Play all audios:
ABSTRACT The gut microbiome has major roles in modulating host physiology. One such function is colonization resistance, or the ability of the microbial collective to protect the host
against enteric pathogens1,2,3, including enterohaemorrhagic _Escherichia coli_ (EHEC) serotype O157:H7, an attaching and effacing (AE) food-borne pathogen that causes severe
gastroenteritis, enterocolitis, bloody diarrhea and acute renal failure4,5 (haemolytic uremic syndrome). Although gut microorganisms can provide colonization resistance by outcompeting some
pathogens or modulating host defence provided by the gut barrier and intestinal immune cells6,7, this phenomenon remains poorly understood. Here, we show that activation of the
neurotransmitter receptor dopamine receptor D2 (DRD2) in the intestinal epithelium by gut microbial metabolites produced upon dietary supplementation with the essential amino acid
l-tryptophan protects the host against _Citrobacter rodentium_, a mouse AE pathogen that is widely used as a model for EHEC infection8,9. We further find that DRD2 activation by these
tryptophan-derived metabolites decreases expression of a host actin regulatory protein involved in _C. rodentium_ and EHEC attachment to the gut epithelium via formation of actin pedestals.
Our results reveal a noncanonical colonization resistance pathway against AE pathogens that features an unconventional role for DRD2 outside the nervous system in controlling actin
cytoskeletal organization in the gut epithelium. Our findings may inspire prophylactic and therapeutic approaches targeting DRD2 with dietary or pharmacological interventions to improve gut
health and treat gastrointestinal infections, which afflict millions globally. Access through your institution Buy or subscribe This is a preview of subscription content, access via your
institution ACCESS OPTIONS Access through your institution Access Nature and 54 other Nature Portfolio journals Get Nature+, our best-value online-access subscription $32.99 / 30 days cancel
any time Learn more Subscribe to this journal Receive 51 print issues and online access $199.00 per year only $3.90 per issue Learn more Buy this article * Purchase on SpringerLink *
Instant access to full article PDF Buy now Prices may be subject to local taxes which are calculated during checkout ADDITIONAL ACCESS OPTIONS: * Log in * Learn about institutional
subscriptions * Read our FAQs * Contact customer support SIMILAR CONTENT BEING VIEWED BY OTHERS VITAMIN B12 PRODUCED BY GUT BACTERIA MODULATES CHOLINERGIC SIGNALLING Article 02 January 2024
A COMMENSAL PROTOZOAN ATTENUATES _CLOSTRIDIOIDES DIFFICILE_ PATHOGENESIS IN MICE VIA ARGININE-ORNITHINE METABOLISM AND HOST INTESTINAL IMMUNE RESPONSE Article Open access 02 April 2024
_DUBOSIELLA NEWYORKENSIS_ MODULATES IMMUNE TOLERANCE IN COLITIS VIA THE L-LYSINE-ACTIVATED AHR-IDO1-KYN PATHWAY Article Open access 13 February 2024 DATA AVAILABILITY Next-generation
sequencing reads have been deposited at NCBI BioProject under accession number PRJNA1049399. Source data are provided with this paper. CODE AVAILABILITY R script for statistical analysis,
box pots, and QIIME2 code are available at https://zenodo.org/records/10535214. REFERENCES * Buffie, C. G. & Pamer, E. G. Microbiota-mediated colonization resistance against intestinal
pathogens. _Nat. Rev. Immunol._ 13, 790–801 (2013). CAS PubMed PubMed Central Google Scholar * McKenney, P. T. & Pamer, E. G. From hype to hope: the gut microbiota in enteric
infectious disease. _Cell_ 163, 1326–1332 (2015). CAS PubMed PubMed Central Google Scholar * Caballero, S. & Pamer, E. G. Microbiota-mediated inflammation and antimicrobial defense
in the intestine. _Annu. Rev. Immunol._ 33, 227–256 (2015). CAS PubMed PubMed Central Google Scholar * Croxen, M. A. & Finlay, B. B. Molecular mechanisms of _Escherichia coli_
pathogenicity. _Nat. Rev. Microbiol._ 8, 26–38 (2010). CAS PubMed Google Scholar * Kaper, J. B., Nataro, J. P. & Mobley, H. L. T. Pathogenic _Escherichia coli_. _Nat. Rev. Microbiol._
2, 123–140 (2004). CAS PubMed Google Scholar * Kamada, N. et al. Regulated virulence controls the ability of a pathogen to compete with the gut microbiota. _Science_ 336, 1325–1329
(2012). ADS CAS PubMed PubMed Central Google Scholar * Willing, B. P., Vacharaksa, A., Croxen, M., Thanachayanont, T. & Finlay, B. B. Altering host resistance to infections through
microbial transplantation. _PLoS ONE_ 6, 2–10 (2011). Google Scholar * Collins, J. W. et al. _Citrobacter rodentium_: infection, inflammation and the microbiota. _Nat. Rev. Microbiol._ 12,
612–623 (2014). CAS PubMed Google Scholar * Mullineaux-Sanders, C. et al. _Citrobacter rodentium_–host–microbiota interactions: immunity, bioenergetics and metabolism. _Nat. Rev.
