] [CrossRef] [PubMed]
- Couto, M.R.; Gonçalves, P.; Magro, F.; Martel, F. Microbiota-derived butyrate regulates intestinal inflammation: Focus on inflammatory bowel disease. Pharmacol. Res. 2020, 159, 104947. [Google Scholar] [CrossRef] [PubMed]
- Lewis, K.; Lutgendorff, F.; Phan, V.; Söderholm, J.D.; Sherman, P.M.; McKay, D.M. Enhanced translocation of bacteria across metabolically stressed epithelia is reduced by butyrate. Inflamm. Bowel Dis. 2010, 16, 1138–1148. [Google Scholar] [CrossRef] [PubMed]
- Leclercq, S.; Mian, F.M.; Stanisz, A.M.; Bindels, L.B.; Cambier, E.; Ben-Amram, H.; Koren, O.; Forsythe, P.; Bienenstock, J. Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nat. Commun. 2017, 8, 15062. [Google Scholar] [CrossRef] [Green Version]
- Hurst, R.D.; Clark, J.B. Butyric acid mediated induction of enhanced transendothelial resistance in an in vitro model blood–brain barrier system. Neurochem. Int. 1999, 35, 261–267. [Google Scholar] [CrossRef]
- Knox, E.G.; Aburto, M.R.; Tessier, C.; Nagpal, J.; Clarke, G.; O’Driscoll, C.M.; Cryan, J.F. Microbial-derived metabolites induce actin cytoskeletal rearrangement and protect blood-brain barrier function. iscience 2022, 25, 105648. [Google Scholar] [CrossRef]
- Wang, Z.; Leng, Y.; Tsai, L.-K.; Leeds, P.; Chuang, D.-M. Valproic acid attenuates blood-brain barrier disruption in a rat model of transient focal cerebral ischemia: The roles of HDAC and MMP-9 inhibition. J. Cereb. Blood Flow Metab. 2011, 31, 52–57. [Google Scholar] [CrossRef]
- Nguyen, T.D.; Hållenius, F.F.; Lin, X.; Nyman, M.; Prykhodko, O. Monobutyrin and Monovalerin Affect Brain Short-Chain Fatty Acid Profiles and Tight-Junction Protein Expression in ApoE-Knockout Rats Fed High-Fat Diets. Nutrients 2020, 12, 1202. [Google Scholar] [CrossRef]
- Yang, Y.; Rosenberg, G.A. Blood-brain barrier breakdown in acute and chronic cerebrovascular disease. Stroke 2011, 42, 3323–3328. [Google Scholar] [CrossRef] [Green Version]
- Feng, W.; Wu, Y.; Chen, G.; Fu, S.; Li, B.; Huang, B.; Wang, D.; Wang, W.; Liu, J. Sodium Butyrate Attenuates Diarrhea in Weaned Piglets and Promotes Tight Junction Protein Expression in Colon in a GPR109A-Dependent Manner. Cell. Physiol. Biochem. 2018, 47, 1617–1629. [Google Scholar] [CrossRef]
- Peng, L.; Li, Z.-R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate Enhances the Intestinal Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef] [Green Version]
- Yan, H.; Ajuwon, K.M. Butyrate modifies intestinal barrier function in IPEC-J2 cells through a selective upregulation of tight junction proteins and activation of the Akt signaling pathway. PLoS ONE 2017, 12, e0179586. [Google Scholar] [CrossRef] [Green Version]
- Yang, T.; Datsomor, O.; Jiang, M.; Ma, X.; Zhao, G.; Zhan, K. Protective Roles of Sodium Butyrate in Lipopolysaccharide-Induced Bovine Ruminal Epithelial Cells by Activating G Protein-Coupled Receptors 41. Front. Nutr. 2022, 9, 842634. [Google Scholar] [CrossRef]
- Zhan, K.; Gong, X.; Chen, Y.; Jiang, M.; Yang, T.; Zhao, G. Short-Chain Fatty Acids Regulate the Immune Responses via G Protein-Coupled Receptor 41 in Bovine Rumen Epithelial Cells. Front. Immunol. 2019, 10, 2042. [Google Scholar] [CrossRef]
- Karoor, V.; Strassheim, D.; Sullivan, T.; Verin, A.; Umapathy, N.S.; Dempsey, E.C.; Frank, D.N.; Stenmark, K.R.; Gerasimovskaya, E. The Short-Chain Fatty Acid Butyrate Attenuates Pulmonary Vascular Remodeling and Inflammation in Hypoxia-Induced Pulmonary Hypertension. Int. J. Mol. Sci. 2021, 22, 9916. [Google Scholar] [CrossRef]
- Miyoshi, M.; Usami, M.; Ohata, A. Short-chain fatty acids and trichostatin A alter tight junction permeability in human umbilical vein endothelial cells. Nutrition 2008, 24, 1189–1198. [Google Scholar] [CrossRef] [Green Version]
- Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.Y.; Lannoy, V.; Decobecq, M.-E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Van Damme, J.; et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef] [Green Version]
- Robles-Vera, I.; Toral, M.; De La Visitación, N.; Aguilera-Sánchez, N.; Redondo, J.M.; Duarte, J. Protective Effects of Short-Chain Fatty Acids on Endothelial Dysfunction Induced by Angiotensin II. Front. Physiol. 2020, 11, 277. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Z.; Xu, N.; Matei, N.; McBride, D.W.; Ding, Y.; Liang, H.; Tang, J.; Zhang, J.H. Sodium butyrate attenuated neuronal apoptosis via GPR41/Gβγ/PI3K/Akt pathway after MCAO in rats. J. Cereb. Blood Flow Metab. 2020, 41, 267–281. [Google Scholar] [CrossRef]
- Layden, B.T.; Angueira, A.R.; Brodsky, M.; Durai, V.; Lowe, W.L., Jr. Short chain fatty acids and their receptors: New metabolic targets. Transl. Res. 2013, 161, 131–140. [Google Scholar] [CrossRef]
- Li, H.B.; Xu, M.L.; Du, M.M.; Yu, X.J.; Bai, J.; Xia, W.J.; Dai, Z.M.; Li, C.X.; Li, Y.; Su, Q.; et al. Curcumin ameliorates hypertension via gut-brain communication in spontaneously hypertensive rat. Toxicol. Appl. Pharmacol. 2021, 429, 115701. [Google Scholar] [CrossRef]
