Altered microbiota-host metabolic cross talk preceding neutropenic fever in patients with acute leukemia

Armin Rashidi, Maryam Ebadi, Tauseef Ur Rehman, Heba Elhusseini, Harika Nalluri, Thomas Kaiser, Sivapriya Ramamoorthy, Shernan G Holtan, Alexander Khoruts, Daniel J Weisdorf, Christopher Staley, Armin Rashidi, Maryam Ebadi, Tauseef Ur Rehman, Heba Elhusseini, Harika Nalluri, Thomas Kaiser, Sivapriya Ramamoorthy, Shernan G Holtan, Alexander Khoruts, Daniel J Weisdorf, Christopher Staley

Abstract

Despite antibiotic prophylaxis, most patients with acute leukemia receiving mucotoxic chemotherapy develop neutropenic fever (NF), many cases of which remain without a documented etiology. Antibiotics disrupt the gut microbiota, with adverse clinical consequences, such as Clostridioides difficile infection. A better understanding of NF pathogenesis could inform the development of novel therapeutics without deleterious effects on the microbiota. We hypothesized that metabolites absorbed from the gut to the bloodstream modulate pyrogenic and inflammatory pathways. Longitudinal profiling of the gut microbiota in 2 cohorts of patients with acute leukemia showed that Akkermansia expansion in the gut was associated with an increased risk for NF. As a prototype mucolytic genus, Akkermansia may influence the absorption of luminal metabolites; thus, its association with NF supported our metabolomics hypothesis. Longitudinal profiling of the serum metabolome identified a signature associated with gut Akkermansia and 1 with NF. Importantly, these 2 signatures overlapped in metabolites in the γ-glutamyl cycle, suggesting oxidative stress as a mediator involved in Akkermansia-related NF. In addition, the level of gut microbial-derived indole compounds increased after Akkermansia expansion and decreased before NF, suggesting their role in mediating the anti-inflammatory effects of Akkermansia, as seen predominantly in healthy individuals. These results suggest that Akkermansia regulates microbiota-host metabolic cross talk by modulating the mucosal interface. The clinical context, including factors influencing microbiota composition, determines the type of metabolites absorbed through the gut barrier and their net effect on the host. Our findings identify novel aspects of NF pathogenesis that could be targets for precision therapeutics. This trial was registered at www.clinicaltrials.gov as #NCT03316456.

