3,427
Views
63
CrossRef citations to date
0
Altmetric
Review

Microbiota and the control of blood-tissue barriers

&
Article: e1039691 | Received 05 Feb 2015, Accepted 03 Apr 2015, Published online: 15 Jul 2015

References

  • Ribbert H. Die Abscheidung intravenos injizierten gelosten Karmins in den Geweben. Z Allgem Physiol 1904; 4:201-14
  • Goldmann EE. [Die aüßere und innere Sekretion des gesunden und kranken Organismus im Lichte der “vitalen Farbung”]. Beitr Klin Chir 1909; 64:192-265
  • Cheng CY, Mruk DD. The blood-testis barrier and its implications for male contraception. Pharmacol Rev 2012; 64:16-64; PMID:22039149; http://dx.doi.org/10.1124/pr.110.002790
  • Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci 2006; 7:41-53; PMID:16371949; http://dx.doi.org/10.1038/nrn1824
  • Ransom BR. The neuronal microenvironment. In: Boron WF, Boulpaep, E., ed. Medical Physiology (BORON). Philadelphia: Saunders, 2009:298-301
  • Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 2010; 468:562-6; PMID:20944625; http://dx.doi.org/10.1038/nature09513
  • Ben-Zvi A, Lacoste B, Kur E, Andreone BJ, Mayshar Y, Yan H, Gu C. Mfsd2a is critical for the formation and function of the blood-brain barrier. Nature 2014; 509:507-11; PMID:24828040; http://dx.doi.org/10.1038/nature13324
  • Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood-brain barrier. Nat Med 2013; 19:1584-96; PMID:24309662; http://dx.doi.org/10.1038/nm.3407
  • Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiol Dis 2010; 37:13-25; PMID:19664713; http://dx.doi.org/10.1016/j.nbd.2009.07.030
  • Stewart PA, Wiley MJ. Developing nervous tissue induces formation of blood-brain barrier characteristics in invading endothelial cells: a study using quail–chick transplantation chimeras. Dev Biol 1981; 84:183-92; PMID:7250491; http://dx.doi.org/10.1016/0012-1606(81)90382-1
  • Igarashi Y, Utsumi H, Chiba H, Yamada-Sasamori Y, Tobioka H, Kamimura Y, Furuuchi K, Kokai Y, Nakagawa T, Mori M, et al. Glial cell line-derived neurotrophic factor induces barrier function of endothelial cells forming the blood-brain barrier. Biochem Biophy Res Commun 1999; 261:108-12; PMID:10405331; http://dx.doi.org/10.1006/bbrc.1999.0992
  • Armulik A, Genove G, Mae M, Nisancioglu MH, Wallgard E, Niaudet C, He L, Norlin J, Lindblom P, Strittmatter K, et al. Pericytes regulate the blood-brain barrier. Nature 2010; 468:557-61; PMID:20944627; http://dx.doi.org/10.1038/nature09522
  • Zenker D, Begley D, Bratzke H, Rubsamen-Waigmann H, von Briesen H. Human blood-derived macrophages enhance barrier function of cultured primary bovine and human brain capillary endothelial cells. J Physiol 2003; 551:1023-32; PMID:12829721; http://dx.doi.org/10.1113/jphysiol.2003.045880
  • Abbott NJ. Dynamics of CNS barriers: evolution, differentiation, and modulation. Cell Mol Neurobiol 2005; 25:5-23; PMID:15962506; http://dx.doi.org/10.1007/s10571-004-1374-y
  • Ploen L, Setchell BP. Blood-testis barriers revisited. A homage to Lennart Nicander. Int J Androl 1992; 15:1-4; PMID:1544694; http://dx.doi.org/10.1111/j.1365-2605.1992.tb01108.x
  • Holash JA, Harik SI, Perry G, Stewart PA. Barrier properties of testis microvessels. Proc Natl Acad Sci U S A 1993; 90:11069-73; PMID:7902579; http://dx.doi.org/10.1073/pnas.90.23.11069