- Ikeda, T.; Nishida, A.; Yamano, M.; Kimura, I. Short-chain fatty acid receptors and gut microbiota as therapeutic targets in metabolic, immune, and neurological diseases. Pharmacol. Ther. 2022, 239, 108273. [Google Scholar] [CrossRef]
- Luo, Q.-J.; Sun, M.-X.; Guo, Y.-W.; Tan, S.-W.; Wu, X.-Y.; Abassa, K.-K.; Lin, L.; Liu, H.-L.; Jiang, J.; Wei, X.-Q. Sodium butyrate protects against lipopolysaccharide-induced liver injury partially via the GPR43/β-arrestin-2/NF-κB network. Gastroenterol. Rep. 2020, 9, 154–165. [Google Scholar] [CrossRef]
- Bolognini, D.; Tobin, A.B.; Milligan, G.; Moss, C.E. The Pharmacology and Function of Receptors for Short-Chain Fatty Acids. Mol. Pharmacol. 2015, 89, 388–398. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.H.; Kang, S.G.; Park, J.H.; Yanagisawa, M.; Kim, C.H. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 2013, 145, 396–406. [Google Scholar] [CrossRef]
- Kobayashi, M.; Mikami, D.; Kimura, H.; Kamiyama, K.; Morikawa, Y.; Yokoi, S.; Kasuno, K.; Takahashi, N.; Taniguchi, T.; Iwano, M. Short-chain fatty acids, GPR41 and GPR43 ligands, inhibit TNF-α-induced MCP-1 expression by modulating p38 and JNK signaling pathways in human renal cortical epithelial cells. Biochem. Biophys. Res. Commun. 2017, 486, 499–505. [Google Scholar] [CrossRef]
- Hung, K.-Y.; Wu, S.-Y.; Pao, H.-P.; Liao, W.-I.; Chu, S.-J. Acetate, a gut bacterial product, ameliorates ischemia-reperfusion induced acute lung injury in rats. Int. Immunopharmacol. 2022, 111, 109136. [Google Scholar] [CrossRef]
- Wen, C.; Xie, T.; Pan, K.; Deng, Y.; Zhao, Z.; Li, N.; Bian, J.; Deng, X.; Zha, Y. Acetate attenuates perioperative neurocognitive disorders in aged mice. Aging 2020, 12, 3862–3879. [Google Scholar] [CrossRef]
- Razazan, A.; Karunakar, P.; Mishra, S.P.; Sharma, S.; Miller, B.; Jain, S.; Yadav, H. Activation of Microbiota Sensing—Free Fatty Acid Receptor 2 Signaling Ameliorates Amyloid-β Induced Neurotoxicity by Modulating Proteolysis-Senescence Axis. Front. Aging Neurosci. 2021, 13, 735933. [Google Scholar] [CrossRef]
- Wise, A.; Foord, S.M.; Fraser, N.J.; Barnes, A.A.; Elshourbagy, N.; Eilert, M.; Ignar, D.M.; Murdock, P.R.; Steplewski, K.; Green, A.; et al. Molecular identification of high and low affinity receptors for nicotinic acid. J. Biol. Chem. 2003, 278, 9869–9874. [Google Scholar] [CrossRef] [Green Version]
- Taggart, A.K.; Kero, J.; Gan, X.; Cai, T.-Q.; Cheng, K.; Ippolito, M.; Ren, N.; Kaplan, R.; Wu, K.; Wu, T.-J.; et al. (D)-beta-Hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. J. Biol. Chem. 2005, 280, 26649–26652. [Google Scholar] [CrossRef] [Green Version]
- Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef] [Green Version]
- Guo, W.; Li, W.; Su, Y.; Liu, S.; Kan, X.; Ran, X.; Cao, Y.; Fu, S.; Liu, J. GPR109A alleviate mastitis and enhances the blood milk barrier by activating AMPK/Nrf2 and autophagy. Int. J. Biol. Sci. 2021, 17, 4271–4284. [Google Scholar] [CrossRef]
- Walters, R.W.; Shukla, A.K.; Kovacs, J.J.; Violin, J.D.; DeWire, S.M.; Lam, C.M.; Chen, J.R.; Muehlbauer, M.J.; Whalen, E.J.; Lefkowitz, R.J. β-Arrestin1 mediates nicotinic acid–induced flushing, but not its antilipolytic effect, in mice. J. Clin. Investig. 2009, 119, 1312–1321. [Google Scholar] [CrossRef] [Green Version]
- Cresci, G.A.; Thangaraju, M.; Mellinger, J.D.; Liu, K.; Ganapathy, V. Colonic gene expression in conventional and germ-free mice with a focus on the butyrate receptor GPR109A and the butyrate transporter SLC5A8. J. Gastrointest. Surg. 2010, 14, 449–461. [Google Scholar] [CrossRef]
- Guo, W.; Liu, J.; Sun, J.; Gong, Q.; Ma, H.; Kan, X.; Cao, Y.; Wang, J.; Fu, S. Butyrate alleviates oxidative stress by regulating NRF2 nuclear accumulation and H3K9/14 acetylation via GPR109A in bovine mammary epithelial cells and mammary glands. Free. Radic. Biol. Med. 2020, 152, 728–742. [Google Scholar] [CrossRef]
- Ibrahim, W.W.; Sayed, R.H.; Kandil, E.A.; Wadie, W. Niacin mitigates blood–brain barrier tight junctional proteins dysregulation and cerebral inflammation in ketamine rat model of psychosis: Role of GPR109A receptor. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2022, 119, 110583. [Google Scholar] [CrossRef]
- Seidel, C.; Schnekenburger, M.; Dicato, M.; Diederich, M. Histone deacetylase modulators provided by Mother Nature. Genes Nutr. 2012, 7, 357–367. [Google Scholar] [CrossRef] [Green Version]
- Fessler, E.; Chibane, F.; Wang, Z.; Chuang, D.-M. Potential Roles of HDAC Inhibitors in Mitigating Ischemia-induced Brain Damage and Facilitating Endogenous Regeneration and Recovery. Curr. Pharm. Des. 2013, 19, 5105–5120. [Google Scholar] [CrossRef]
- Spange, S.; Wagner, T.; Heinzel, T.; Krämer, O.H. Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int. J. Biochem. Cell Biol. 2009, 41, 185–198. [Google Scholar] [CrossRef]
- Chuang, D.-M.; Leng, Y.; Marinova, Z.; Kim, H.-J.; Chiu, C.-T. Multiple roles of HDAC inhibition in neurodegenerative conditions. Trends Neurosci. 2009, 32, 591–601. [Google Scholar] [CrossRef] [Green Version]