© 2021 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Consort diagram of patients, samples, and major analytic steps. All serum samples from cohort 1 and some from cohort 2 had been exhausted in prior projects; thus, the remaining samples from cohort 2 were used for analysis.
Figure 2.
Figure 2.
Gut microbiome and NF in patients with AML. (A) Relative abundances of genera in the 2 cohorts. Mean relative abundances across all samples are shown. Genera with a relative abundance < 1% are shown in aggregate. (B) LDA coupled with effect size measurements (LEfSe) in the 2 cohorts. Differentially abundant OTUs (LDA score > 3.0; P < .05) in high-risk samples are shown as red bars to the right; those more abundant in low-risk samples are shown as blue bars to the left. “f” and “g” in the plot for cohort 2 indicate family and genus, respectively. All significant taxa in cohort 1 were classifiable to the genus level, thus “f” and “g” notations are not used. High- and low-risk samples are defined by whether were followed or not by a new episode of NF within 7 days, respectively. (C) Relative abundance of Akkermansia in stool samples followed vs not followed by a new episode of NF within 7 days, with the 2 cohorts combined. A horizontal jitter was added for better visualization. The P value was derived from Welch’s t test. (D) Receiver operating characteristic curve analysis for the relative abundance of Akkermansia as a predictor of a new episode of NF within the next 7 days. AUC, area under the curve; CI, confidence interval.
Figure 3.
Figure 3.
Serum metabolomics associated with NF. (A) Distribution of serum metabolites into pathways. (B) Volcano plot showing the magnitude and significance of metabolomics changes within 24 hours before NF. High-risk (preceding NF within 24 hours) and low-risk samples (not preceding NF within 24 hours) were compared. Points to the right (left) of the vertical line (no change) represent metabolites that were increased (decreased) before NF. Select metabolites of interest in this work are labeled (the orange circle representing p-cresol sulfate is nearly completely hidden behind 2 green circles). The horizontal lines represent q = .05 and q = .1, above which the metabolites are statistically significant according to the corresponding threshold. p values from Welch’s t-test were corrected for multiple testing to derive the q values. (C) Distribution of metabolites with q < .1 in (B) into pathways. (D) Overrepresentation analysis using subpathways. Metabolites with q < .05 in (B) were considered for overrepresentation of their subpathways using a hypergeometric test with a corrected P value threshold of .05. Each central node represents an overrepresented subpathway, and the peripheral nodes connected to the central node represent the metabolites within that subpathway that are significant in (B). The number of these metabolites in each overrepresented subpathway is proportional to the size of the central node. The complete list of metabolites is provided in supplemental Data 1.
Figure 4.
Figure 4.
Serum metabolomics associated with gut Akkermansia. (A) Distribution of serum metabolites into pathways, provided to facilitate comparison with (C). (B) Volcano plot showing the strength and significance of the association between serum metabolite levels (outcome variable) and abundance of gut Akkermansia (predictor) in the nearest prior stool sample. For each metabolite, a separate mixed effect regression was created, adjusting for sex, use of parenteral nutrition, serum sample day, and patient number (random effect). The regression coefficient for Akkermansia was considered its effect size (x-axis), and the corresponding P value was corrected for multiple testing to derive the q value (y-axis). Select metabolites of interest in this work are labeled. The horizontal lines represent q = .05 and q = .1, above which the metabolites are statistically significant according to the corresponding threshold. (C) Distribution of metabolites with q < .1 in (B) into pathways. (D) Overrepresentation analysis using subpathways. Metabolites with q < .05 in (B) were considered for overrepresentation of their subpathways using a hypergeometric test with a corrected P value threshold of .05. Each central node represents an overrepresented subpathway, and the peripheral nodes connected to the central node represent the metabolites within that subpathway that are significant in (B). The number of these metabolites in each overrepresented subpathway is proportional to the size of the central node. The complete list of metabolites is provided in supplemental Data 2.
Figure 5.
Figure 5.
Changes in the level of select metabolites before NF. (A) A total of 310 serum metabolites were associated with NF, 51 metabolites were associated with abundance of gut Akkermansia, and 9 metabolites were associated with both. (B-I) Serum samples preceding vs not preceding NF within 24 hours were compared for select metabolites using Welch’s t test. (B-E) show colonic bacteria–derived metabolites from dietary tyrosine and tryptophan degradation. (F-H) show select metabolites in the γ-glutamyl cycle. Metabolites in (B-H) were measured by UPLC-MS/MS. GGT in (I) was measured fluorometrically. The boxplots show the median and interquartile range.