Microbiol._ 17, 701–715 (2019). CAS PubMed Google Scholar * Garmendia, J., Frankel, G. & Crepin, V. F. Enteropathogenic and enterohemorrhagic _Escherichia coli_ infections:
translocation, translocation, translocation. _Infect. Immun._ 73, 2573–2585 (2005). CAS PubMed PubMed Central Google Scholar * Battle, S. E., Brady, M. J., Vanaja, S. K., Leong, J. M.
& Hecht, G. A. Actin pedestal formation by enterohemorrhagic _Escherichia coli_ enhances bacterial host cell attachment and concomitant type III translocation. _Infect. Immun._ 82,
3713–3722 (2014). PubMed PubMed Central Google Scholar * Mallick, E. M. et al. The ability of an attaching and effacing pathogen to trigger localized actin assembly contributes to
virulence by promoting mucosal attachment. _Cell Microbiol._ 16, 1405–1424 (2014). CAS PubMed PubMed Central Google Scholar * Law, R. J., Gur-Arie, L., Rosenshine, I. & Finlay, B. B.
In vitro and in vivo model systems for studying enteropathogenic _Escherichia coli_ infections. _Cold Spring Harb. Perspect. Med._ 3, a009977–a009977 (2013). PubMed PubMed Central Google
Scholar * Dorrestein, P. C., Mazmanian, S. K. & Knight, R. Finding the missing links among metabolites, microbes, and the host. _Immunity_ 40, 824–832 (2014). CAS PubMed PubMed
Central Google Scholar * Donia, M. S. & Fischbach, M. A. Small molecules from the human microbiota. _Science_ 349, 1254766 (2015). PubMed PubMed Central Google Scholar * Furusawa,
Y. et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. _Nature_ 504, 446–450 (2013). ADS CAS PubMed Google Scholar * Smith, P. M. et al.
The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. _Science_ 341, 569–573 (2013). ADS CAS PubMed Google Scholar * Arpaia, N. et al. Metabolites
produced by commensal bacteria promote peripheral regulatory T-cell generation. _Nature_ 504, 451–455 (2013). ADS CAS PubMed PubMed Central Google Scholar * Zhao, L. et al. Gut bacteria
selectively promoted by dietary fibers alleviate type 2 diabetes. _Science_ 359, 1151–1156 (2018). ADS CAS PubMed Google Scholar * Hang, S. et al. Bile acid metabolites control TH17 and
Treg cell differentiation. _Nature_ 576, 143–148 (2019). ADS CAS PubMed PubMed Central Google Scholar * Bender, M. J. et al. Dietary tryptophan metabolite released by intratumoral
_Lactobacillus reuteri_ facilitates immune checkpoint inhibitor treatment. _Cell_ 186, 1846–1862 (2023). CAS PubMed Google Scholar * Rangan, K. J. & Hang, H. C. Biochemical mechanisms
of pathogen restriction by intestinal bacteria. _Trends Biochem. Sci._ 42, 887–898 (2017). CAS PubMed PubMed Central Google Scholar * Chang, P. V. Chemical mechanisms of colonization
resistance by the gut microbial metabolome. _ACS Chem. Biol._ 15, 1119–1126 (2020). ADS CAS PubMed PubMed Central Google Scholar * Rooks, M. G. & Garrett, W. S. Gut microbiota,
metabolites and host immunity. _Nat. Rev. Immunol._ 16, 341–352 (2016). CAS PubMed PubMed Central Google Scholar * Postler, T. S. & Ghosh, S. Understanding the holobiont: how
microbial metabolites affect human health and shape the immune system. _Cell Metab._ 26, 110–130 (2017). CAS PubMed PubMed Central Google Scholar * Belkaid, Y. & Hand, T. W. Role of
the microbiota in immunity and inflammation. _Cell_ 157, 121–141 (2014). CAS PubMed PubMed Central Google Scholar * Brestoff, J. R. & Artis, D. Commensal bacteria at the interface of
host metabolism and the immune system. _Nat. Immunol._ 14, 676–684 (2013). CAS PubMed PubMed Central Google Scholar * Scott, S. A., Fu, J. & Chang, P. V. Microbial tryptophan
metabolites regulate gut barrier function via the aryl hydrocarbon receptor. _Proc. Natl Acad. Sci. USA_ 117, 19376–19387 (2020). ADS CAS PubMed PubMed Central Google Scholar * Kumar,
A. & Sperandio, V. Indole signaling at the host–microbiota–pathogen interface. _MBio_ 10, e01031–19 (2019). CAS PubMed PubMed Central Google Scholar * Bansal, T., Alaniz, R. C.,
Wood, T. K. & Jayaraman, A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. _Proc. Natl Acad. Sci. USA_ 107,
228–233 (2010). ADS CAS PubMed Google Scholar * Bommarius, B. et al. A family of indoles regulate virulence and Shiga toxin production in pathogenic _E. coli_. _PLoS ONE_ 8, e54456–16
(2013). ADS CAS PubMed PubMed Central Google Scholar * Keiser, M. J. et al. Relating protein pharmacology by ligand chemistry. _Nat. Biotechnol._ 25, 197–206 (2007). CAS PubMed Google
Scholar * Missale, C., Russel Nash, S., Robinson, S. W., Jaber, M. & Caron, M. G. Dopamine receptors: from structure to function. _Physiol. Rev._ 78, 189–225 (1998). CAS PubMed
Google Scholar * Beaulieu, J. & Gainetdinov, R. R. The physiology, signaling, and pharmacology of dopamine receptors. _Pharmacol. Rev._ 63, 182–217 (2011). CAS PubMed Google Scholar
* Kulagowski, J. J. et al. 3-[[4-(4-Chlorophenyl)piperazin-1-yl]-methyl]-1H-pyrrolo[2,3-b]pyridine: an antagonist with high affinity and selectivity for the human dopamine D4 receptor. _J.