- Gupta, R.; Ambasta, R.K.; Kumar, P. Pharmacological intervention of histone deacetylase enzymes in the neurodegenerative disorders. Life Sci. 2020, 243, 117278. [Google Scholar] [CrossRef]
- Zhao, Q.; Yu, Z.; Zhang, F.; Huang, L.; Xing, C.; Liu, N.; Xu, Y.; Wang, X. HDAC3 inhibition prevents oxygen glucose deprivation/reoxygenation-induced transendothelial permeability by elevating PPAR γ activity in vitro. J. Neurochem. 2019, 149, 298–310. [Google Scholar] [CrossRef]
- Zhao, Q.; Zhang, F.; Yu, Z.; Guo, S.; Liu, N.; Jiang, Y.; Lo, E.H.; Xu, Y.; Wang, X. HDAC3 inhibition prevents blood-brain barrier permeability through Nrf2 activation in type 2 diabetes male mice. J. Neuroinflamm. 2019, 16, 103. [Google Scholar] [CrossRef] [Green Version]
- Shi, W.; Wei, X.; Wang, Z.; Han, H.; Fu, Y.; Liu, J.; Zhang, Y.; Guo, J.; Dong, C.; Zhou, D.; et al. HDAC 9 exacerbates endothelial injury in cerebral ischaemia/reperfusion injury. J. Cell. Mol. Med. 2016, 20, 1139–1149. [Google Scholar] [CrossRef] [Green Version]
- Cousens, L.S.; Gallwitz, D.; Alberts, B.M. Different accessibilities in chromatin to histone acetylase. J. Biol. Chem. 1979, 254, 1716–1723. [Google Scholar] [CrossRef]
- Luu, M.; Pautz, S.; Kohl, V.; Singh, R.; Romero, R.; Lucas, S.; Hofmann, J.; Raifer, H.; Vachharajani, N.; Carrascosa, L.C.; et al. The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat. Commun. 2019, 10, 760. [Google Scholar] [CrossRef] [Green Version]
- Soliman, M.L.; Rosenberger, T.A. Acetate supplementation increases brain histone acetylation and inhibits histone deacetylase activity and expression. Mol. Cell. Biochem. 2011, 352, 173–180. [Google Scholar] [CrossRef]
- Ma, X.; Wang, Q. Short-Chain Fatty Acids Attenuate Renal Fibrosis and Enhance Autophagy of Renal Tubular Cells in Diabetic Mice through the HDAC2/ULK1 Axis. Endocrinol. Metab. 2022, 37, 432–443. [Google Scholar] [CrossRef]
- Yu, F.; Wang, Z.; Tanaka, M.; Chiu, C.-T.; Leeds, P.; Zhang, Y.; Chuang, D.-M. Posttrauma cotreatment with lithium and valproate: Reduction of lesion volume, attenuation of blood-brain barrier disruption, and improvement in motor coordination in mice with traumatic brain injury. J. Neurosurg. 2013, 119, 766–773. [Google Scholar] [CrossRef]
- Dash, P.K.; Orsi, S.A.; Zhang, M.; Grill, R.J.; Pati, S.; Zhao, J.; Moore, A.N. Valproate administered after traumatic brain injury provides neuroprotection and improves cognitive function in rats. PLoS ONE 2010, 5, e11383. [Google Scholar] [CrossRef] [Green Version]
- Zhao, W.; Zhao, L.; Guo, Z.; Hou, Y.; Jiang, J.; Song, Y. Valproate Sodium Protects Blood Brain Barrier Integrity in Intracerebral Hemorrhage Mice. Oxidative Med. Cell. Longev. 2020, 2020, 8884320. [Google Scholar] [CrossRef]
- Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, M.; Mikami, D.; Uwada, J.; Yazawa, T.; Kamiyama, K.; Kimura, H.; Taniguchi, T.; Iwano, M. A short-chain fatty acid, propionate, enhances the cytotoxic effect of cisplatin by modulating GPR41 signaling pathways in HepG2 cells. Oncotarget 2018, 9, 31342–31354. [Google Scholar] [CrossRef] [Green Version]
- Ahmed, S.M.U.; Luo, L.; Namani, A.; Wang, X.J.; Tang, X. Nrf2 signaling pathway: Pivotal roles in inflammation. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 585–597. [Google Scholar] [CrossRef]
- Bellezza, I.; Giambanco, I.; Minelli, A.; Donato, R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2018, 1865, 721–733. [Google Scholar] [CrossRef]
- Zapolska-Downar, D.; Siennicka, A.; Kaczmarczyk, M.; Kołodziej, B.; Naruszewicz, M. Butyrate inhibits cytokine-induced VCAM-1 and ICAM-1 expression in cultured endothelial cells: The role of NF-κB and PPARα. J. Nutr. Biochem. 2004, 15, 220–228. [Google Scholar] [CrossRef]
- Segain, J.P.; de la Blétière, D.R.; Bourreille, A.; Leray, V.; Gervois, N.; Rosales, C.; Ferrier, L.; Bonnet, C.; Blottière, H.M.; Galmiche, J.P. Butyrate inhibits inflammatory responses through NFkappa B inhibition: Implications for Crohn’s disease. Gut 2000, 47, 397–403. [Google Scholar] [CrossRef] [Green Version]
- Tedelind, S.; Westberg, F.; Kjerrulf, M.; Vidal, A. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: A study with relevance to inflammatory bowel disease. World J. Gastroenterol. 2007, 13, 2826–2832. [Google Scholar] [CrossRef] [PubMed]
- Pun, P.B.L.; Lu, J.; Moochhala, S. Involvement of ROS in BBB dysfunction. Free Radic. Res. 2009, 43, 348–364. [Google Scholar] [CrossRef] [PubMed]
- Haorah, J.; Ramirez, S.H.; Schall, K.; Smith, D.; Pandya, R.; Persidsky, Y. Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to blood-brain barrier dysfunction. J. Neurochem. 2006, 101, 566–576. [Google Scholar] [CrossRef]
- Schreibelt, G.; Kooij, G.; Reijerkerk, A.; van Doorn, R.; Gringhuis, S.I.; van de Pol, S.; Weksler, B.B.; Romero, I.A.; Couraud, P.; Piontek, J.; et al. Reactive oxygen species alter brain endothelial tight junction dynamics via RhoA, PI3 kinase, and PKB signaling. FASEB J. 2007, 21, 3666–3676. [Google Scholar] [CrossRef] [Green Version]
