Thromb Haemost 2022; 122(04): 540-551
DOI: 10.1055/s-0041-1733906
Blood Cells, Inflammation and Infection

Impact of a Vancomycin-Induced Shift of the Gut Microbiome in a Gram-Negative Direction on Plasma Factor VIII:C Levels: Results from a Randomized Controlled Trial

Gro Grimnes
1   Department of Clinical Medicine, K. G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT—The Arctic University of Norway, Tromsø, Norway
2   Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
,
1   Department of Clinical Medicine, K. G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT—The Arctic University of Norway, Tromsø, Norway
3   Division of Haemostasis and Thrombosis, Department of Haematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
,
Kristian Hindberg
1   Department of Clinical Medicine, K. G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT—The Arctic University of Norway, Tromsø, Norway
,
Mark Davids
4   Department of Vascular Medicine, Amsterdam University Medical Centers—location AMC, University of Amsterdam, Amsterdam, The Netherlands
,
Max Nieuwdorp
4   Department of Vascular Medicine, Amsterdam University Medical Centers—location AMC, University of Amsterdam, Amsterdam, The Netherlands
5   Department of Internal Medicine, Diabetes Center, Amsterdam University Medical Centers—location VUmc, Amsterdam, The Netherlands
6   Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
,
Tom E. Mollnes
1   Department of Clinical Medicine, K. G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT—The Arctic University of Norway, Tromsø, Norway
7   Research Laboratory, Nordland Hospital, Bodø, Norway
8   Department of Immunology, Oslo University Hospital and K.G. Jebsen IRC, University of Oslo, Oslo, Norway
,
Annika E. Michelsen
9   Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
10   Faculty of Medicine, University of Oslo, Oslo, Norway
,
Thor Ueland
1   Department of Clinical Medicine, K. G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT—The Arctic University of Norway, Tromsø, Norway
9   Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
10   Faculty of Medicine, University of Oslo, Oslo, Norway
,
Sigrid K. Brækkan
1   Department of Clinical Medicine, K. G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT—The Arctic University of Norway, Tromsø, Norway
2   Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
,
John-Bjarne Hansen
1   Department of Clinical Medicine, K. G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT—The Arctic University of Norway, Tromsø, Norway
2   Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
,
Vladimir Tichelaar
1   Department of Clinical Medicine, K. G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT—The Arctic University of Norway, Tromsø, Norway
3   Division of Haemostasis and Thrombosis, Department of Haematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
11   Certe Thrombosis Service, Groningen, The Netherlands
› Author Affiliations
Funding G.G. is in receipt of an independent grant from the North Norwegian Health Authorities. V.T. is in receipt of an independent grant (Mandema Stipendium) from University Medical Center Groningen, University of Groningen, the Netherlands. M.N. is supported by a ZONMW-VIDI grant 2013 (016.146.327) and CVON Young Talent grant 2012, and supported by Le Ducq consortium grant 17CVD01 and Novo Nordisk Foundation. The laboratory analyses were financially supported to T.E.M. by The Norwegian Council on Cardiovascular Disease and The Odd Fellow Foundation.

Abstract

Background Inflammation is present in several conditions associated with risk of venous thromboembolism. The gut microbiome might be a source of systemic inflammation and activation of coagulation, by translocation of lipopolysaccharides from gram-negative bacteria to the systemic circulation.

Objective To investigate whether a vancomycin-induced shift of the gut microbiome in a gram-negative direction influences systemic inflammation and plasma factor (F) VIII procoagulant activity (FVIII:C).