  • Mok KW, Lie PP, Mruk DD, Mannu J, Mathur PP, Silvestrini B, Cheng CY. The apical ectoplasmic specialization-blood-testis barrier functional axis is a novel target for male contraception. Adv Exp Med Biol 2012; 763:334-55; PMID:23397633
  • Cyr DG, Hermo L, Egenberger N, Mertineit C, Trasler JM, Laird DW. Cellular immunolocalization of occludin during embryonic and postnatal development of the mouse testis and epididymis. Endocrinology 1999; 140:3815-25; PMID:10433243
  • Cheng CY, Wong EW, Lie PP, Li MW, Mruk DD, Yan HH, Mok KW, Mannu J, Mathur PP, Lui WY, et al. Regulation of blood-testis barrier dynamics by desmosome, gap junction, hemidesmosome and polarity proteins: An unexpected turn of events. Spermatogenesis 2011; 1:105-15; PMID:22319658; http://dx.doi.org/10.4161/spmg.1.2.15745
  • Orth JM. Proliferation of Sertoli cells in fetal and postnatal rats: a quantitative autoradiographic study. Anatom Rec 1982; 203:485-92; PMID:7137603; http://dx.doi.org/10.1002/ar.1092030408
  • Petersen C, Soder O. The sertoli cell–a hormonal target and ‘super’ nurse for germ cells that determines testicular size. Horm Res 2006; 66:153-61; PMID:16804315; http://dx.doi.org/10.1159/000094142
  • Sharpe RM, McKinnell C, Kivlin C, Fisher JS. Proliferation and functional maturation of Sertoli cells, and their relevance to disorders of testis function in adulthood. Reproduction 2003; 125:769-84; PMID:12773099; http://dx.doi.org/10.1530/rep.0.1250769
  • Tarulli GA, Stanton PG, Meachem SJ. Is the adult Sertoli cell terminally differentiated? Biol Reprod 2012; 87:13, 1-1; http://dx.doi.org/10.1095/biolreprod.111.095091
  • Ahmed EA, Barten-van Rijbroek AD, Kal HB, Sadri-Ardekani H, Mizrak SC, van Pelt AM, de Rooij DG. Proliferative activity in vitro and DNA repair indicate that adult mouse and human Sertoli cells are not terminally differentiated, quiescent cells. Biol Reprod 2009; 80:1084-91; PMID:19164176; http://dx.doi.org/10.1095/biolreprod.108.071662
  • Mital P, Hinton BT, Dufour JM. The blood-testis and blood-epididymis barriers are more than just their tight junctions. Biol Reprod 2011; 84:851-8; PMID:21209417; http://dx.doi.org/10.1095/biolreprod.110.087452
  • Wong V, Russell LD. Three-dimensional reconstruction of a rat stage V Sertoli cell: I. Methods, basic configuration, and dimensions. Am J Anat 1983; 167:143-61; PMID:6351582; http://dx.doi.org/10.1002/aja.1001670202
  • Xia W, Mruk DD, Lee WM, Cheng CY. Cytokines and junction restructuring during spermatogenesis–a lesson to learn from the testis. Cytokine Growth Factor Rev 2005; 16:469-93; PMID:16023885; http://dx.doi.org/10.1016/j.cytogfr.2005.05.007
  • Cheng CY, Mruk DD. A local autocrine axis in the testes that regulates spermatogenesis. Nat Rev Endocrinol 2010; 6:380-95; PMID:20571538; http://dx.doi.org/10.1038/nrendo.2010.71
  • Li MW, Mruk DD, Lee WM, Cheng CY. Cytokines and junction restructuring events during spermatogenesis in the testis: an emerging concept of regulation. Cytokine Growth Factor Rev 2009; 20:329-38; PMID:19651533; http://dx.doi.org/10.1016/j.cytogfr.2009.07.007