- Aslam, M.; Ahmad, N.; Srivastava, R.; Hemmer, B. TNF-alpha induced NFκB signaling and p65 (RelA) overexpression repress Cldn5 promoter in mouse brain endothelial cells. Cytokine 2012, 57, 269–275. [Google Scholar] [CrossRef]
- Blecharz-Lang, K.G.; Wagner, J.; Fries, A.; Nieminen-Kelhä, M.; Rösner, 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]
- Qin, W.; Li, J.; Zhu, R.; Gao, S.; Fan, J.; Xia, M.; Zhao, R.C.; Zhang, J. Melatonin protects blood-brain barrier integrity and permeability by inhibiting matrix metalloproteinase-9 via the NOTCH3/NF-κB pathway. Aging 2019, 11, 11391–11415. [Google Scholar] [CrossRef]
- Zhu, H.; Dai, R.; Zhou, Y.; Fu, H.; Meng, Q. TLR2 Ligand Pam3CSK4 Regulates MMP-2/9 Expression by MAPK/NF-κB Signaling Pathways in Primary Brain Microvascular Endothelial Cells. Neurochem. Res. 2018, 43, 1897–1904. [Google Scholar] [CrossRef] [Green Version]
- Chen, G.; Ran, X.; Li, B.; Li, Y.; He, D.; Huang, B.; Fu, S.; Liu, J.; Wang, W. Sodium Butyrate Inhibits Inflammation and Maintains Epithelium Barrier Integrity in a TNBS-induced Inflammatory Bowel Disease Mice Model. EBioMedicine 2018, 30, 317–325. [Google Scholar] [CrossRef] [Green Version]
- Fu, J.; Li, G.; Wu, X.; Zang, B. Sodium Butyrate Ameliorates Intestinal Injury and Improves Survival in a Rat Model of Cecal Ligation and Puncture-Induced Sepsis. Inflammation 2019, 42, 1276–1286. [Google Scholar] [CrossRef]
- Yue, X.; Wen, S.; Long-Kun, D.; Man, Y.; Chang, S.; Min, Z.; Shuang-Yu, L.; Xin, Q.; Jie, M.; Liang, W. Three important short-chain fatty acids (SCFAs) attenuate the inflammatory response induced by 5-FU and maintain the integrity of intestinal mucosal tight junction. BMC Immunol. 2022, 23, 19. [Google Scholar] [CrossRef]
- Sun, X.; Luo, S.; Jiang, C.; Tang, Y.; Cao, Z.; Jia, H.; Xu, Q.; Zhao, C.; Loor, J.J.; Xu, C. Sodium butyrate reduces bovine mammary epithelial cell inflammatory responses induced by exogenous lipopolysaccharide, by inactivating NF-κB signaling. J. Dairy Sci. 2020, 103, 8388–8397. [Google Scholar] [CrossRef]
- Chen, L.-F.; Fischle, W.; Verdin, E.; Greene, W.C. Duration of nuclear NF-kappaB action regulated by reversible acetylation. Science 2001, 293, 1653–1657. [Google Scholar] [CrossRef] [Green Version]
- Chen, S.; Ye, J.; Chen, X.; Shi, J.; Wu, W.; Lin, W.; Lin, W.; Li, Y.; Fu, H.; Li, S. Valproic acid attenuates traumatic spinal cord injury-induced inflammation via STAT1 and NF-κB pathway dependent of HDAC3. J. Neuroinflamm. 2018, 15, 150. [Google Scholar] [CrossRef]
- Lu, H.; Ashiqueali, R.; Lin, C.I.; Walchale, A.; Clendaniel, V.; Matheson, R.; Fisher, M.; Lo, E.H.; Selim, M.; Shehadah, A. Histone Deacetylase 3 Inhibition Decreases Cerebral Edema and Protects the Blood–Brain Barrier after Stroke. Mol. Neurobiol. 2022, 60, 235–246. [Google Scholar] [CrossRef]
- Lee, S.U.; In, H.J.; Kwon, M.S.; Park, B.-O.; Jo, M.; Kim, M.-O.; Cho, S.; Lee, S.; Lee, H.-J.; Kwak, Y.S.; et al. β-Arrestin 2 mediates G protein-coupled receptor 43 signals to nuclear factor-κB. Biol. Pharm. Bull. 2013, 36, 1754–1759. [Google Scholar] [CrossRef] [Green Version]
- Gao, H.; Sun, Y.; Wu, Y.; Luan, B.; Wang, Y.; Qu, B.; Pei, G. Identification of β-arrestin2 as a G protein-coupled receptor-stimulated regulator of NF-κB pathways. Mol. Cell 2004, 14, 303–317. [Google Scholar] [CrossRef]
- Bai, B.; Yang, Y.; Wang, Q.; Li, M.; Tian, C.; Liu, Y.; Aung, L.H.H.; Li, P.-F.; Yu, T.; Chu, X.-M. NLRP3 inflammasome in endothelial dysfunction. Cell Death Dis. 2020, 11, 776. [Google Scholar] [CrossRef]
- Chen, S.; Tang, C.; Ding, H.; Wang, Z.; Liu, X.; Chai, Y.; Jiang, W.; Han, Y.; Zeng, H. Maf1 Ameliorates Sepsis-Associated Encephalopathy by Suppressing the NF-kB/NLRP3 Inflammasome Signaling Pathway. Front. Immunol. 2020, 11, 594071. [Google Scholar] [CrossRef]
- Li, X.; Wang, C.; Zhu, J.; Lin, Q.; Yu, M.; Wen, J.; Feng, J.; Hu, C. Sodium Butyrate Ameliorates Oxidative Stress-Induced Intestinal Epithelium Barrier Injury and Mitochondrial Damage through AMPK-Mitophagy Pathway. Oxidative Med. Cell. Longev. 2022, 2022, 3745135. [Google Scholar] [CrossRef]
- Feng, Y.; Wang, Y.; Wang, P.; Huang, Y.; Wang, F. Short-Chain Fatty Acids Manifest Stimulative and Protective Effects on Intestinal Barrier Function through the Inhibition of NLRP3 Inflammasome and Autophagy. Cell. Physiol. Biochem. 2018, 49, 190–205. [Google Scholar] [CrossRef]
- Yang, C.; Hawkins, K.E.; Doré, S.; Candelario-Jalil, E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am. J. Physiol. Cell Physiol. 2019, 316, C135–C153. [Google Scholar] [CrossRef]
- Turner, R.J.; Sharp, F.R. Implications of MMP9 for Blood Brain Barrier Disruption and Hemorrhagic Transformation Following Ischemic Stroke. Front. Cell. Neurosci. 2016, 10, 56. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Estrada, E.Y.; Thompson, J.F.; Liu, W.; Rosenberg, G.A. Matrix Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. J. Cereb. Blood Flow Metab. 2007, 27, 697–709. [Google Scholar] [CrossRef]
