Histamine derived from probiotic Lactobacillus reuteri suppresses TNF via modulation of PKA and ERK signaling

Carissa M Thomas, Teresa Hong, Jan Peter van Pijkeren, Peera Hemarajata, Dan V Trinh, Weidong Hu, Robert A Britton, Markus Kalkum, James Versalovic, Carissa M Thomas, Teresa Hong, Jan Peter van Pijkeren, Peera Hemarajata, Dan V Trinh, Weidong Hu, Robert A Britton, Markus Kalkum, James Versalovic

Abstract

Beneficial microbes and probiotic species, such as Lactobacillus reuteri, produce biologically active compounds that can modulate host mucosal immunity. Previously, immunomodulatory factors secreted by L. reuteri ATCC PTA 6475 were unknown. A combined metabolomics and bacterial genetics strategy was utilized to identify small compound(s) produced by L. reuteri that were TNF-inhibitory. Hydrophilic interaction liquid chromatography-high performance liquid chromatography (HILIC-HPLC) separation isolated TNF-inhibitory compounds, and HILIC-HPLC fraction composition was determined by NMR and mass spectrometry analyses. Histamine was identified and quantified in TNF-inhibitory HILIC-HPLC fractions. Histamine is produced from L-histidine via histidine decarboxylase by some fermentative bacteria including lactobacilli. Targeted mutagenesis of each gene present in the histidine decarboxylase gene cluster in L. reuteri 6475 demonstrated the involvement of histidine decarboxylase pyruvoyl type A (hdcA), histidine/histamine antiporter (hdcP), and hdcB in production of the TNF-inhibitory factor. The mechanism of TNF inhibition by L. reuteri-derived histamine was investigated using Toll-like receptor 2 (TLR2)-activated human monocytoid cells. Bacterial histamine suppressed TNF production via activation of the H(2) receptor. Histamine from L. reuteri 6475 stimulated increased levels of cAMP, which inhibited downstream MEK/ERK MAPK signaling via protein kinase A (PKA) and resulted in suppression of TNF production by transcriptional regulation. In summary, a component of the gut microbiome, L. reuteri, is able to convert a dietary component, L-histidine, into an immunoregulatory signal, histamine, which suppresses pro-inflammatory TNF production. The identification of bacterial bioactive metabolites and their corresponding mechanisms of action with respect to immunomodulation may lead to improved anti-inflammatory strategies for chronic immune-mediated diseases.

Conflict of interest statement

Competing Interests: James Versalovic receives unrestricted research support from BioGaia AB. This does not alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Identification of histamine in TNF-inhibitory…
Figure 1. Identification of histamine in TNF-inhibitory fractions of L. reuteri 6475 by NMR.
1H NMR analysis demonstrated differences in the composition of HILIC-HPLC fractions from L. reuteri 6475. A. Top and bottom spectra show the 1D 1H NMR of TNF-inhibitory fraction B3 and fraction B4, respectively. The assigned peaks of phenylalanine (Phe), tryptophan (Trp), and histamine from fraction B3 are labeled on the top spectrum. Two complementary 2D NMR techniques were used to identify compounds unique to TNF-inhibitory fraction B3. B. Aromatic peaks of 1H-13C-HSQC from Phe, Trp, and histamine. C. Aliphatic peaks of 1H-13C-HSQC from Phe, Trp, and histamine. D. The TOCSY spectrum further identified the spin system of Phe, Trp, and histamine unique to TNF-inhibitory fraction B3.
Figure 2. The production of histamine by…
Figure 2. The production of histamine by L. reuteri 6475 was confirmed by mass spectrometry.
Examination of TNF-inhibitory HILIC-HPLC fractions from L. reuteri 6475 grown in LDMIIIG confirmed the presence of histamine by ESI qTOF MS. A. ESI qTOF MS of TNF-inhibitory HILIC-HPLC fraction B3 from strain 6475 grown in glucose. Histamine was detected at m/z 112.08. B. ESI qTOF MS of non-TNF inhibitory HILIC-HPLC fraction B3 from strain 6475 grown in sucrose. Histamine was not detected at m/z 112.08. C. MS/MS analysis of m/z 112.08 confirmed that the compound is histamine. D.L. reuteri culture supernatants from sucrose or glucose-containing media were analyzed for histamine concentration by triple quadrupole mass spectrometry. Histamine (pg/mL) was detected only in culture supernatants from glucose-containing medium cultures.
Figure 3. Quantification of histamine by triple…
Figure 3. Quantification of histamine by triple quadrupole mass spectrometry.