Med. Chem._ 39, 1941–1942 (1996). CAS PubMed Google Scholar * Binkowski, B. F. et al. A luminescent biosensor with increased dynamic range for intracellular cAMP. _ACS Chem. Biol._ 6,
1193–1197 (2011). CAS PubMed Google Scholar * Barnea, G. et al. The genetic design of signaling cascades to record receptor activation. _Proc. Natl Acad. Sci. USA_ 105, 64–69 (2008). ADS
CAS PubMed Google Scholar * Murata, K., Noda, K., Kohno, K. & Samejima, M. Bioavailability and pharmacokinetics of oral dopamine in dogs. _J. Pharm. Sci._ 77, 565–568 (1988). CAS
PubMed Google Scholar * Alex, K. D. & Pehek, E. A. Pharmacologic mechanisms of serotonergic regulation of dopamine neurotransmission. _Pharmacol. Ther._ 113, 296–320 (2007). CAS
PubMed Google Scholar * Łukasiewicz, S., Błasiak, E., Szafran-Pilch, K. & Dziedzicka-Wasylewska, M. Dopamine D2 and serotonin 5-HT1A receptor interaction in the context of the effects
of antipsychotics—in vitro studies. _J. Neurochem._ 137, 549–560 (2016). PubMed Google Scholar * Kumar, A. et al. The serotonin neurotransmitter modulates virulence of enteric pathogens.
_Cell Host Microbe_ 28, 41–53 (2020). CAS PubMed PubMed Central Google Scholar * Suetsugu, S. et al. Sustained activation of N-WASP through phosphorylation is essential for neurite
extension. _Dev. Cell_ 3, 645–658 (2002). CAS PubMed Google Scholar * Sasahara, Y. et al. Mechanism of recruitment of WASP to the immunological synapse and of its activation following TCR
ligation. _Mol. Cell_ 10, 1269–1281 (2002). CAS PubMed Google Scholar * Martinez-Quiles, N. et al. WIP regulates N-WASP-mediated actin polymerization and filopodium formation. _Nat. Cell
Biol._ 3, 484–491 (2001). CAS PubMed Google Scholar * Desai, M. S. et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility.
_Cell_ 167, 1339–1353 (2016). CAS PubMed PubMed Central Google Scholar * Schulthess, J. et al. The short chain fatty acid butyrate imprints an antimicrobial program in macrophages.
_Immunity_ 50, 432–445 (2019). CAS PubMed PubMed Central Google Scholar * Chen, H. et al. A forward chemical genetic screen reveals gut microbiota metabolites that modulate host
physiology. _Cell_ 177, 1217–1231.e18 (2019). CAS PubMed PubMed Central Google Scholar * Colosimo, D. A. et al. Mapping interactions of microbial metabolites with human G-protein-coupled
receptors. _Cell Host Microbe_ 26, 273–282 (2019). CAS PubMed PubMed Central Google Scholar * Zhao, X. et al. Chemoproteomics reveals microbiota-derived aromatic monoamine agonists for
GPRC5A. _Nat. Chem. Biol._ 19, 1205–1214 (2023). CAS PubMed Google Scholar * Thompson, L. R. et al. A communal catalogue reveals Earth’s multiscale microbial diversity. _Nature_ 551,
457–463 (2017). ADS CAS PubMed PubMed Central Google Scholar * Bolyen, E. et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. _Nat.
Biotechnol._ 37, 852–857 (2019). CAS PubMed PubMed Central Google Scholar * Callahan, B. J. et al. DADA2: High-resolution sample inference from Illumina amplicon data. _Nat. Methods_ 13,
581–583 (2016). CAS PubMed PubMed Central Google Scholar * DeSantis, T. Z. et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. _Appl. Environ.
