Iron at the host-microbe interface

https://doi.org/10.1016/j.mam.2020.100895Get rights and content

Abstract

Iron is an essential micronutrient for nearly all living organisms. In addition to facilitating redox reactions, iron is bound by metalloproteins that participate in a variety of biological processes. As the bioavailability of free iron in host environments is extremely low, iron lies at the center of a battle for nutrients between microbes and their host. Mucosal surfaces such as the respiratory and gastrointestinal tracts are constantly exposed to commensal and pathogenic microorganisms. Whereas a key strategy of mammalian antimicrobial defense is to deprive microbes of iron, pathogens and some commensals have evolved effective strategies to circumvent iron limitation. Here we provide an overview of mechanisms underpinning the tug-of-war for iron between microbes and their host, with a particular focus on mucosal surfaces.

Introduction

Iron (Fe) is a vital micronutrient for nearly all forms of life, and is involved in critical physiological processes including oxygen transportation, energy metabolism, and DNA replication (Lieu et al., 2001). By existing primarily in two oxidation states, ferrous (Fe2+) and ferric (Fe3+) iron, this element has the flexibility to serve as a key cofactor in many enzymes as well as play a crucial role in facilitating a variety of redox reactions. Nevertheless, despite iron's essential roles in the beneficial chemistry of life, its redox potential can negatively impact biological molecules and cells; for example, the Fenton reaction involving Fe2+ and hydrogen peroxide yields reactive oxygen species (ROS) that can damage biomolecules including lipids, proteins, and DNA. Oxidative stress, in turn, is linked to aging, cardiovascular disorders, neurodegenerative diseases, and cancer (Liu et al., 2018). To minimize iron's reactivity and toxicity, the vast majority of iron in mammals is found in intracellular compartments, complexed with hemoproteins (e.g., hemoglobin, myoglobin), or the iron-storage protein ferritin (Lieu et al., 2001). Thus, the bioavailability of free iron is low in the mammalian environment, even during homeostasis. Moreover, in a process known as nutritional immunity, extracellular and intracellular iron is further decreased during infection to prevent the outgrowth of pathogens (Hood and Skaar, 2012). Successful pathogens have therefore evolved strategies to compete for and obtain iron from the host in order to replicate and thrive despite limited nutrient availability.

Mucosal surfaces constitute a physical barrier between the external environment and the inner body. During homeostasis, many of these surfaces are colonized by an assortment of microbes that are collectively referred to as the commensal microbiota, and typically include non-pathogenic bacteria, fungi, archaea, and viruses (Lozupone et al., 2012). Among the body sites with a commensal microbiota, the gastrointestinal (GI) tract, and in particular the colon, is home to the body's largest and most diverse microbial community. The host and its commensal microbiota have evolved a mutualistic interdependency that significantly contributes to many aspects of the host's normal physiology, ranging from metabolism, to resisting pathogen colonization (Dethlefsen et al., 2007), to shaping and maintaining the host's immune system (Belkaid and Hand, 2014). The host, in turn, provides its microbiota with nutrients and environments suitable for growth. Even so, not all microbes are able to colonize and/or flourish in a given host, as the availability of specific nutrients controls the abundance of certain microbes (Pereira and Berry, 2017). The ability to acquire and utilize nutrients is thus essential for commensal microbes to stably colonize their host.

Despite this coevolved mutualism, the mucosal immune system must still prevent microorganisms, be they commensals or pathogens, from disseminating beyond mucosal surfaces. On this front, iron sequestration by the host represents a conserved innate immune mechanism for limiting microbial growth. Nevertheless, pathogens have evolved strategies to circumvent these defenses. Here we provide an overview of mechanisms underpinning the tug-of-war for iron between microbes and their host, which ultimately determines the health of the host and the survival of pathogenic microbes.

Section snippets

Iron regulation in humans

Regulation of iron metabolism is a balance of iron absorption, recycling, and loss. Iron levels must be tightly controlled to avoid iron deficiency (which can result in anemia) and to prevent toxicity due to excess iron. Trafficking and systemic metabolism of iron have been the subject of excellent reviews (e.g., (Hentze et al., 2010)). In this section, we provide a brief synopsis. Dietary iron is generally acquired as heme or non-heme iron, and is absorbed in the duodenum and proximal jejunum (

Iron limitation at mucosal surfaces during health and disease

A number of mechanisms contribute to defense at mucosal surfaces, including colonization resistance mediated by the microbiota (Sassone-Corsi and Raffatellu, 2015) and microbial growth restriction mediated by nutritional immunity (Hood and Skaar, 2012). Iron represents an important battleground in both of these processes as it is a key nutrient that must be scavenged from the host environment by commensals and by pathogens. On the host side, intestinal homeostasis and restriction of pathogen