Histamine in HILIC-HPLC fractions B2–B7 from L. reuteri 6475 grown in a glucose-containing medium was quantified by triple quadrupole mass spectrometry. The y-axis indicates TNF production from activated human monocytoid cells. The concentration of histamine (pg/µL) is indicated above each bar graph. Elevated levels of histamine correlated with TNF inhibition.
Figure 4. The histidine decarboxylase gene cluster…
Figure 4. The histidine decarboxylase gene cluster contributed to the TNF-inhibitory phenotype of L. reuteri 6475.
The histidine decarboxylase gene cluster consists of three genes, hdcP (RPRB3004), hdcA (RPRB3005), and hdcB (RPRB3006). The organization of the cluster is depicted above the bar graph. Inactivation by targeted mutagenesis in any one of these genes resulted in a partial loss (approximately 40%) of TNF suppression by L. reuteri 6475. Results represent the mean ± SD (n = 9), *p value<0.05 compared to medium control, #p value<0.05 compared to L. reuteri 6475.
Figure 5. L. reuteri -derived histamine inhibited…
Figure 5. L. reuteri-derived histamine inhibited TNF production via the H2 receptor.
A. Histamine inhibited TNF production, and suppression of TNF was blocked by specific H2 receptor antagonists in a concentration-dependent manner. L. reuteri 6475 conditioned media (CM) containing histamine inhibited TNF production, an effect partially blocked by specific H2R antagonists. The presence or absence of histamine (10 nM) or L. reuteri 6475 CM is indicated by a “+” or “−” respectively. Colored bars indicate treatment with select antagonists - ranitidine (H2R; blue), cimetidine (H2R; green), chlorpheniramine (H1R; red), and pyrilamine (H1R; purple). H1R antagonists were tested at 100 nM. Results represent the mean ± SD (n = 3), *p value<0.05 compared to medium control, #p value<0.05 compared to histamine, ##p value<0.05 compared to L. reuteri 6475 CM. B. TFA-treated cell pellets containing histamine suppressed TNF production, an effect partially blocked by specific H2R antagonists. HILIC-HPLC fraction B3, which was enriched for histamine, inhibited TNF production, an effect completely blocked by specific H2R antagonists. All antagonists were tested at 100 nM. The presence or absence of L. reuteri 6475 CP or Fraction B3 is indicated by a “+” or “−” respectively. Results represent the mean ± SD (n = 3), *p value<0.05 compared to medium control, #p value<0.05 compared to L. reuteri 6475 CP, ##p value<0.05 compared to fraction B3. C. Histamine receptor antagonists alone did not affect TNF production. All antagonists were tested at 100 nM. The H1R antagonist, chlorpheniramine (CPM), did not prevent the H2R antagonist, ranitidine (RAN), from blocking TNF suppression by histamine (10 nM) and L. reuteri 6475 CM. Blue bars with red stripes indicate combined treatment with CPM plus RAN. Results represent the mean ± SD (n = 3), *p value<0.05 compared to medium control, #p value<0.05 compared to histamine, ##p value<0.05 compared to L. reuteri 6475 CM.
Figure 6. Stimulation of the H 2…
Figure 6. Stimulation of the H2 receptor increased cAMP and PKA activity was required for TNF suppression.
Signaling pathway studies were performed to determine the effect of histamine on cAMP production and the role of PKA activity in TNF suppression. A. Treatment of activated THP-1 cells with L. reuteri 6475 CM or histamine increased intracellular cAMP. The increase in cAMP was blocked by a specific H2 receptor antagonist, ranitidine. Results represent the mean ± SD (n = 2), *p value<0.05 compared to medium control, **p value<0.05 compared to L. reuteri 6475, #p value<0.05 compared to histamine. B. Inhibition of PKA activity partially blocked TNF suppression by TNF-inhibitory compounds L. reuteri 6475 CM, HILIC-HPLC fraction B3, histamine, and dcAMP. Results represent the mean ± SD (n = 9), *p value<0.05 compared to H89 treated samples.
Figure 7. Stimulation of the H 2…
Figure 7. Stimulation of the H2 receptor inhibited the ERK signaling pathway.
Immunoblot studies were performed to determine the effect of histamine on activation of the ERK signaling pathway. ERK was activated by phosphorylation on Thr202/Tyr204 (P-ERK). Treatment of TLR-stimulated THP-1 cells with L. reuteri 6475, histamine, or U0126 suppressed activation of ERK1/2, an effect that was significantly blocked in the presence of ranitidine. The β-actin antibody served as the protein loading control.