Microbiol._ 72, 5069–5072 (2006). ADS CAS PubMed PubMed Central Google Scholar Download references ACKNOWLEDGEMENTS The authors thank the Arnold and Mabel Beckman Foundation (Beckman
Young Investigator Award to P.V.C.) and a President’s Council for Cornell Women Affinito-Stewart Grant (P.V.C.) for support. This work was supported in part by a grant from the National
Institutes of Health (NIH R35GM133501). J.F. was supported by a Cornell Institute of Host-Microbe Interactions and Disease (CIHMID) Postdoctoral Fellowship. Imaging data was acquired through
the Cornell Institute of Biotechnology BRC Imaging Facility (RRID:SCR_021741), with NYSTEM (C029155) and NIH (S10OD018516) funding for the shared Zeiss LSM 880 confocal/multiphoton
microscope. We thank the Weill Institute for Cell and Molecular Biology for additional resources and reagents. AUTHOR INFORMATION AUTHORS AND AFFILIATIONS * Department of Microbiology,
Cornell University, Ithaca, NY, USA Samantha A. Scott * Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA Samantha A. Scott, Jingjing Fu & Pamela V. Chang *
Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA Pamela V. Chang * Cornell Center for Immunology, Cornell University, Ithaca, NY, USA Pamela V. Chang *
Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, USA Pamela V. Chang Authors * Samantha A. Scott View author publications You can also search for
this author inPubMed Google Scholar * Jingjing Fu View author publications You can also search for this author inPubMed Google Scholar * Pamela V. Chang View author publications You can also
search for this author inPubMed Google Scholar CONTRIBUTIONS S.A.S. and P.V.C. conceptualized the study. S.A.S., J.F. and P.V.C. designed the experiments. J.F. performed the targeted
metabolomic studies. S.A.S. carried out all other studies and bioinformatic analyses. S.A.S., J.F. and P.V.C. wrote the manuscript. CORRESPONDING AUTHOR Correspondence to Pamela V. Chang.
ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing interests. PEER REVIEW PEER REVIEW INFORMATION _Nature_ thanks Vanessa Sperandio and the other, anonymous,
reviewer(s) for their contribution to the peer review of this work. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published
maps and institutional affiliations. EXTENDED DATA FIGURES AND TABLES EXTENDED DATA FIG. 1 THE TRP METABOLITES I3A, IPYA, AND IET PROTECT AGAINST _CITROBACTER RODENTIUM_ INFECTION IN MICE.
C57Bl/6 mice were pre-treated with antibiotics (ABX) for 7 d, followed by Trp metabolites, I3A (1000 mg/kg), IPyA (2900 mg/kg), or IEt (600 mg/kg) by oral gavage daily for 2 d. The mice were
then administered _C. rodentium_ (CR, oral gavage, 108 CFU) with continued ABX (except neomycin) and metabolite treatment for 10 d. Colon sections were stained with H&E. Shown are
representative images. Scale bar: 50 μm. Data are representative of at least 3 independent experiments, n = 10 mice per group. EXTENDED DATA FIG. 2 TRYPTOPHAN METABOLITES DO NOT AFFECT _C.
RODENTIUM_ AND EHEC GROWTH AND VIRULENCE IN VITRO. _C. rodentium_ (CR) (A–E) and EHEC O157:H7 (F–K) were cultured in the presence of I3A, IPyA, or IEt (100 μM). A, F, Growth was monitored by
measuring OD600 absorbance readings over 24 h. B, G, Cultures were plated after 24 h, and CFUs were counted. (C–E, H–K) Bacteria were cultured in low glucose DMEM under anaerobic (ana),
microaerophilic (micro), and aerobic (aero) conditions to activate locus of enterocyte effacement-pathogenicity island expression with I3A, IPyA, or IEt (100 μM). RNA was isolated after
cultures reached late-log phase (OD600 = 0.6–0.8), and cDNA was synthesized and analyzed by qPCR for the indicated genes. Relative expression of mRNA transcripts was normalized to the RNA
polymerase subunit alpha _rpoA_. Data are represented as the fold induction over control samples, bars = mean, error bars = standard deviation. Statistical analysis was performed using a
two-tailed Student’s t-test (A–B, F–G), or one-way ANOVA, followed by post-hoc Tukey multiple comparison test (C–E, H–K), n = 3 biological replicates examined over 3 independent experiments.