  • Wong CH, Cheng CY. The blood-testis barrier: its biology, regulation, and physiological role in spermatogenesis. Curr Top Dev Biol 2005; 71:263-96; PMID:16344108; http://dx.doi.org/10.1016/S0070-2153(05)71008-5
  • Russell LD, Peterson RN. Sertoli cell junctions: morphological and functional correlates. Int Rev Cytol 1985; 94:177-211; PMID:3894273; http://dx.doi.org/10.1016/S0074-7696(08)60397-6
  • Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, Furuse M, Tsukita S. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J Cell Biol 2003; 161:653-60; PMID:12743111; http://dx.doi.org/10.1083/jcb.200302070
  • Saitou M, Furuse M, Sasaki H, Schulzke JD, Fromm M, Takano H, Noda T, Tsukita S. Complex phenotype of mice lacking occludin, a component of tight junction strands. Mol Biol Cell 2000; 11:4131-42; PMID:11102513; http://dx.doi.org/10.1091/mbc.11.12.4131
  • Mazaud-Guittot S, Meugnier E, Pesenti S, Wu X, Vidal H, Gow A, Le Magueresse-Battistoni B. Claudin 11 deficiency in mice results in loss of the Sertoli cell epithelial phenotype in the testis. Biol Reprod 2010; 82:202-13; PMID:19741204; http://dx.doi.org/10.1095/biolreprod.109.078907
  • Chihara M, Ikebuchi R, Otsuka S, Ichii O, Hashimoto Y, Suzuki A, Saga Y, Kon Y. Mice stage-specific claudin 3 expression regulates progression of meiosis in early stage spermatocytes. Biol Reprod 2013; 89:3; PMID:23677978; http://dx.doi.org/10.1095/biolreprod.113.107847
  • Sandoval KE, Witt KA. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis 2008; 32:200-19; PMID:18790057; http://dx.doi.org/10.1016/j.nbd.2008.08.005
  • Correale J, Villa A. The blood-brain-barrier in multiple sclerosis: functional roles and therapeutic targeting. Autoimmunity 2007; 40:148-60; PMID:17453713; http://dx.doi.org/10.1080/08916930601183522
  • Erickson MA, Banks WA. Blood-brain barrier dysfunction as a cause and consequence of Alzheimer's disease. J Cereb Blood Flow Metab 2013; 33:1500-13; PMID:23921899; http://dx.doi.org/10.1038/jcbfm.2013.135
  • Fijak M, Meinhardt A. The testis in immune privilege. Immunol Rev 2006; 213:66-81; PMID:16972897; http://dx.doi.org/10.1111/j.1600-065X.2006.00438.x
  • Alves MG, Oliveira PF, Socorro S, Moreira PI. Impact of diabetes in blood-testis and blood-brain barriers: resemblances and differences. Curr Diab Rev 2012; 8:401-12; PMID:22934551; http://dx.doi.org/10.2174/157339912803529896
  • Lee JY, Huerta PT, Zhang J, Kowal C, Bertini E, Volpe BT, Diamond B. Neurotoxic autoantibodies mediate congenital cortical impairment of offspring in maternal lupus. Nat Med 2009; 15:91-6; PMID:19079257; http://dx.doi.org/10.1038/nm.1892
  • Carson MJ, Doose JM, Melchior B, Schmid CD, Ploix CC. CNS immune privilege: hiding in plain sight. Immunol Rev 2006; 213:48-65; PMID:16972896; http://dx.doi.org/10.1111/j.1600-065X.2006.00441.x
  • Moens E, Veldhoen M. Epithelial barrier biology: good fences make good neighbours. Immunology 2012; 135:1-8; PMID:22044254; http://dx.doi.org/10.1111/j.1365-2567.2011.03506.x
  • Korecka A, Arulampalam V. The gut microbiome: scourge, sentinel or spectator? J Oral Microbiol 2012; 4:9367-80; PMID:22368769; http://dx.doi.org/10.3402/jom.v4i0.9367