Figure 8. Pathway diagram of the mechanism…
Figure 8. Pathway diagram of the mechanism of histamine inhibition of ERK activation.
THP-1 cells activated with a TLR2 agonist activated the MEK/ERK MAPK signaling cascade to induce production of TNF. Histamine produced by L. reuteri 6475 engaged the H2 receptor on activated THP-1 cells. This G protein-coupled receptor activated adenylate cyclase (AC) to increase intracellular cAMP, which subsequently activated protein kinase A (PKA). PKA inhibited MEK/ERK activation and suppressed production of TNF. It was predicted that PKA inhibited MEK/ERK activation by inhibiting the activity of c-Raf. Specific H2 receptor antagonists, such as ranitidine, prevented histamine from activating the H2 receptor and blocked TNF suppression by histamine. H89, a specific inhibitor of PKA, prevented TNF suppression by histamine, demonstrating that PKA activity was necessary for histamine's effect on TNF. Blocking MEK activity with U0126 suppressed activation of ERK and downstream TNF production, demonstrating that ERK activation was necessary for production of TNF.

References

    1. Yan F, Cao H, Cover TL, Whitehead R, Washington MK, et al. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology. 2007;132:562–575.
    1. Clarke MB, Sperandio V. Events at the host-microbial interface of the gastrointestinal tract III. Cell-to-cell signaling among microbial flora, host, and pathogens: there is a whole lot of talking going on. Am J Physiol Gastrointest Liver Physiol. 2005;288:G1105–1109.
    1. Reuter G. The Lactobacillus and Bifidobacterium microflora of the human intestine: composition and succession. Curr Issues Intest Microbiol. 2001;2:43–53.
    1. FAO/WHO. Health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria. Basel, Switzerland: World Health Organization; 2001.
    1. Spinler JK, Taweechotipatr M, Rognerud CL, Ou CN, Tumwasorn S, et al. Human-derived probiotic Lactobacillus reuteri demonstrate antimicrobial activities targeting diverse enteric bacterial pathogens. Anaerobe. 2008;14:166–171.
    1. Morita H, Toh H, Fukuda S, Horikawa H, Oshima K, et al. Comparative genome analysis of Lactobacillus reuteri and Lactobacillus fermentum reveal a genomic island for reuterin and cobalamin production. DNA Res. 2008;15:151–161.
    1. Santos F, Spinler JK, Saulnier DM, Molenaar D, Teusink B, et al. Functional identification in Lactobacillus reuteri of a PocR-like transcription factor regulating glycerol utilization and vitamin B12 synthesis. Microb Cell Fact. 2011;10:55.
    1. Lin YP, Thibodeaux CH, Pena JA, Ferry GD, Versalovic J. Probiotic Lactobacillus reuteri suppress proinflammatory cytokines via c-Jun. Inflamm Bowel Dis. 2008;14:1068–1083.
    1. Iyer C, Kosters A, Sethi G, Kunnumakkara AB, Aggarwal BB, et al. Probiotic Lactobacillus reuteri promotes TNF-induced apoptosis in human myeloid leukemia-derived cells by modulation of NF-kappaB and MAPK signalling. Cell Microbiol. 2008;10:1442–1452.
    1. Watanabe T, Nishio H, Tanigawa T, Yamagami H, Okazaki H, et al. Probiotic Lactobacillus casei strain Shirota prevents indomethacin-induced small intestinal injury: involvement of lactic acid. Am J Physiol Gastrointest Liver Physiol. 2009;297:G506–513.
    1. Konstantinov SR, Smidt H, de Vos WM, Bruijns SC, Singh SK, et al. S layer protein A of Lactobacillus acidophilus NCFM regulates immature dendritic cell and T cell functions. Proc Natl Acad Sci U S A. 2008;105:19474–19479.
    1. Fernandez EM, Valenti V, Rockel C, Hermann C, Pot B, et al. Anti-inflammatory capacity of selected lactobacilli in experimental colitis is driven by NOD2-mediated recognition of a specific peptidoglycan-derived muropeptide. Gut. 2011;60:1050–1059.
    1. Murzyn A, Krasowska A, Stefanowicz P, Dziadkowiec D, Lukaszewicz M. Capric acid secreted by S. boulardii inhibits C. albicans filamentous growth, adhesion and biofilm formation. PLoS One. 2010;5:e12050.