Source Data EXTENDED DATA FIG. 3 EFFECTS OF I3A, IPYA, AND IET DEPEND ON DOPAMINE RECEPTOR D2 (DRD2). A–B, Polarized Caco-2 monolayers were pre-treated with haloperidol (HAL, 10 μM) for 24
h, followed by metabolites (I3A, IPyA, or IEt, 100 μM) for 2 d, and then infection with EHEC O157:H7 for 16 h. B, Representative images of pedestals (denoted by arrows) from Caco-2 cells
stained with DAPI and Alexa Fluor 647-phalloidin and imaged by confocal microscopy. Shown are maximum intensity z-projections. Scale bar: 5 μm. C, Western blot analysis of Caco-2 monolayers
to verify CRISPR/Cas9-mediated knockout (KO) of _Drd2_, _Drd3_, and _Drd4_. GAPDH is shown as a loading control. D–F, Caco-2 monolayers (WT vs. KO) were pre-treated with metabolites (I3A,
IPyA, or IEt, 100 μM) for 2 d and then infected with EHEC O157:H7 for 16 h. (A, D–F) Pedestal formation = # of pedestals per Caco-2 cell (HAL: I3A, n = 955; IPyA, n = 970; IEt, n = 1024;
_Drd3_ KO: I3A, n = 1089; IPyA, n = 1094; IEt, n = 1012; _Drd4_ KO: I3A, n = 1032; IPyA, n = 887; IEt, n = 1026; _Drd2_ KO: I3A, n = 1032; IPyA, n = 1026; IEt, n = 1073 cells examined over 3
independent experiments). For box plots, interquartile ranges (IQRs, boxes), median values (line within box), whiskers (lowest and highest values within 1.5 times IQR from the first and
third quartiles), and outliers beyond whiskers (dots), are shown. G-M, Caco-2 cells (WT vs KO) were pre-treated with metabolites (I3A, IPyA, or IEt, 100 μM) for 24 h and then infected with
EHEC O157:H7 for 12 h. Cell lysates were analyzed by Western blotting with the indicated antibodies. Source data are provided in Supplementary Fig. 9. (H, J, L, M) Densitometry was performed
using FIJI, bars = mean, error bars = standard deviation. Data are representative of at least 3 independent experiments, n = 3 biological replicates. Statistical analysis was performed
using one-way ANOVA, followed by post-hoc Tukey multiple comparison test. Source Data EXTENDED DATA FIG. 4 I3A, IPYA, AND IET ARE LIGANDS OF DOPAMINE RECEPTOR D2 (DRD2), WHICH SIGNALS VIA
GΑI, AND _DRD2_ IS KNOCKED OUT IN INTESTINAL EPITHELIAL CELLS IN _DRD2_FL/FL X VILLIN-CRE MICE FED TRP DIET DURING _C. RODENTIUM_ INFECTION. A, C–F, HEK 293 T cells overexpressing either
DRD2 and a split luciferase-based cAMP sensor (GloSensor) or B, DRD2-Tango and a β-arrestin-TEV fusion were incubated with dopamine (DA), I3A, IPyA, or IEt (1 mM each in A–B; concentrations
indicated in C–F) for 15 min (A, C–F) or 24 h (B), after which luminescence was measured to quantify ligand-induced (A) decrease in cAMP or (B) increase in β-arrestin recruitment. RLU =
relative luminescence units. EC50 and Kd values were calculated using GraphPad Prism. (G–H) _Drd2_fl/fl x Villin (Vil)-Cre or _Drd2_fl/fl mice were fed a conventional (2 g Trp/kg diet, ad
libitum) or Trp (42 g Trp/kg diet, ad libitum) diet for 7 d and then infected with _C. rodentium_ (CR, oral gavage, 108 CFU) with continued Trp feeding. Ten days post-infection, intestinal
cryosections were stained with DAPI and an anti-DRD2 antibody, followed by an anti-mouse Alexa Fluor 594 antibody. (G) Shown are representative z-slices. Scale bar: 20 μm. (H) Image
brightness was quantified using FIJI. Statistical analysis was performed using one-way ANOVA, followed by post-hoc Tukey multiple comparison test; bars = mean, error bars = standard
deviation, (A–F) n = 3 biological replicates and (G–H) n = 10 mice per group examined over 3 independent experiments. Source Data EXTENDED DATA FIG. 5 _DRD2_ IS KNOCKED OUT IN INTESTINAL
EPITHELIAL CELLS IN _DRD2_FL/FL X VILLIN-CRE MICE ADMINISTERED TRP METABOLITES DURING _C. RODENTIUM_ INFECTION. _Drd2_fl/fl x Villin (Vil)-Cre or _Drd2_fl/fl mice were treated with Trp
metabolites, I3A (1000 mg/kg), IPyA (2900 mg/kg), or IEt (600 mg/kg), by oral gavage daily for 2 d, and then infected with _C. rodentium_ (CR, oral gavage, 108 CFU) with continued metabolite
treatment. Ten days post-infection, intestinal cryosections were stained with DAPI and an anti-DRD2 antibody, followed by an anti-mouse Alexa Fluor 594 antibody. (A) Shown are
representative z-slices. Scale bar: 20 μm. (B–D) Image brightness was quantified using FIJI. Data are representative of at least 3 independent experiments, n = 10 mice per group, bars =
mean, error bars = standard deviation. Statistical analysis was performed using one-way ANOVA, followed by post-hoc Tukey multiple comparison test. Source Data EXTENDED DATA FIG. 6 EFFECTS
OF THE TRYPTOPHAN (TRP) DIET AND METABOLITES I3A, IPYA, AND IET IN PROTECTING AGAINST _C. RODENTIUM_ INFECTION DEPEND ON DOPAMINE RECEPTOR D2 (DRD2) IN INTESTINAL EPITHELIAL CELLS (IECS).