Methods and Results We performed a randomized controlled trial including 43 healthy volunteers aged 19 to 37 years. Twenty-one were randomized to 7 days of oral vancomycin intake and 22 served as controls. Feces and blood were sampled at baseline, the day after the end of intervention, and 3 weeks after intervention. Gut microbiome composition was assessed by amplicon sequencing. FVIII:C was measured using an activated partial thromboplastin time-based assay, cytokines were measured using multiplex technology, complement activation was measured using the enzyme-linked immunosorbent assay, and high-sensitivity C-reactive protein (CRP) was measured by an immunoturbidimetric assay. Vancomycin intake reduced gut microbiome diversity and increased the abundance of gram-negative bacteria. Change in FVIII:C in the intervention group was +4 IU/dL versus −6 IU/dL (p = 0.01) in the control group. A similar change was observed for log-transformed CRP (+0.21 mg/dL vs. −0.25 mg/dL, p = 0.04). The cytokines and complement activation markers remained similar in the two groups.

Conclusion The found slight increases in FVIII:C and CRP levels might support the hypothesis that a vancomycin-induced gram-negative shift in the gut microbiome could induce increased systemic inflammation and thereby a procoagulant state.

Author Contributions

G.G. and V.T. conceived the idea of the study. V.T. and J-.B.H. obtained funding for the study. G.G., V.T., and J-.B.H. designed the study. G.G. and V.T. wrote the protocol and obtained approval of the Regional Ethics Committee and the Clinical Research Unit at the University Hospital of North Norway. G.G. ran the trial as the principal investigator, included the volunteers, and collected the data. G.G. and K.H. performed statistical analysis on the inflammatory and coagulation outcomes. M.D. and M.N. performed statistical analysis of the microbiome data. M.D., M.N., V.T., S.B., and G.G. interpreted the results of the microbiome data. A.E.M. and T.U. performed the zonulin assays. T.E.M. analyzed the cytokines and complement activation products and interpreted the data. G.G., V.T., S.B., K.H., J-.B.H., and S.K.B. interpreted all results of all outcomes in the light of the hypothesis of the study. G.G. and S.B. drafted the manuscript with supervision of V.T., J-.B.H., and S.K.B. K.H., T.E.M., M.D., A.E.M., T.U., M.N., and S.K.B. critically reviewed the manuscript.