    1. Thomas CM, Versalovic J. Probiotics-host communication: Modulation of signaling pathways in the intestine. Gut Microbes. 2010;1:148–163.
    1. Preidis GA, Versalovic J. Targeting the human microbiome with antibiotics, probiotics, and prebiotics: gastroenterology enters the metagenomics era. Gastroenterology. 2009;136:2015–2031.
    1. Gionchetti P, Rizzello F, Morselli C, Poggioli G, Tambasco R, et al. High-dose probiotics for the treatment of active pouchitis. Dis Colon Rectum. 2007;50:2075–2082; discussion 2082–2074.
    1. Alfaleh K, Bassler D. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev; 2008. CD005496
    1. Pena JA, Rogers AB, Ge Z, Ng V, Li SY, et al. Probiotic Lactobacillus spp. diminish Helicobacter hepaticus-induced inflammatory bowel disease in interleukin-10-deficient mice. Infect Immun. 2005;73:912–920.
    1. Christensen HR, Frokiaer H, Pestka JJ. Lactobacilli differentially modulate expression of cytokines and maturation surface markers in murine dendritic cells. J Immunol. 2002;168:171–178.
    1. Banerjee A, Gerondakis S. Coordinating TLR-activated signaling pathways in cells of the immune system. Immunol Cell Biol. 2007;85:420–424.
    1. Carter AB, Monick MM, Hunninghake GW. Both Erk and p38 kinases are necessary for cytokine gene transcription. Am J Respir Cell Mol Biol. 1999;20:751–758.
    1. Guha M, Mackman N. LPS induction of gene expression in human monocytes. Cell Signal. 2001;13:85–94.
    1. Waltereit R, Weller M. Signaling from cAMP/PKA to MAPK and synaptic plasticity. Mol Neurobiol. 2003;27:99–106.
    1. Funaki C, Hodges RR, Dartt DA. Identification of the Raf-1 signaling pathway used by cAMP to inhibit p42/p44 MAPK in rat lacrimal gland acini: role in potentiation of protein secretion. Invest Ophthalmol Vis Sci. 2010;51:6321–6328.
    1. Pena JA, Thompson-Snipes L, Calkins PR, Tatevian N, Puppi M, et al. Alterations in myeloid dendritic cell innate immune responses in the Galphai2-deficient mouse model of colitis. Inflamm Bowel Dis. 2009;15:248–260.
    1. Guma M, Stepniak D, Shaked H, Spehlmann ME, Shenouda S, et al. Constitutive intestinal NF-{kappa}B does not trigger destructive inflammation unless accompanied by MAPK activation. J Exp Med. 2011;208:1889–1900.
    1. Xia J, Bjorndahl TC, Tang P, Wishart DS. MetaboMiner-semi-automated identification of metabolites from 2D NMR spectra of complex biofluids. BMC Bioinformatics. 2008;9:507–523.
    1. Koyama J, Taga S, Shimizu K, Shimizu M, Morita I, et al. Simultaneous determination of histamine and prostaglandin D(2) using an LC-ESI-MS/MS method with positive/negative ion-switching ionization modes: application to the study of anti-allergic flavonoids on the degranulation of KU812 cells. Anal Bioanal Chem. 2011;401:1385–1392.
    1. Trip H, Mulder NL, Rattray FP, Lolkema JS. HdcB, a novel enzyme catalysing maturation of pyruvoyl-dependent histidine decarboxylase. Mol Microbiol. 2011;79:861–871.
    1. Martin MC, Fernandez M, Linares DM, Alvarez MA. Sequencing, characterization and transcriptional analysis of the histidine decarboxylase operon of Lactobacillus buchneri. Microbiology. 2005;151:1219–1228.
    1. Lucas PM, Wolken WA, Claisse O, Lolkema JS, Lonvaud-Funel A. Histamine-producing pathway encoded on an unstable plasmid in Lactobacillus hilgardii 0006. Appl Environ Microbiol. 2005;71:1417–1424.
    1. Thomason L, Court DL, Bubunenko M, Costantino N, Wilson H, et al. Recombineering: genetic engineering in bacteria using homologous recombination. Curr Protoc Mol Biol Chapter. 2007;1:Unit 1 16.
    1. van Pijkeren JP, Britton RA. High efficiency recombineering in lactic acid bacteria. Nucleic Acids Res. 2012 in press.
    1. Sander LE, Lorentz A, Sellge G, Coeffier M, Neipp M, et al. Selective expression of histamine receptors H1R, H2R, and H4R, but not H3R, in the human intestinal tract. Gut. 2006;55:498–504.