References

    1. Sickles EA, Greene WH, Wiernik PH.. Clinical presentation of infection in granulocytopenic patients. Arch Intern Med. 1975;135(5):715-719.
    1. Hansen B-A, Wendelbo Ø, Bruserud Ø, Hemsing AL, Mosevoll KA, Reikvam H.. Febrile neutropenia in acute leukemia. Epidemiology, etiology, pathophysiology and treatment. Mediterr J Hematol Infect Dis. 2020;12(1):e2020009.
    1. Bucaneve G, Micozzi A, Menichetti F, et al. ; Gruppo Italiano Malattie Ematologiche dell’Adulto (GIMEMA) Infection Program . Levofloxacin to prevent bacterial infection in patients with cancer and neutropenia. N Engl J Med. 2005;353(10):977-987.
    1. Peseski AM, McClean M, Green SD, Beeler C, Konig H.. Management of fever and neutropenia in the adult patient with acute myeloid leukemia. Expert Rev Anti Infect Ther. 2021;19(3):359-378.
    1. Gyssens IC, Kern WV, Livermore DM; ECIL-4, a joint venture of EBMT, EORTC, ICHS and ESGICH of ESCMID .The role of antibiotic stewardship in limiting antibacterial resistance among hematology patients. Haematologica. 2013;98(12):1821-1825.
    1. Spellberg B. The new antibiotic mantra - “shorter is better”. JAMA Intern Med. 2016;176(9):1254-1255.
    1. Belkaid Y, Harrison OJ.. Homeostatic immunity and the microbiota. Immunity. 2017;46(4):562-576.
    1. Belkaid Y, Hand TW.. Role of the microbiota in immunity and inflammation. Cell. 2014;157(1):121-141.
    1. Galloway-Peña JR, Shi Y, Peterson CB, et al. . Gut microbiome signatures are predictive of infectious risk following induction therapy for acute myeloid leukemia. Clin Infect Dis. 2020;71(1):63-71.
    1. Rashidi A, Kaiser T, Shields-Cutler R, et al. . Dysbiosis patterns during re-induction/salvage versus induction chemotherapy for acute leukemia. Sci Rep. 2019;9(1):6083.
    1. Rashidi A, Kaiser T, Graiziger C, et al. . Specific gut microbiota changes heralding bloodstream infection and neutropenic fever during intensive chemotherapy. Leukemia. 2020;34(1):312-316.
    1. Wikoff WR, Anfora AT, Liu J, et al. . Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci USA. 2009;106(10):3698-3703.
    1. Visconti A, Le Roy CI, Rosa F, et al. . Interplay between the human gut microbiome and host metabolism. Nat Commun. 2019;10(1):4505.
    1. Derrien M, Vaughan EE, Plugge CM, de Vos WM.. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol. 2004;54(Pt 5):1469-1476.
    1. Derrien M, Collado MC, Ben-Amor K, Salminen S, de Vos WM.. The mucin degrader Akkermansia muciniphila is an abundant resident of the human intestinal tract. Appl Environ Microbiol. 2008;74(5):1646-1648.
    1. Rashidi A, Kaiser T, Graiziger C, et al. . Gut dysbiosis during antileukemia chemotherapy versus allogeneic hematopoietic cell transplantation. Cancer. 2020;126(7):1434-1447.
    1. Freifeld AG, Bow EJ, Sepkowitz KA, et al. ; Infectious Diseases Society of America . Clinical practice guideline for the use of antimicrobial agents in neutropenic patients with cancer: 2010 update by the infectious diseases society of america. Clin Infect Dis. 2011;52(4):e56-e93.
    1. Benjamini Y, Hochberg Y.. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol. 1995;57(1):289-300.
    1. Segata N, Izard J, Waldron L, et al. . Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(6):R60.
    1. Keizer RJ, Jansen RS, Rosing H, et al. . Incorporation of concentration data below the limit of quantification in population pharmacokinetic analyses. Pharmacol Res Perspect. 2015;3(2):e00131.
    1. Leong HS, Kipling D. Text-based over-representation analysis of microarray gene lists with annotation bias. Nucleic Acids Res. 2009;37(11):e79.
    1. van den Berg RA, Hoefsloot HCJ, Westerhuis JA, Smilde AK, van der Werf MJ.. Centering, scaling, and transformations: improving the biological information content of metabolomics data. BMC Genomics. 2006;7(1):142.
    1. Ridlon JM, Harris SC, Bhowmik S, Kang D-J, Hylemon PB.. Consequences of bile salt biotransformations by intestinal bacteria [published correction appears in Gut Microbe. 2016; 7(3):262]. Gut Microbes. 2016;7(1):22-39.
    1. Smith EA, Macfarlane GT.. Enumeration of human colonic bacteria producing phenolic and indolic compounds: effects of pH, carbohydrate availability and retention time on dissimilatory aromatic amino acid metabolism. J Appl Bacteriol. 1996;81(3):288-302.
    1. Sumner LW, Amberg A, Barrett D, et al. . Proposed minimum reporting standards for chemical analysis Chemical Analysis Working Group (CAWG) Metabolomics Standards Initiative (MSI). Metabolomics. 2007;3(3):211-221.