_Drd2_fl/fl x Villin (Vil)-Cre or _Drd2_fl/fl mice were fed a conventional (2 g Trp/kg diet, ad libitum) or Trp (42 g Trp/kg diet, ad libitum) diet for 7 d or Trp metabolites, I3A (1000
mg/kg), IPyA (2900 mg/kg), or IEt (600 mg/kg), by oral gavage daily for 2 d, and then infected with _C. rodentium_ (CR, oral gavage, 108 CFU) with continued Trp feeding or metabolite
treatment. A–B, Bacterial load in (A) feces and (B) colon tissue was measured (A) every 1–2 d for 24 d post-infection and (B) at the peak of infection, 10 d post-infection. C–E, Colon
sections were stained with H&E and (C) blindly scored for submucosal edema (0-3), goblet cell depletion (0-3), epithelial hyperplasia (0-3), epithelial integrity (0-4), and neutrophil
and mononuclear cell infiltration (0-3). Data are expressed as the sum of these individual scores (0-16). See Methods for full description of scoring rubric. (D) Crypt heights were measured.
(E) Representative images. Scale bar: 50 μm. (F) Representative images of pedestals from Fig. 4a (denoted by arrows) stained with DAPI and Alexa Fluor 647-phalloidin and imaged by confocal
microscopy. Shown are maximum intensity z-projections. Scale bar: 5 μm. (G) Pedestal formation = # of pedestals per host cell (I3A, n = 1079; IPyA, n = 1027 cells examined over 3 independent
experiments). For box plots, interquartile ranges (IQRs, boxes), median values (line within box), whiskers (lowest and highest values within 1.5 times IQR from the first and third
quartiles), and outliers beyond whiskers (dots), are shown. H, I, Intestinal epithelial cells were isolated, and cell lysates were analyzed by Western blotting with the indicated antibodies.
Source data are provided in Supplementary Fig. 9. (I) Densitometry was performed using FIJI, n = 3 biological replicates. Data are representative of at least 3 independent experiments, n =
10 mice per group, bars = mean, error bars = standard deviation. Statistical analysis was performed using the two-tailed Student’s t-test (A) or one-way ANOVA, followed by post-hoc Tukey
multiple comparison test: *p < 0.05, **p < 0.01, ***p < 0.001. Source Data EXTENDED DATA FIG. 7 SYNTHETIC DRD2 AGONIST PROTECTS AGAINST A MOUSE MODEL OF EHEC INFECTION USING _C.
RODENTIUM_, STRAIN DBS100, WHEREAS A DRD2 ANTAGONIST BLOCKS THE EFFECTS. A–I, _Drd2_fl/fl x Villin (Vil)-Cre or _Drd2_fl/fl mice were pre-treated with DRD2 agonist sumanirole (SUM, 4 mg/kg,
IP injection) daily for 2 d. J–R, _Drd2_fl/fl x Vil-Cre or _Drd2_fl/fl mice were pre-treated with DRD2 antagonist L-741,626 (1 mg/kg, IP injection) daily for 2 d, followed by conventional (2
g Trp/kg diet, ad libitum) or Trp (42 g Trp/kg diet, ad libitum) diet for 7 d. A–R, The mice were then administered _C. rodentium_ (CR, oral gavage, 108 colony-forming units, CFU) with
continued (A–I) SUM treatment or (J–R) L-741,626 and Trp feeding. A, J Timeline for (A) SUM and (J) L-741,626 study. B–C, K–L, Bacterial load in (B, K) feces and (C, L) colon tissue was
measured (B, K) every 1–2 d for 10 d post-infection and (C, L) at the peak of infection, 10 d post-infection. (D–F, M–O) Colon sections were stained with H&E and (D, M) blindly scored
for submucosal edema (0-3), goblet cell depletion (0-3), epithelial hyperplasia (0-3), epithelial integrity (0-4), and neutrophil and mononuclear cell infiltration (0-3). Data are expressed
as the sum of these individual scores (0-16). See Methods for full description of scoring rubric. (E, N) Crypt heights were measured. (F, O) Representative images. Scale bar: 50 μm. (G, P)
Intestinal cryosections were stained with DAPI and Alexa Fluor 647-phalloidin. Pedestal formation = # of pedestals per host cell (SUM, n = 883; L-741,626, n = 1775 cells examined over 3
independent experiments). For box plots, interquartile ranges (IQRs, boxes), median values (line within box), whiskers (lowest and highest values within 1.5 times IQR from the first and
third quartiles), and outliers beyond whiskers (dots), are shown. H–I, Q–R, Intestinal epithelial cells were isolated, and cell lysates were analyzed by Western blotting with the indicated
antibodies. Source data are provided in Supplementary Fig. 9. (Q) Samples derive from the same experiment, and Western blots were processed in parallel. (I, R) Densitometry was performed