- Li, X.-F.; Zhang, X.-J.; Zhang, C.; Wang, L.-N.; Li, Y.-R.; Zhang, Y.; He, T.-T.; Zhu, X.-Y.; Cui, L.-L.; Gao, B.-L. Ulinastatin protects brain against cerebral ischemia/reperfusion injury through inhibiting MMP-9 and alleviating loss of ZO-1 and occludin proteins in mice. Exp. Neurol. 2018, 302, 68–74. [Google Scholar] [CrossRef] [PubMed]
- Tsai, M.-M.; Chen, J.-L.; Lee, T.-H.; Liu, H.; Shanmugam, V.; Hsieh, H.-L. Brain Protective Effect of Resveratrol via Ameliorating Interleukin-1β-Induced MMP-9-Mediated Disruption of ZO-1 Arranged Integrity. Biomedicines 2022, 10, 1270. [Google Scholar] [CrossRef] [PubMed]
- Pirozzi, C.; Francisco, V.; Di Guida, F.; Gómez, R.; Lago, F.; Pino, J.; Meli, R.; Gualillo, O. Butyrate Modulates Inflammation in Chondrocytes via GPR43 Receptor. Cell. Physiol. Biochem. 2018, 51, 228–243. [Google Scholar] [CrossRef]
- Zhang, H.; Xu, J.; Wu, Q.; Fang, H.; Shao, X.; Ouyang, X.; He, Z.; Deng, Y.; Chen, C. Gut Microbiota Mediates the Susceptibility of Mice to Sepsis-Associated Encephalopathy by Butyric Acid. J. Inflamm. Res. 2022, ume 15, 2103–2119. [Google Scholar] [CrossRef]
- Aguilar, E.C.; dos Santos, L.C.; Leonel, A.J.; de Oliveira, J.S.; Santos, E.A.; Navia-Pelaez, J.M.; da Silva, J.F.; Mendes, B.P.; Capettini, L.S.; Teixeira, L.G.; et al. Oral butyrate reduces oxidative stress in atherosclerotic lesion sites by a mechanism involving NADPH oxidase down-regulation in endothelial cells. J. Nutr. Biochem. 2016, 34, 99–105. [Google Scholar] [CrossRef] [PubMed]
- Tian, Q.; Leung, F.P.; Chen, F.M.; Tian, X.Y.; Chen, Z.; Tse, G.; Ma, S.; Wong, W.T. Butyrate protects endothelial function through PPARδ/miR-181b signaling. Pharmacol. Res. 2021, 169, 105681. [Google Scholar] [CrossRef]
- Li, D.; Bai, X.; Jiang, Y.; Cheng, Y. Butyrate alleviates PTZ-induced mitochondrial dysfunction, oxidative stress and neuron apoptosis in mice via Keap1/Nrf2/HO-1 pathway. Brain Res. Bull. 2021, 168, 25–35. [Google Scholar] [CrossRef]
- Kim, S.Y.; Chae, C.W.; Lee, H.J.; Jung, Y.H.; Choi, G.E.; Kim, J.S.; Lim, J.R.; Lee, J.E.; Cho, J.H.; Park, H.; et al. Sodium butyrate inhibits high cholesterol-induced neuronal amyloidogenesis by modulating NRF2 stabilization-mediated ROS levels: Involvement of NOX2 and SOD1. Cell Death Dis. 2020, 11, 469. [Google Scholar] [CrossRef]
- Wu, P.; Tian, L.; Zhou, X.-Q.; Jiang, W.-D.; Liu, Y.; Jiang, J.; Xie, F.; Kuang, S.-Y.; Tang, L.; Tang, W.-N.; et al. Sodium butyrate enhanced physical barrier function referring to Nrf2, JNK and MLCK signaling pathways in the intestine of young grass carp (Ctenopharyngodon idella). Fish Shellfish Immunol. 2018, 73, 121–132. [Google Scholar] [CrossRef]
- Tebay, L.E.; Robertson, H.; Durant, S.T.; Vitale, S.R.; Penning, T.M.; Dinkova-Kostova, A.T.; Hayes, J.D. Mechanisms of activation of the transcription factor Nrf2 by redox stressors, nutrient cues, and energy status and the pathways through which it attenuates degenerative disease. Free Radic. Biol. Med. 2015, 88, 108–146. [Google Scholar] [CrossRef] [Green Version]
- Zhao, J.; Moore, A.N.; Redell, J.B.; Dash, P.K. Enhancing expression of Nrf2-driven genes protects the blood brain barrier after brain injury. J. Neurosci. 2007, 27, 10240–10248. [Google Scholar] [CrossRef] [Green Version]
- Alfieri, A.; Srivastava, S.; Siow, R.C.; Cash, D.; Modo, M.; Duchen, M.R.; Fraser, P.A.; Williams, S.C.; Mann, G.E. Sulforaphane preconditioning of the Nrf2/HO-1 defense pathway protects the cerebral vasculature against blood–brain barrier disruption and neurological deficits in stroke. Free. Radic. Biol. Med. 2013, 65, 1012–1022. [Google Scholar] [CrossRef]
- Prasad, S.; Sajja, R.K.; Kaisar, M.A.; Park, J.H.; Villalba, H.; Liles, T.; Abbruscato, T.; Cucullo, L. Role of Nrf2 and protective effects of Metformin against tobacco smoke-induced cerebrovascular toxicity. Redox Biol. 2017, 12, 58–69. [Google Scholar] [CrossRef]
- Zolotoff, C.; Voirin, A.-C.; Puech, C.; Roche, F.; Perek, N. Intermittent Hypoxia and Its Impact on Nrf2/HIF-1α Expression and ABC Transporters: An in Vitro Human Blood-Brain Barrier Model Study. Cell. Physiol. Biochem. 2020, 54, 1231–1248. [Google Scholar] [CrossRef]
- Sajja, R.K.; Green, K.N.; Cucullo, L. Altered Nrf2 signaling mediates hypoglycemia-induced blood-brain barrier endothelial dysfunction In Vitro. PLoS ONE 2015, 10, e0122358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, T.; Xu, S.; Wu, P.; Zhou, K.; Wu, L.; Xie, Z.; Xu, W.; Luo, X.; Li, P.; Ocak, U.; et al. Mitoquinone attenuates blood-brain barrier disruption through Nrf2/PHB2/OPA1 pathway after subarachnoid hemorrhage in rats. Exp. Neurol. 2019, 317, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Fock, E.M.; Parnova, R.G. Protective Effect of Mitochondria-Targeted Antioxidants against Inflammatory Response to Lipopolysaccharide Challenge: A Review. Pharmaceutics 2021, 13, 144. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.; Geng, J.; Gao, J.; Zhao, H.; Li, J.; Shi, Y.; Yang, B.; Xiao, C.; Linghu, Y.; Sun, X.; et al. Macrophage achieves self-protection against oxidative stress-induced ageing through the Mst-Nrf2 axis. Nat. Commun. 2019, 10, 755. [Google Scholar] [CrossRef]