    1. Mair RD, Sirich TL, Plummer NS, Meyer TW.. Characteristics of colon-derived uremic solutes. Clin J Am Soc Nephrol. 2018;13(9):1398-1404.
    1. Lees HJ, Swann JR, Wilson ID, Nicholson JK, Holmes E.. Hippurate: the natural history of a mammalian-microbial cometabolite. J Proteome Res. 2013;12(4):1527-1546.
    1. Depommier C, Everard A, Druart C, et al. . Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med. 2019;25(7):1096-1103.
    1. Everard A, Belzer C, Geurts L, et al. . Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci USA. 2013;110(22):9066-9071.
    1. Ballatori N, Krance SM, Marchan R, Hammond CL. Plasma membrane glutathione transporters and their roles in cell physiology and pathophysiology. Mol Aspects Med. 2009;30(1-2):13-28.
    1. Zhang H, Forman HJ.. Redox regulation of γ-glutamyl transpeptidase. Am J Respir Cell Mol Biol. 2009;41(5):509-515.
    1. Mardinoglu A, Shoaie S, Bergentall M, et al. . The gut microbiota modulates host amino acid and glutathione metabolism in mice. Mol Syst Biol. 2015; 11(10):834.
    1. Suzuki H, Hashimoto W, Kumagai H.. Escherichia coli K-12 can utilize an exogenous gamma-glutamyl peptide as an amino acid source, for which gamma-glutamyltranspeptidase is essential. J Bacteriol. 1993;175(18):6038-6040.
    1. Suzuki H, Kumagai H, Tochikura T.. gamma-Glutamyltranspeptidase from Escherichia coli K-12: formation and localization. J Bacteriol. 1986;168(3):1332-1335.
    1. Alam A, Leoni G, Wentworth CC, et al. . Redox signaling regulates commensal-mediated mucosal homeostasis and restitution and requires formyl peptide receptor 1. Mucosal Immunol. 2014;7(3):645-655.
    1. Jones RM, Luo L, Ardita CS, et al. . Symbiotic lactobacilli stimulate gut epithelial proliferation via Nox-mediated generation of reactive oxygen species. EMBO J. 2013;32(23):3017-3028.
    1. Neish AS, Jones RM.. Redox signaling mediates symbiosis between the gut microbiota and the intestine. Gut Microbes. 2014;5(2):250-253.
    1. Dinarello CA. Cytokines as endogenous pyrogens. J Infect Dis. 1999;179(suppl 2):S294-S304.
    1. Reuter S, Schnekenburger M, Cristofanon S, et al. . Tumor necrosis factor α induces γ-glutamyltransferase expression via nuclear factor-kappaB in cooperation with Sp1. Biochem Pharmacol. 2009;77(3):397-411.
    1. Riedel W, Maulik G.. Fever: an integrated response of the central nervous system to oxidative stress. Mol Cell Biochem. 1999;196(1-2):125-132.
    1. Hou C-C, Lin H, Chang C-P, Huang W-T, Lin M-T.. Oxidative stress and pyrogenic fever pathogenesis. Eur J Pharmacol. 2011;667(1-3):6-12.
    1. Roager HM, Licht TR.. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018;9(1):3294.
    1. Schirmer M, Smeekens SP, Vlamakis H, et al. . Linking the human gut microbiome to inflammatory cytokine production capacity [published correction appears in Cell. 2016;167(7):1897]. Cell. 2016;167(4):1125-1136.e8.
    1. Wlodarska M, Luo C, Kolde R, et al. . Indoleacrylic acid produced by commensal Peptostreptococcus species suppresses inflammation. Cell Host Microbe. 2017;22(1):25-37.e6.
    1. Png CW, Lindén SK, Gilshenan KS, et al. . Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol. 2010;105(11):2420-2428.
    1. Koropatkin NM, Cameron EA, Martens EC.. How glycan metabolism shapes the human gut microbiota. Nat Rev Microbiol. 2012;10(5):323-335.
    1. Verhoog S, Taneri PE, Roa Díaz ZM, et al. . Dietary factors and modulation of bacteria strains of Akkermansia muciniphila and Faecalibacterium prausnitzii: a systematic review. Nutrients. 2019;11(7):1565.
    1. Desai MS, Seekatz AM, Koropatkin NM, et al. . A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. 2016;167(5):1339-1353.e21.
    1. Ribera J-M, Oriol A, Morgades M, et al. . Treatment of high-risk Philadelphia chromosome-negative acute lymphoblastic leukemia in adolescents and adults according to early cytologic response and minimal residual disease after consolidation assessed by flow cytometry: final results of the PETHEMA ALL-AR-03 trial. J Clin Oncol. 2014;32(15):1595-1604.
    1. Chalandon Y, Thomas X, Hayette S, et al. ; Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) . Randomized study of reduced-intensity chemotherapy combined with imatinib in adults with Ph-positive acute lymphoblastic leukemia [published correction appears in Blood. 2015;126(10):1261]. Blood. 2015;125(24):3711-3719.

Source: PubMed

3
Subskrybuj