  • Al-Asmakh M, Anuar F, Zadjali F, Rafter J, Pettersson S. Gut microbial communities modulating brain development and function. Gut Microbes 2012; 3:366-73; PMID:22743758; http://dx.doi.org/10.4161/gmic.21287
  • Stappenbeck TS, Hooper LV, Gordon JI. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc Natl Acad Sci U S A 2002; 99:15451-5; PMID:12432102; http://dx.doi.org/10.1073/pnas.202604299
  • Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci U S A 2011; 108:5354-9; PMID:21402903; http://dx.doi.org/10.1073/pnas.1019378108
  • Diaz Heijtz R, Wang S, Anuar F, Qian Y, Bjorkholm B, Samuelsson A, Hibberd ML, Forssberg H, Pettersson S. Normal gut microbiota modulates brain development and behavior. Proceed Natl Acad Sci U S A 2011; 108:3047-52; PMID:21282636; http://dx.doi.org/10.1073/pnas.1010529108
  • Amaral FA, Sachs D, Costa VV, Fagundes CT, Cisalpino D, Cunha TM, Ferreira SH, Cunha FQ, Silva TA, Nicoli JR, et al. Commensal microbiota is fundamental for the development of inflammatory pain. Proc Natl Acad Sci U S A 2008; 105:2193-7; PMID:18268332; http://dx.doi.org/10.1073/pnas.0711891105
  • Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science 2012; 336:1262-7; PMID:22674330; http://dx.doi.org/10.1126/science.1223813
  • Sjogren K, Engdahl C, Henning P, Lerner UH, Tremaroli V, Lagerquist MK, Backhed F, Ohlsson C. The gut microbiota regulates bone mass in mice. J Bone Miner Res 2012; 27:1357-67; PMID:22407806; http://dx.doi.org/10.1002/jbmr.1588
  • Wilson ID. Drugs, bugs, and personalized medicine: pharmacometabonomics enters the ring. Proc Natl Acad Sci U S A 2009; 106:14187-8; PMID:19706501; http://dx.doi.org/10.1073/pnas.0907721106
  • Bjorkholm B, Bok CM, Lundin A, Rafter J, Hibberd ML, Pettersson S. Intestinal microbiota regulate xenobiotic metabolism in the liver. PloS One 2009; 4:e6958; PMID:19742318; http://dx.doi.org/10.1371/journal.pone.0006958
  • Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiol Rev 2001; 81:1031-64; PMID:11427691
  • Bordin M, D'Atri F, Guillemot L, Citi S. Histone deacetylase inhibitors up-regulate the expression of tight junction proteins. Mol Cancer Res 2004; 2:692-701; PMID:15634758
  • Meijer K, de Vos P, Priebe MG. Butyrate and other short-chain fatty acids as modulators of immunity: what relevance for health? Curr Opin Clin Nutr Metab Care 2010; 13:715-21; PMID:20823773; http://dx.doi.org/10.1097/MCO.0b013e32833eebe5
  • Kles KA, Chang EB. Short-chain fatty acids impact on intestinal adaptation, inflammation, carcinoma, and failure. Gastroenterology 2006; 130:S100-5; PMID:16473056; http://dx.doi.org/10.1053/j.gastro.2005.11.048
  • Cook SI, Sellin JH. Review article: short chain fatty acids in health and disease. Aliment Pharmacol Ther 1998; 12:499-507; PMID:9678808; http://dx.doi.org/10.1046/j.1365-2036.1998.00337.x
  • Ganapathy V, Thangaraju M, Prasad PD, Martin PM, Singh N. Transporters and receptors for short-chain fatty acids as the molecular link between colonic bacteria and the host. Curr Opin Pharmacol 2013; 13:869-74; PMID:23978504; http://dx.doi.org/10.1016/j.coph.2013.08.006
  • Bollrath J, Powrie F. Immunology. Feed your Tregs more fiber. Science 2013; 341:463-4; PMID:23908210; http://dx.doi.org/10.1126/science.1242674