- Cen, M.; Ouyang, W.; Zhang, W.; Yang, L.; Lin, X.; Dai, M.; Hu, H.; Tang, H.; Liu, H.; Xia, J.; et al. MitoQ protects against hyperpermeability of endothelium barrier in acute lung injury via a Nrf2-dependent mechanism. Redox Biol. 2021, 41, 101936. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Zhou, Q.; Li, Y.; Zhang, Y.; Wu, Y. MitoQ Modulates Lipopolysaccharide-Induced Intestinal Barrier Dysfunction via Regulating Nrf2 Signaling. Mediat. Inflamm. 2020, 2020, 3276148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hou, L.; Zhang, J.; Liu, Y.; Fang, H.; Liao, L.; Wang, Z.; Yuan, J.; Wang, X.; Sun, J.; Tang, B.; et al. MitoQ alleviates LPS-mediated acute lung injury through regulating Nrf2/Drp1 pathway. Free. Radic. Biol. Med. 2021, 165, 219–228. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Jiang, Z.; Zhang, H.; Liang, W.; Huang, W.; Zhang, H.; Li, Y.; Wang, Z.; Wang, J.; Jia, Y.; et al. Sodium butyrate attenuates diabetes-induced aortic endothelial dysfunction via P300-mediated transcriptional activation of Nrf2. Free. Radic. Biol. Med. 2018, 124, 454–465. [Google Scholar] [CrossRef] [PubMed]
- Dong, W.; Jia, Y.; Liu, X.; Zhang, H.; Li, T.; Huang, W.; Chen, X.; Wang, F.; Sun, W.; Wu, H. Sodium butyrate activates NRF2 to ameliorate diabetic nephropathy possibly via inhibition of HDAC. J. Endocrinol. 2017, 232, 71–83. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Zheng, M.; Shimoni, O.; Banks, W.A.; Bush, A.I.; Gamble, J.R.; Shi, B. Development of Novel Therapeutics Targeting the Blood–Brain Barrier: From Barrier to Carrier. Adv. Sci. 2021, 8, e2101090. [Google Scholar] [CrossRef] [PubMed]
- Brunmeir, R.; Xu, F. Functional Regulation of PPARs through Post-Translational Modifications. Int. J. Mol. Sci. 2018, 19, 1738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, X.; Ye, X.; Guo, W.; Lu, H.; Gao, Z. Inhibition of HDAC3 promotes ligand-independent PPARγ activation by protein acetylation. J. Mol. Endocrinol. 2014, 53, 191–200. [Google Scholar] [CrossRef] [Green Version]
- Mysiorek, C.; Culot, M.; Dehouck, L.; Derudas, B.; Staels, B.; Bordet, R.; Cecchelli, R.; Fenart, L.; Berezowski, V. Peroxisome Peroxisome-proliferator-activated receptor-α activation protects brain capillary endothelial cells from oxygen-glucose deprivation-induced hyperpermeability in the blood-brain barrier. Curr. Neurovasc. Res. 2009, 6, 181–193. [Google Scholar] [CrossRef]
- Hind, W.H.; England, T.J.; O’Sullivan, S.E. Cannabidiol protects an in vitro model of the blood-brain barrier from oxygen-glucose deprivation via PPARγ and 5-HT1A receptors. Br. J. Pharmacol. 2015, 173, 815–825. [Google Scholar] [CrossRef] [Green Version]
- Li, J.M.; Yu, R.; Zhang, L.P.; Wen, S.Y.; Wang, S.J.; Zhang, X.Y.; Xu, Q.; Kong, L.D. Dietary fructose-induced gut dysbiosis promotes mouse hippocampal neuroinflammation: A benefit of short-chain fatty acids. Microbiome 2019, 7, 98. [Google Scholar] [CrossRef] [Green Version]
- Hicks, K.; O’Neil, R.G.; Dubinsky, W.S.; Brown, R.C. TRPC-mediated actin-myosin contraction is critical for BBB disruption following hypoxic stress. Am. J. Physiol. Physiol. 2010, 298, C1583–C1593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cunningham, K.E.; Turner, J.R. Myosin light chain kinase: Pulling the strings of epithelial tight junction function. Ann. N. Y. Acad. Sci. 2012, 1258, 34–42. [Google Scholar] [CrossRef] [Green Version]
- Huppert, J.; Closhen, D.; Croxford, A.; White, R.; Kulig, P.; Pietrowski, E.; Bechmann, I.; Becher, B.; Luhmann, H.J.; Waisman, A.; et al. Cellular mechanisms of IL-17-induced blood-brain barrier disruption. FASEB J. 2010, 24, 1023–1034. [Google Scholar] [CrossRef] [PubMed]
- Kuhlmann, C.R.; Tamaki, R.; Gamerdinger, M.; Lessmann, V.; Behl, C.; Kempski, O.S.; Luhmann, H.J. Inhibition of the myosin light chain kinase prevents hypoxia-induced blood-brain barrier disruption. J. Neurochem. 2007, 102, 501–507. [Google Scholar] [CrossRef]
- Sun, C.; Wu, M.H.; Yuan, S.Y. Nonmuscle myosin light-chain kinase deficiency attenuates atherosclerosis in apolipoprotein E-deficient mice via reduced endothelial barrier dysfunction and monocyte migration. Circulation 2011, 124, 48–57. [Google Scholar] [CrossRef] [Green Version]
- Beard, R.S., Jr.; Haines, R.J.; Wu, K.Y.; Reynolds, J.J.; Davis, S.M.; Elliott, J.E.; Malinin, N.L.; Chatterjee, V.; Cha, B.J.; Wu, M.H.; et al. Non-muscle Mlck is required for β-catenin- and FoxO1-dependent downregulation of Cldn5 in IL-1β-mediated barrier dysfunction in brain endothelial cells. J. Cell Sci. 2014, 127, 1840–1853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miao, W.; Wu, X.; Wang, K.; Wang, W.; Wang, Y.; Li, Z.; Liu, J.; Li, L.; Peng, L. Sodium Butyrate Promotes Reassembly of Tight Junctions in Caco-2 Monolayers Involving Inhibition of MLCK/MLC2 Pathway and Phosphorylation of PKCβ2. Int. J. Mol. Sci. 2016, 17, 1696. [Google Scholar] [CrossRef] [Green Version]