Publication History

Received: 02 March 2021

Accepted: 01 July 2021

Article published online:
24 August 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Heit JA. Epidemiology of venous thromboembolism. Nat Rev Cardiol 2015; 12 (08) 464-474
  • 2 Arshad N, Isaksen T, Hansen JB, Brækkan SK. Time trends in incidence rates of venous thromboembolism in a large cohort recruited from the general population. Eur J Epidemiol 2017; 32 (04) 299-305
  • 3 Esmon CT, Xu J, Lupu F. Innate immunity and coagulation. J Thromb Haemost 2011; 9 (01, Suppl 1): 182-188
  • 4 Rittirsch D, Flierl MA, Ward PA. Harmful molecular mechanisms in sepsis. Nat Rev Immunol 2008; 8 (10) 776-787
  • 5 Rasmussen LD, Dybdal M, Gerstoft J. et al. HIV and risk of venous thromboembolism: a Danish nationwide population-based cohort study. HIV Med 2011; 12 (04) 202-210
  • 6 Grainge MJ, West J, Card TR. Venous thromboembolism during active disease and remission in inflammatory bowel disease: a cohort study. Lancet 2010; 375 (9715): 657-663
  • 7 Timp JF, Cannegieter SC, Tichelaar V. et al. Antibiotic use as a marker of acute infection and risk of first and recurrent venous thrombosis. Br J Haematol 2017; 176 (06) 961-970
  • 8 Clayton TC, Gaskin M, Meade TW. Recent respiratory infection and risk of venous thromboembolism: case-control study through a general practice database. Int J Epidemiol 2011; 40 (03) 819-827
  • 9 Smeeth L, Cook C, Thomas S, Hall AJ, Hubbard R, Vallance P. Risk of deep vein thrombosis and pulmonary embolism after acute infection in a community setting. Lancet 2006; 367 (9516): 1075-1079
  • 10 Eckburg PB, Bik EM, Bernstein CN. et al. Diversity of the human intestinal microbial flora. Science 2005; 308 (5728): 1635-1638
  • 11 Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature 2007; 449 (7164): 804-810
  • 12 Zhao L. The gut microbiota and obesity: from correlation to causality. Nat Rev Microbiol 2013; 11 (09) 639-647
  • 13 Smits LP, Bouter KEC, de Vos WM, Borody TJ, Nieuwdorp M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology 2013; 145 (05) 946-953
  • 14 Khoruts A, Dicksved J, Jansson JK, Sadowsky MJ. Changes in the composition of the human fecal microbiome after bacteriotherapy for recurrent Clostridium difficile-associated diarrhea. J Clin Gastroenterol 2010; 44 (05) 354-360
  • 15 Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer 2013; 13 (11) 800-812
  • 16 Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol 2008; 6 (11) e280
  • 17 Isaac S, Scher JU, Djukovic A. et al. Short- and long-term effects of oral vancomycin on the human intestinal microbiota. J Antimicrob Chemother 2017; 72 (01) 128-136
  • 18 Guo S, Al-Sadi R, Said HM, Ma TY. Lipopolysaccharide causes an increase in intestinal tight junction permeability in vitro and in vivo by inducing enterocyte membrane expression and localization of TLR-4 and CD14. Am J Pathol 2013; 182 (02) 375-387
  • 19 Cani PD, Amar J, Iglesias MA. et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007; 56 (07) 1761-1772
  • 20 Hersoug LG, Møller P, Loft S. Gut microbiota-derived lipopolysaccharide uptake and trafficking to adipose tissue: implications for inflammation and obesity. Obes Rev 2016; 17 (04) 297-312
  • 21 Carnevale R, Raparelli V, Nocella C. et al. Gut-derived endotoxin stimulates factor VIII secretion from endothelial cells. Implications for hypercoagulability in cirrhosis. J Hepatol 2017; 67 (05) 950-956
  • 22 Reitsma PH, Branger J, Van Den Blink B, Weijer S, Van Der Poll T, Meijers JCM. Procoagulant protein levels are differentially increased during human endotoxemia. J Thromb Haemost 2003; 1 (05) 1019-1023
  • 23 Koster T, Blann AD, Briët E, Vandenbroucke JP, Rosendaal FR. Role of clotting factor VIII in effect of von Willebrand factor on occurrence of deep-vein thrombosis. Lancet 1995; 345 (8943): 152-155
  • 24 Kyrle PA, Minar E, Hirschl M. et al. High plasma levels of factor VIII and the risk of recurrent venous thromboembolism. N Engl J Med 2000; 343 (07) 457-462
  • 25 Horvei LD, Grimnes G, Hindberg K. et al. C-reactive protein, obesity, and the risk of arterial and venous thrombosis. J Thromb Haemost 2016; 14 (08) 1561-1571
  • 26 Mold C, Gewurz H, Du Clos TW. Regulation of complement activation by C-reactive protein. Immunopharmacology 1999; 42 (1–3): 23-30