    1. Vannier E, Miller LC, Dinarello CA. Histamine suppresses gene expression and synthesis of tumor necrosis factor alpha via histamine H2 receptors. J Exp Med. 1991;174:281–284.
    1. Wang L, Gantz I, DelValle J. Histamine H2 receptor activates adenylate cyclase and PLC via separate GTP-dependent pathways. Am J Physiol. 1996;271:G613–620.
    1. Taffet SM, Singhel KJ, Overholtzer JF, Shurtleff SA. Regulation of tumor necrosis factor expression in a macrophage-like cell line by lipopolysaccharide and cyclic AMP. Cell Immunol. 1989;120:291–300.
    1. Dohlsten M, Kalland T, Sjogren HO, Carlsson R. Histamine inhibits interleukin 1 production by lipopolysaccharide-stimulated human peripheral blood monocytes. Scand J Immunol. 1988;27:527–532.
    1. Elenkov IJ, Webster E, Papanicolaou DA, Fleisher TA, Chrousos GP, et al. Histamine potently suppresses human IL-12 and stimulates IL-10 production via H2 receptors. J Immunol. 1998;161:2586–2593.
    1. Takahashi HK, Morichika T, Iwagaki H, Tamura R, Kubo S, et al. Histamine downregulates CD14 expression via H2 receptors on human monocytes. Clin Immunol. 2003;108:274–281.
    1. Nakamura T, Ueno Y, Goda Y, Nakamura A, Shinjo K, et al. Efficacy of a selective histamine H2 receptor agonist, dimaprit, in experimental models of endotoxin shock and hepatitis in mice. Eur J Pharmacol. 1997;322:83–89.
    1. Masaki T, Chiba S, Tatsukawa H, Noguchi H, Kakuma T, et al. The role of histamine H1 receptor and H2 receptor in LPS-induced liver injury. FASEB J. 2005;19:1245–1252.
    1. Handley SA, Dube PH, Miller VL. Histamine signaling through the H(2) receptor in the Peyer's patch is important for controlling Yersinia enterocolitica infection. Proc Natl Acad Sci U S A. 2006;103:9268–9273.
    1. Aschenbach JR, Honscha KU, von Vietinghoff V, Gabel G. Bioelimination of histamine in epithelia of the porcine proximal colon of pigs. Inflamm Res. 2009;58:269–276.
    1. Jutel M, Watanabe T, Akdis M, Blaser K, Akdis CA. Immune regulation by histamine. Curr Opin Immunol. 2002;14:735–740.
    1. Jutel M, Watanabe T, Klunker S, Akdis M, Thomet OA, et al. Histamine regulates T-cell and antibody responses by differential expression of H1 and H2 receptors. Nature. 2001;413:420–425.
    1. Guillet R, Stoll BJ, Cotten CM, Gantz M, McDonald S, et al. Association of H2-blocker therapy and higher incidence of necrotizing enterocolitis in very low birth weight infants. Pediatrics. 2006;117:e137–142.
    1. Wu G. Amino acids: metabolism, functions, and nutrition. Amino Acids. 2009;37:1–17.
    1. Coeffier M, Marion-Letellier R, Dechelotte P. Potential for amino acids supplementation during inflammatory bowel diseases. Inflamm Bowel Dis. 2010;16:518–524.
    1. Son DO, Satsu H, Shimizu M. Histidine inhibits oxidative stress- and TNF-alpha-induced interleukin-8 secretion in intestinal epithelial cells. FEBS Lett. 2005;579:4671–4677.
    1. Andou A, Hisamatsu T, Okamoto S, Chinen H, Kamada N, et al. Dietary histidine ameliorates murine colitis by inhibition of proinflammatory cytokine production from macrophages. Gastroenterology. 2009;136:564–574 e562.
    1. Fuller MF, Reeds PJ. Nitrogen cycling in the gut. Annu Rev Nutr. 1998;18:385–411.
    1. Yang XD, Ai W, Asfaha S, Bhagat G, Friedman RA, et al. Histamine deficiency promotes inflammation-associated carcinogenesis through reduced myeloid maturation and accumulation of CD11b+Ly6G+immature myeloid cells. Nat Med. 2011;17:87–95.
    1. Nelson KE, Weinstock GM, Highlander SK, Worley KC, Creasy HH, et al. A catalog of reference genomes from the human microbiome. Science. 2010;328:994–999.
    1. Frese SA, Benson AK, Tannock GW, Loach DM, Kim J, et al. The evolution of host specialization in the vertebrate gut symbiont Lactobacillus reuteri. PLoS Genet. 2011;7:e1001314.