using FIJI, n = 3 biological replicates. Data are representative of at least 3 independent experiments, n = 10 mice per group, bars = mean, error bars = standard deviation. Statistical
analysis was performed using the two-tailed Student’s t-test (B, K) or one-way ANOVA, followed by post-hoc Tukey multiple comparison test: ***p < 0.001. Source Data EXTENDED DATA FIG. 8
EFFECTS OF TRP METABOLITES DO NOT DEPEND ON GΑI AND Β-ARRESTIN SIGNALING. Caco-2 cells were pre-treated with (A–D) Gαi inhibitor pertussis toxin (PTx, 100 ng/mL) for 18 h or (E–H) β-arrestin
inhibitor barbadin (100 μM) for 30 min, followed by Trp metabolite (I3A, IPyA, or IEt, 100 μM) for 24 h. The cells were then infected with EHEC (MOI 50) for 12 h, (A, E) fixed, and stained
with DAPI and Alexa Fluor 647-phalloidin. Pedestal formation = # of pedestals per Caco-2 cell (PTx: I3A, n = 994; IPyA, n = 916; IEt, n = 905; barbadin: I3A, n = 934; IPyA, n = 993; IEt, n =
1087 cells examined over 3 independent experiments). For box plots, interquartile ranges (IQRs, boxes), median values (line within box), whiskers (lowest and highest values within 1.5 times
IQR from the first and third quartiles), and outliers beyond whiskers (dots), are shown. B–D, F–H, Alternatively, cells were lysed and analyzed by Western blotting with the indicated
antibodies. Source data are provided in Supplementary Fig. 9. (C–D, G–H) Densitometry was performed using FIJI. Data are representative of at least 3 independent experiments, n = 3
biological replicates, bars = mean, error bars = standard deviation. Statistical analysis was performed using one-way ANOVA, followed by post-hoc Tukey multiple comparison test. Source Data
EXTENDED DATA FIG. 9 EFFECTS OF TRP METABOLITES DEPEND ON GΒΓ AND PHOSPHOLIPASE C. Caco-2 cells were pre-treated with (A–H) Gβγ inhibitor gallein (10 μM) for 30 min or (I–P) PLC inhibitor
U-73122 (10 μM) for 30 min, followed by Trp metabolite (I3A, IPyA, or IEt, 100 μM) for 24 h. The cells were then infected with EHEC (MOI 50) for 12 h, (A, I) fixed, and stained with DAPI and
Alexa Fluor 647-phalloidin. Pedestal formation = # of pedestals per Caco-2 cell (gallein: I3A, n = 944; IPyA, n = 1073; IEt, n = 905; U-73122: I3A, n = 977; IPyA, n = 1068; IEt, n = 1075
cells examined over 3 independent experiments). For box plots, interquartile ranges (IQRs, boxes), median values (line within box), whiskers (lowest and highest values within 1.5 times IQR
from the first and third quartiles), and outliers beyond whiskers (dots), are shown. B–H, J–P, Alternatively, cells were lysed and analyzed by Western blotting with the indicated antibodies.
Source data are provided in Supplementary Fig. 9. (C, E, G, H, K, M, O, P) Densitometry was performed using FIJI. Data are representative of at least 3 independent experiments, n = 3
biological replicates, bars = mean, error bars = standard deviation. Statistical analysis was performed using one-way ANOVA, followed by post-hoc Tukey multiple comparison test. Source Data
EXTENDED DATA FIG. 10 EFFECTS OF TRP METABOLITES DEPEND ON PROTEIN KINASE C (PKC). Caco-2 cells were pre-treated with pan-PKC inhibitor sotrastaurin (Sotra, 5 μM) for 30 min, followed by Trp
metabolite (I3A, IPyA, or IEt, 100 μM) for 24 h. The cells were then infected with EHEC (MOI 50) for 12 h, (A) fixed, and stained with DAPI and Alexa Fluor 647-phalloidin. Pedestal
formation = # of pedestals per Caco-2 cell (I3A, n = 912; IPyA, n = 1138; IEt, n = 1086 cells examined over 3 independent experiments). For box plots, interquartile ranges (IQRs, boxes),
median values (line within box), whiskers (lowest and highest values within 1.5 times IQR from the first and third quartiles), and outliers beyond whiskers (dots), are shown. B–F,
Alternatively, cells were lysed and analyzed by Western blotting with the indicated antibodies. Source data are provided in Supplementary Fig. 9. (C, E, F) Densitometry was performed using
FIJI. Data are representative of at least 3 independent experiments, n = 3 biological replicates, bars = mean, error bars = standard deviation. Statistical analysis was performed using