  • 27 Høiland II, Liang RA, Braekkan SK. et al. Complement activation assessed by the plasma terminal complement complex and future risk of venous thromboembolism. J Thromb Haemost 2019; 17 (06) 934-943
  • 28 Nørgaard I, Nielsen SF, Nordestgaard BG. Complement C3 and high risk of venous thromboembolism: 80517 individuals from the Copenhagen general population study. Clin Chem 2016; 62 (03) 525-534
  • 29 Swerdlow DI, Holmes MV, Kuchenbaecker KB. et al; Interleukin-6 Receptor Mendelian Randomisation Analysis (IL6R MR) Consortium. The interleukin-6 receptor as a target for prevention of coronary heart disease: a mendelian randomisation analysis. Lancet 2012; 379 (9822): 1214-1224
  • 30 Christiansen SC, Naess IA, Cannegieter SC, Hammerstrøm J, Rosendaal FR, Reitsma PH. Inflammatory cytokines as risk factors for a first venous thrombosis: a prospective population-based study. PLoS Med 2006; 3 (08) e334
  • 31 Vormittag R, Hsieh K, Kaider A. et al. Interleukin-6 and interleukin-6 promoter polymorphism (-174) G > C in patients with spontaneous venous thromboembolism. Thromb Haemost 2006; 95 (05) 802-806
  • 32 Matos MF, Lourenço DM, Orikaza CM, Bajerl JAH, Noguti MAE, Morelli VM. The role of IL-6, IL-8 and MCP-1 and their promoter polymorphisms IL-6 -174GC, IL-8 -251AT and MCP-1 -2518AG in the risk of venous thromboembolism: a case-control study. Thromb Res 2011; 128 (03) 216-220
  • 33 Vrieze A, Out C, Fuentes S. et al. Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J Hepatol 2014; 60 (04) 824-831
  • 34 Rubinstein E, Keynan Y. Vancomycin revisited - 60 years later. Front Public Health 2014; 2 (OCT): 217
  • 35 Fasano A. All disease begins in the (leaky) gut: role of zonulin-mediated gut permeability in the pathogenesis of some chronic inflammatory diseases. F1000 Res 2020; 9: 1-13
  • 36 Citronberg JS, Wilkens LR, Lim U. et al. Reliability of plasma lipopolysaccharide-binding protein (LBP) from repeated measures in healthy adults. Cancer Causes Control 2016; 27 (09) 1163-1166
  • 37 Sprenger HG, Lisman JA, Mulder AB. et al. Hypercoagulability and hypofibrinolysis in patients with human immunodeficiency virus infection partially resolve after antiretroviral treatment. J Thromb Haemost 2013; 11: 1003
  • 38 Costea PI, Zeller G, Sunagawa S. et al. Towards standards for human fecal sample processing in metagenomic studies. Nat Biotechnol 2017; 35 (11) 1069-1076
  • 39 Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Microbiol 2013; 79 (17) 5112-5120
  • 40 Quast C, Pruesse E, Yilmaz P. et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res 2013; 41 (Database issue, D1): D590-D596
  • 41 McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One 2013; 8 (04) e61217
  • 42 Oksanen JF, Guillaume Blanchet RK, Legendre P. et al. Package ‘vegan’. R Packag. 2019
  • 43 Kembel SW, Cowan PD, Helmus MR. et al. Picante: R tools for integrating phylogenies and ecology. Bioinformatics 2010; 26 (11) 1463-1464
  • 44 Anderson MJ. A new method for non-parametric multivariate analysis of variance. Austral Ecol 2011; 26: 32-46
  • 45 de Maat MPM, van Schie M, Kluft C, Leebeek FWG, Meijer P. Biological variation of hemostasis variables in thrombosis and bleeding: consequences for performance specifications. Clin Chem 2016; 62 (12) 1639-1646
  • 46 Reijnders D, Goossens GH, Hermes GDA. et al. Effects of gut microbiota manipulation by antibiotics on host metabolism in obese humans: a randomized double-blind placebo-controlled trial. Cell Metab 2016; 24 (01) 63-74
  • 47 Massier L, Chakaroun R, Kovacs P, Heiker JT. Blurring the picture in leaky gut research: how shortcomings of zonulin as a biomarker mislead the field of intestinal permeability. Gut 2020; 0 (00) 1-2
  • 48 Jäckel S, Kiouptsi K, Lillich M. et al. Gut microbiota regulate hepatic von Willebrand factor synthesis and arterial thrombus formation via Toll-like receptor-2. Blood 2017; 130 (04) 542-553
  • 49 Alzahrani J, Hussain T, Simar D. et al. Inflammatory and immunometabolic consequences of gut dysfunction in HIV: Parallels with IBD and implications for reservoir persistence and non-AIDS comorbidities. EBioMedicine 2019; 46: 522-531
  • 50 Leech B, McIntyre E, Steel A, Sibbritt D. Risk factors associated with intestinal permeability in an adult population: a systematic review. Int J Clin Pract 2019; 73 (10) e13385