- Daneman, R.; Agalliu, D.; Zhou, L.; Kuhnert, F.; Kuo, C.J.; Barres, B.A. Wnt/β-catenin signaling is required for CNS, but not non-CNS, angiogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 641–646. [Google Scholar] [CrossRef] [Green Version]
- Tran, K.A.; Zhang, X.; Predescu, D.; Huang, X.; Machado, R.; Göthert, J.; Malik, A.B.; Valyi-Nagy, T.; Zhao, Y.-Y. Endothelial β-Catenin Signaling Is Required for Maintaining Adult Blood–Brain Barrier Integrity and Central Nervous System Homeostasis. Circulation 2016, 133, 177–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liebner, S.; Corada, M.; Bangsow, T.; Babbage, J.; Taddei, A.; Czupalla, C.J.; Reis, M.; Felici, A.; Wolburg, H.; Fruttiger, M.; et al. Wnt/β-catenin signaling controls development of the blood–brain barrier. J. Cell Biol. 2008, 183, 409–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Artus, C.; Glacial, F.; Ganeshamoorthy, K.; Ziegler, N.; Godet, M.; Guilbert, T.; Liebner, S.; Couraud, P.-O. The Wnt/planar cell polarity signaling pathway contributes to the integrity of tight junctions in brain endothelial cells. J. Cereb. Blood Flow Metab. 2014, 34, 433–440. [Google Scholar] [CrossRef] [PubMed]
- Mou, Y.; Du, Y.; Zhou, L.; Yue, J.; Hu, X.; Liu, Y.; Chen, S.; Lin, X.; Zhang, G.; Xiao, H.; et al. Gut Microbiota Interact with the Brain Through Systemic Chronic Inflammation: Implications on Neuroinflammation, Neurodegeneration, and Aging. Front. Immunol. 2022, 13, 796288. [Google Scholar] [CrossRef] [PubMed]
- Huang, X.; Hussain, B.; Chang, J. Peripheral inflammation and blood–brain barrier disruption: Effects and mechanisms. CNS Neurosci. Ther. 2020, 27, 36–47. [Google Scholar] [CrossRef]
- Versele, R.; Sevin, E.; Gosselet, F.; Fenart, L.; Candela, P. TNF-α and IL-1β Modulate Blood-Brain Barrier Permeability and Decrease Amyloid-β Peptide Efflux in a Human Blood-Brain Barrier Model. Int. J. Mol. Sci. 2022, 23, 10235. [Google Scholar] [CrossRef]
- Li, M.; van Esch, B.C.; Wagenaar, G.T.; Garssen, J.; Folkerts, G.; Henricks, P.A. Pro- and anti-inflammatory effects of short chain fatty acids on immune and endothelial cells. Eur. J. Pharmacol. 2018, 831, 52–59. [Google Scholar] [CrossRef]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; Van Der Veeken, J.; DeRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [Green Version]
- Siddiqui, M.T.; Cresci, G.A.M. The Immunomodulatory Functions of Butyrate. J. Inflamm. Res. 2021, 14, 6025–6041. [Google Scholar] [CrossRef]
- Vinolo, M.A.R.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of inflammation by short chain fatty acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef] [Green Version]
- Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A.N.; Mackay, C.R.; Macia, L. The role of short-chain fatty acids in health and disease. Adv. Immunol. 2014, 121, 91–119. [Google Scholar] [CrossRef]
- Wallimann, A.; Magrath, W.; Pugliese, B.; Stocker, N.; Westermann, P.; Heider, A.; Gehweiler, D.; Zeiter, S.; Claesson, M.J.; Richards, R.G.; et al. Butyrate Inhibits Osteoclast Activity In Vitro and Regulates Systemic Inflammation and Bone Healing in a Murine Osteotomy Model Compared to Antibiotic-Treated Mice. Mediat. Inflamm. 2021, 2021, 8817421. [Google Scholar] [CrossRef]
- Eslick, S.; Williams, E.J.; Berthon, B.S.; Wright, T.; Karihaloo, C.; Gately, M.; Wood, L.G. Weight Loss and Short-Chain Fatty Acids Reduce Systemic Inflammation in Monocytes and Adipose Tissue Macrophages from Obese Subjects. Nutrients 2022, 14, 765. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Frister, A.; Schmidt, C.; Schneble, N.; Brodhun, M.; Gonnert, F.A.; Bauer, M.; Hirsch, E.; Müller, J.P.; Wetzker, R.; Bauer, R. Phosphoinositide 3-kinase γ affects LPS-induced disturbance of blood-brain barrier via lipid kinase-independent control of cAMP in microglial cells. NeuroMol. Med. 2014, 16, 704–713. [Google Scholar] [CrossRef] [PubMed]
- Peng, X.; Luo, Z.; He, S.; Zhang, L.; Li, Y. Blood-Brain Barrier Disruption by Lipopolysaccharide and Sepsis-Associated Encephalopathy. Front. Cell. Infect. Microbiol. 2021, 11, 768108. [Google Scholar] [CrossRef]
- Fu, S.-P.; Wang, J.-F.; Xue, W.-J.; Liu, H.-M.; Liu, B.-R.; Zeng, Y.-L.; Li, S.-N.; Huang, B.-X.; Lv, Q.-K.; Wang, W.; et al. Anti-inflammatory effects of BHBA in both in vivo and in vitro Parkinson’s disease models are mediated by GPR109A-dependent mechanisms. J. Neuroinflamm. 2015, 12, 9. [Google Scholar] [CrossRef] [Green Version]
- Zhang, S.; Zhan, L.; Li, X.; Yang, Z.; Luo, Y.; Zhao, H. Preclinical and clinical progress for HDAC as a putative target for epigenetic remodeling and functionality of immune cells. Int. J. Biol. Sci. 2021, 17, 3381–3400. [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, 1–17. [Google Scholar] [CrossRef] [Green Version]
- Ziabska, K.; Gargas, J.; Sypecka, J.; Ziemka-Nalecz, M. The impact of the histone deacetylase inhibitor sodium butyrate on microglial polarization after oxygen and glucose deprivation. Pharmacol. Rep. 2022, 74, 909–919. [Google Scholar] [CrossRef]