  • Grunstein M. Histone acetylation in chromatin structure and transcription. Nature 1997; 389:349-52; PMID:9311776; http://dx.doi.org/10.1038/38664
  • Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, Muir AI, Wigglesworth MJ, Kinghorn I, Fraser NJ, 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; 278:11312-9; PMID:12496283; http://dx.doi.org/10.1074/jbc.M211609200
  • Ichimura A, Hirasawa A, Hara T, Tsujimoto G. Free fatty acid receptors act as nutrient sensors to regulate energy homeostasis. Prostaglandins Other Lipid Mediat 2009; 89:82-8; PMID:19460454; http://dx.doi.org/10.1016/j.prostaglandins.2009.05.003
  • Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009; 461:1282-6; PMID:19865172; http://dx.doi.org/10.1038/nature08530
  • Lievin-Le Moal V, Servin AL. The front line of enteric host defense against unwelcome intrusion of harmful microorganisms: mucins, antimicrobial peptides, and microbiota. Clin Microbiol Rev 2006; 19:315-37; PMID:16614252; http://dx.doi.org/10.1128/CMR.19.2.315-337.2006
  • Turner JR, Buschmann MM, Romero-Calvo I, Sailer A, Shen L. The role of molecular remodeling in differential regulation of tight junction permeability. Semin Cell Dev Biol 2014; 36:204-12; PMID:25263012; http://dx.doi.org/10.1016/j.semcdb.2014.09.022
  • Goncalves A, Ambrosio AF, Fernandes R. Regulation of claudins in blood-tissue barriers under physiological and pathological states. Tissue Barriers 2013; 1:e24782; PMID:24665399; http://dx.doi.org/10.4161/tisb.24782
  • Groschwitz KR, Hogan SP. Intestinal barrier function: molecular regulation and disease pathogenesis. J Allergy Clin Immunol 2009; 124:3-20; quiz 1–2; PMID:19560575; http://dx.doi.org/10.1016/j.jaci.2009.05.038
  • Huang XZ, Zhu LB, Li ZR, Lin J. Bacterial colonization and intestinal mucosal barrier development. World J Clin Pediatr 2013; 2:46-53; PMID:25254174; http://dx.doi.org/10.5409/wjcp.v2.i4.46
  • Sharma R, Young C, Neu J. Molecular modulation of intestinal epithelial barrier: contribution of microbiota. J Biomed Biotechnol 2010; 2010:305879; PMID:20150966
  • Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M, Korecka A, Bakocevic N, Guan NL, Kundu P, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 2014; 6:263ra158; http://dx.doi.org/10.1126/scitranslmed.3009759
  • Al-Asmakh M, Stukenborg JB, Reda A, Anuar F, Strand ML, Hedin L, Pettersson S, Soder O. The gut microbiota and developmental programming of the testis in mice. PloS One 2014; 9:e103809; PMID:25118984; http://dx.doi.org/10.1371/journal.pone.0103809
  • Mercado J, Valenzano MC, Jeffers C, Sedlak J, Cugliari MK, Papanikolaou E, Clouse J, Miao J, Wertan NE, Mullin JM. Enhancement of tight junctional barrier function by micronutrients: compound-specific effects on permeability and claudin composition. PloS One 2013; 8:e78775; PMID:24236048; http://dx.doi.org/10.1371/journal.pone.0078775
  • Andrade-Oliveira V, Amano MT, Correa-Costa M, Castoldi A, Felizardo RJ, de Almeida DC, Bassi EJ, Moraes-Vieira PM, Hiyane MI, Rodas AC, et al. Gut Bacteria Products Prevent AKI Induced by Ischemia-Reperfusion. J A Soc Nephrol 2015; PMID:25589612

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.