    1. Drouault S, Anba J, Corthier G. Streptococcus thermophilus is able to produce a beta-galactosidase active during its transit in the digestive tract of germ-free mice. Appl Environ Microbiol. 2002;68:938–941.
    1. Walter J, Heng NC, Hammes WP, Loach DM, Tannock GW, et al. Identification of Lactobacillus reuteri genes specifically induced in the mouse gastrointestinal tract. Appl Environ Microbiol. 2003;69:2044–2051.
    1. Bron PA, Grangette C, Mercenier A, de Vos WM, Kleerebezem M. Identification of Lactobacillus plantarum genes that are induced in the gastrointestinal tract of mice. J Bacteriol. 2004;186:5721–5729.
    1. Klaassens ES, Boesten RJ, Haarman M, Knol J, Schuren FH, et al. Mixed-species genomic microarray analysis of fecal samples reveals differential transcriptional responses of bifidobacteria in breast- and formula-fed infants. Appl Environ Microbiol. 2009;75:2668–2676.
    1. Corthier G, Delorme C, Ehrlich SD, Renault P. Use of luciferase genes as biosensors to study bacterial physiology in the digestive tract. Appl Environ Microbiol. 1998;64:2721–2722.
    1. Alifano P, Fani R, Lio P, Lazcano A, Bazzicalupo M, et al. Histidine biosynthetic pathway and genes: structure, regulation, and evolution. Microbiol Rev. 1996;60:44–69.
    1. Hungerford JM. Scombroid poisoning: a review. Toxicon. 2010;56:231–243.
    1. Nuutinen S, Panula P. Histamine in Neurotransmission and Brain Diseases. Adv Exp Med Biol. 2011;709:95–107.
    1. Jones SE, Versalovic J. Probiotic Lactobacillus reuteri biofilms produce antimicrobial and anti-inflammatory factors. BMC Microbiol. 2009;9:35.
    1. Weljie AM, Newton J, Mercier P, Carlson E, Slupsky CM. Targeted profiling: quantitative analysis of 1H NMR metabolomics data. Anal Chem. 2006;78:4430–4442.
    1. Braunschweiler L, Ernst RR. Coherence transfer by isotropic mixing: application to proton correlation spectroscopy. J Magn Reson. 1983;53:521–528.
    1. Sklenar V, Piotto M, Leppik R, Saudek V. Gradient-tailored water suppression for 1H-15N HSQC experiments optimized to retain full sensitivity. J Magn Reson, A. 1993;102:241–245.
    1. Shaka AJ, Lee CJ, Pine A. Iterative schemes for bilinear operators; applications to spin decoupling. J Magn Reson. 1988;77:274–293.
    1. Bodenhausen G, Ruben DJ. Natural abundance nitrogen-15 NMR by enhanced heteronuclear spectroscopy. Chem Phys Lett. 1980;69:185–189.
    1. Kay LE, Keifer P, Saarinen T. Pure absorption gradient enhanced heteronuclear single quantum correlation spectroscopy with improved sensitivity. J Am Chem Soc. 1992;114:10663–10665.
    1. Delaglio F, Grzesiek S, Vuister GW, Zhu G, Pfeifer J, et al. NMRPipe: a multidimensional spectral processing system based on UNIX pipes. J Biomol NMR. 1995;6:277–293.
    1. Johnson BA, Blevins RA. NMRView: A computer program for the visualization and analysis of NMR data. J Biomol NMR. 1994;4:603–614.
    1. Zhu G, Bax A. Improved linear prediction for truncated signals of known phase. J Magn Reson. 1990;90:405–410.
    1. Gosetti F, Mazzucco E, Gianotti V, Polati S, Gennaro MC. High performance liquid chromatography/tandem mass spectrometry determination of biogenic amines in typical Piedmont cheeses. J Chromatogr A. 2007;1149:151–157.
    1. Saulnier DM, Santos F, Roos S, Mistretta TA, Spinler JK, et al. Exploring metabolic pathway reconstruction and genome-wide expression profiling in Lactobacillus reuteri to define functional probiotic features. PLoS One. 2011;6:e18783.
    1. Wall T, Bath K, Britton RA, Jonsson H, Versalovic J, et al. The early response to acid shock in Lactobacillus reuteri involves the ClpL chaperone and a putative cell wall-altering esterase. Appl Environ Microbiol. 2007;73:3924–3935.

Source: PubMed

3
Prenumerera