- Patnala, R.; Arumugam, T.V.; Gupta, N.; Dheen, S.T. HDAC Inhibitor Sodium Butyrate-Mediated Epigenetic Regulation Enhances Neuroprotective Function of Microglia during Ischemic Stroke. Mol. Neurobiol. 2016, 54, 6391–6411. [Google Scholar] [CrossRef]
- Kim, H.J.; Rowe, M.; Ren, M.; Hong, J.S.; Chen, P.S.; Chuang, D.-M. Histone deacetylase inhibitors exhibit anti-inflammatory and neuroprotective effects in a rat permanent ischemic model of stroke: Multiple mechanisms of action. J. Pharmacol. Exp. Ther. 2007, 321, 892–901. [Google Scholar] [CrossRef] [PubMed]
- Huuskonen, J.; Suuronen, T.; Nuutinen, T.; Kyrylenko, S.; Salminen, A. Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids. Br. J. Pharmacol. 2004, 141, 874–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.; Li, H.; Gong, T.; Chen, W.; Mao, S.; Kong, Y.; Yu, J.; Sun, J. Anti-neuroinflammatory Effect of Short-Chain Fatty Acid Acetate against Alzheimer’s Disease via Upregulating GPR41 and Inhibiting ERK/JNK/NF-κB. J. Agric. Food Chem. 2020, 68, 7152–7161. [Google Scholar] [CrossRef]
- Song, L.; Sun, Q.; Zheng, H.; Zhang, Y.; Wang, Y.; Liu, S.; Duan, L. Roseburia hominis Alleviates Neuroinflammation via Short-Chain Fatty Acids through Histone Deacetylase Inhibition. Mol. Nutr. Food Res. 2022, 66, 2200164. [Google Scholar] [CrossRef]
- Gao, Y.; Xie, D.; Wang, Y.; Niu, L.; Jiang, H. Short-Chain Fatty Acids Reduce Oligodendrocyte Precursor Cells Loss by Inhibiting the Activation of Astrocytes via the SGK1/IL-6 Signalling Pathway. Neurochem. Res. 2022, 47, 3476–3489. [Google Scholar] [CrossRef] [PubMed]
- Munji, R.N.; Soung, A.L.; Weiner, G.A.; Sohet, F.; Semple, B.D.; Trivedi, A.; Gimlin, K.; Kotoda, M.; Korai, M.; Aydin, S.; et al. Profiling the mouse brain endothelial transcriptome in health and disease models reveals a core blood–brain barrier dysfunction module. Nat. Neurosci. 2019, 22, 1892–1902. [Google Scholar] [CrossRef] [PubMed]
Method | SCFA | Model | Dose and Mode of SCFA Administration | Effect on BBB | Reference |
---|---|---|---|---|---|
In vitro | butyrate | Endothelial/glial cells co-culture | 20–80 μM, 6 h incubation | ↑TEER | [75] |
In vitro | butyrate, propionate | hCMEC/D3 cells, LPS | 1 μM, 24 h | ↑TEER, ↓permeability (FITC-labeled dextran) | [33] |
In vitro | propionate | hCMEC/D3 cells, LPS | 1 μM, 24 h | ↓NFκB, ↓TLR4 signaling pathways, ↑Nrf2-related protein expression, ↑Nrf2 translocation to the nucleus, ↑occludin, claudin-5 mRNA and protein level, ↓ROS | [33] |
In vitro | butyrate, propionate | bEnd.3 cells, LPS | 1 μM, 24 h | ↑TEER, ↑ZO-1 and claudin-5 localization at the cell–cell junctions, ↑area occupied by mitochondria, no changes in occludin, claudin-5 and ZO-1 mRNA level | [76] |
In vivo | butyrate | Stroke (MCAO) | 300 mg/kg, i.p. | ↓permeability (Evans blue extravasation), ↓NF-κB, ↓MMP-9, ↑claudin-5, ZO-1 protein level | [77] |
In vivo | butyrate | Stroke (MCAO) | 300 mg/kg, i.p. | ↓permeability (serum levels of glial fibrillary acidic protein) | [29] |
In vivo | butyrate | A mouse model of Parkinson’s disease | 200 mg/kg, i.g. | ↑occludin, ZO-1 protein level | [32] |
In vivo | butyrate | Traumatic brain injury, mice | 200 mg/kg, i.p. | ↓permeability (Evans blue extravasation) ↑occludin, ZO-1 protein level | [30] |
In vivo | acetate: propionate: butyrate 3:1:1 | Sepsis (cecal ligation and puncture), mice | 500 mg/kg, i.g. | ↑occludin, ZO-1 protein level | [31] |
In vivo | butyrate | Postoperative aged mice | i.g., (dose is not indicated) | ↓permeability (FITC-labeled dextran), ↑occludin, claudin-5, ZO-1 protein level | [71] |
In vivo | SCFAs mixture | High-fructose diet + chronic stress, mice | supplemented to feed, (dose is not indicated) | ↓permeability (FITC-labeled dextran) ↑occludin, claudin-5, ZO-1 protein level | [7] |
In vivo | monobutyrin, monovalerin | ApoE-/- rats + high-fat diet | supplemented to feed in a dose of 1% | ↑occludin protein, ↑occludin, ZO-1 protein level | [78] |
In vivo | butyrate | Germ-free mice | 1 g/kg, i.g. | ↓permeability (Evans blue extravasation, [11C]raclopride) ↑occludin protein level | [25] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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
Fock, E.; Parnova, R. Mechanisms of Blood–Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids. Cells 2023, 12, 657. https://doi.org/10.3390/cells12040657
Fock E, Parnova R. Mechanisms of Blood–Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids. Cells. 2023; 12(4):657. https://doi.org/10.3390/cells12040657
Chicago/Turabian StyleFock, Ekaterina, and Rimma Parnova. 2023. "Mechanisms of Blood–Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids" Cells 12, no. 4: 657. https://doi.org/10.3390/cells12040657
APA StyleFock, E., & Parnova, R. (2023). Mechanisms of Blood–Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids. Cells, 12(4), 657. https://doi.org/10.3390/cells12040657