one-way ANOVA, followed by post-hoc Tukey multiple comparison test. Source Data EXTENDED DATA FIG. 11 EFFECTS OF TRP METABOLITES DEPEND ON PROTEIN KINASE C (PKC)-Θ. Caco-2 cells were
pre-treated with (A–F) isoform-selective PKC-θ inhibitor (PKCθi, 5 μM) for 24 h, or (G–L) PKC-θ was knocked down using two different siRNA duplexes (1 and 2) or a negative control siRNA
duplex (C), followed by Trp metabolite (I3A, IPyA, or IEt, 100 μM) for 24 h. The cells were then infected with EHEC (MOI 50) for 12 h, (A, G) fixed, and stained with DAPI and Alexa Fluor
647-phalloidin. Pedestal formation = # of pedestals per Caco-2 cell (PKCθi: I3A, n = 1063; IPyA, n = 1005; IEt, n = 1033; siRNA: I3A, n = 1680; IPyA, n = 1462; IEt, n = 1502 cells examined
over 3 independent experiments). For box plots, interquartile ranges (IQRs, boxes), median values (line within box), whiskers (lowest and highest values within 1.5 times IQR from the first
and third quartiles), and outliers beyond whiskers (dots), are shown. B–F, H–L, Alternatively, cells were lysed and analyzed by Western blotting with the indicated antibodies. Source data
are provided in Supplementary Fig. 9. (C, E, F, I, K, L) Densitometry was performed using FIJI. Data are representative of at least 3 independent experiments, n = 3 biological replicates,
bars = mean, error bars = standard deviation. Statistical analysis was performed using one-way ANOVA, followed by post-hoc Tukey multiple comparison test. Source Data EXTENDED DATA FIG. 12
EFFECTS OF TRP METABOLITES DEPEND ON PROTEASOMAL DEGRADATION. Caco-2 cells were pre-treated with proteasomal inhibitor MG-132 (10 μM) for 1 h, followed by Trp metabolite (I3A, IPyA, or IEt,
100 μM) for 24 h. The cells were then infected with EHEC (MOI 50) for 12 h, (A) fixed, and stained with DAPI and Alexa Fluor 647-phalloidin. Pedestal formation = # of pedestals per Caco-2
cell (I3A, n = 1079; IPyA, n = 999; IEt, n = 911 cells examined over 3 independent experiments). For box plots, interquartile ranges (IQRs, boxes), median values (line within box), whiskers
(lowest and highest values within 1.5 times IQR from the first and third quartiles), and outliers beyond whiskers (dots), are shown. B–H, Alternatively, cells were lysed and analyzed by
Western blotting with the indicated antibodies. Source data are provided in Supplementary Fig. 9. (C, E, G, H) Densitometry was performed using FIJI. Data are representative of at least 3
independent experiments, n = 3 biological replicates, bars = mean, error bars = standard deviation. Statistical analysis was performed using one-way ANOVA, followed by post-hoc Tukey
multiple comparison test. Source Data SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION Supplementary Figs. 1–9 REPORTING SUMMARY SOURCE DATA SUPPLEMENTARY FIG. 1 SOURCE DATA SUPPLEMENTARY
FIG. 2 SOURCE DATA SUPPLEMENTARY FIG. 3 SOURCE DATA SUPPLEMENTARY FIG. 4 SOURCE DATA SUPPLEMENTARY FIG. 5 SOURCE DATA SUPPLEMENTARY FIG. 6 SOURCE DATA SUPPLEMENTARY FIG. 7 SOURCE DATA
SUPPLEMENTARY FIG. 8 SOURCE DATA SOURCE DATA FIG. 1 SOURCE DATA FIG. 2 SOURCE DATA FIG. 3 SOURCE DATA FIG. 4 SOURCE DATA EXTENDED DATA FIG. 2 SOURCE DATA EXTENDED DATA FIG. 3 SOURCE DATA
EXTENDED DATA FIG. 4 SOURCE DATA EXTENDED DATA FIG. 5 SOURCE DATA EXTENDED DATA FIG. 6 SOURCE DATA EXTENDED DATA FIG. 7 SOURCE DATA EXTENDED DATA FIG. 8 SOURCE DATA EXTENDED DATA FIG. 9
SOURCE DATA EXTENDED DATA FIG. 10 SOURCE DATA EXTENDED DATA FIG. 11 SOURCE DATA EXTENDED DATA FIG. 12 RIGHTS AND PERMISSIONS Springer Nature or its licensor (e.g. a society or other partner)
holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is
solely governed by the terms of such publishing agreement and applicable law. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Scott, S.A., Fu, J. & Chang, P.V. Dopamine
receptor D2 confers colonization resistance via microbial metabolites. _Nature_ 628, 180–185 (2024). https://doi.org/10.1038/s41586-024-07179-5 Download citation * Received: 13 September
2021 * Accepted: 07 February 2024 * Published: 13 March 2024 * Issue Date: 04 April 2024 * DOI: https://doi.org/10.1038/s41586-024-07179-5 SHARE THIS ARTICLE Anyone you share the following
link with will be able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer Nature
SharedIt content-sharing initiative