Microbial iron metabolism, with a focus on Gram-negative bacteria

With the exception of some non-pathogenic lactic acid bacteria and the Lyme disease pathogen Borrelia burgdorferi, most microorganisms rely on micromolar iron concentrations for metabolism and replication (Archibald, 1983; Posey and Gherardini, 2000; Weinberg, 1978). However, iron in the mammalian host is largely bound to host proteins (e.g., ferritin, Tf, Lf, hemoproteins), thereby reducing iron's toxicity and accessibility. As such, microbes have evolved a variety of iron acquisition

Regulation of mucosal iron metabolism during infection and inflammation

Mucosal surfaces including the eyes and oropharynx, as well as the GI, respiratory, and urogenital tracts, use various antimicrobial defense strategies that interfere with microbial adhesion and survival. Among these defenses is the upregulation of Lf and Lcn2. Although both host proteins play additional roles in other cells and body sites (Kruzel et al., 2017; Moschen et al., 2017), here we focus on their roles at mucosal surfaces.

The respiratory tract

Bacterial communities of the respiratory tract are more dynamic, albeit less well characterized, than those of the gut (Huffnagle et al., 2017). Similar to the gut, however, iron acquisition is also important for bacterial pathogens that infect the airways. Correspondingly, Lcn2 is also highly upregulated in human pulmonary tissue during bacterial pneumonia (Warszawska et al., 2013). However, whether this increased expression of Lcn2 results in beneficial or detrimental outcomes strongly

Exploiting microbial iron acquisition for therapy and diagnosis

As many pathogens require iron during infection, their means of iron acquisition constitute attractive therapeutic targets (Fig. 4). For example, as described above, many pathogens require siderophores for iron uptake. As such, siderophore immunization might be a promising approach to limit microbial iron acquisition during infection. To this end, immunization against yersiniabactin and aerobactin in the context of UTIs protected from systemic infection with UPEC (Fig. 4a) (Mike et al., 2016).

Conclusions

With antimicrobial resistance posing an increasing threat to humans, alternative strategies to combat infections are in urgent need. As iron is a key micronutrient for most microbes, its abundance is tightly controlled by the host in order to restrict microbial growth. In addition to host-encoded mechanisms of iron sequestration, the microbiota also contributes to reduce iron availability to pathogens. Thus, to colonize the host, pathogens often deploy a variety of virulence factors that evade

Acknowledgements

Work in MR lab is supported by Public Health Service Grants AI126277, AI114625, AI145325, by the Chiba University-UCSD Center for Mucosal Immunology, Allergy, and Vaccines, and by the UCSD Department of Pediatrics. M.R. also holds an Investigator in the Pathogenesis of Infectious Disease Award from the Burroughs Wellcome Fund. RRG was partly supported by a fellowship from the Max Kade Foundation and by a fellowship from the Crohn's and Colitis Foundation. Figures were created with BioRender.com.

References (90)

  • A.R. Moschen et al.

    Lipocalin 2 protects from inflammation and tumorigenesis associated with gut microbiota alterations

    Cell Host Microbe

    (2016)
  • A.R. Moschen et al.

    Lipocalin-2: a master mediator of intestinal and metabolic inflammation

    Trends Endocrinol. Metab.

    (2017)
  • M. Raffatellu et al.

    Lipocalin-2 resistance confers an advantage to Salmonella enterica serotype typhimurium for growth and survival in the inflamed intestine

    Cell Host Microbe

    (2009)
  • C.R. Shaler et al.

    The unique lifestyle of Crohn's disease-associated adherent-invasive Escherichia coli

    J. Mol. Biol.

    (2019)
  • M. Valdebenito et al.

    Environmental factors influence the production of enterobactin, salmochelin, aerobactin, and yersiniabactin in Escherichia coli strain Nissle 1917

    Int. J. Med. Microbiol.

    (2006)
  • E.M. Zygiel et al.

    The human innate immune protein calprotectin induces iron starvation responses in Pseudomonas aeruginosa

    J. Biol. Chem.

    (2019)
  • R.S. Almeida et al.

    Candida albicans iron acquisition within the host

    FEMS Yeast Res.

    (2009)
  • M.A. Bachman et al.

    Klebsiella pneumoniae yersiniabactin promotes respiratory tract infection through evasion of lipocalin 2

    Infect. Immun.

    (2011)
  • M.A. Bachman et al.

    Interaction of lipocalin 2, transferrin, and siderophores determines the replicative niche of Klebsiella pneumoniae during pneumonia

    mBio

    (2012)
  • G. Bairwa et al.

    Iron acquisition in fungal pathogens of humans

    Met. Integr. Biometal Sci.

    (2017)
  • A.J. Bäumler et al.

    Interactions between the microbiota and pathogenic bacteria in the gut

    Nature

    (2016)
  • N.J. Bessman et al.

    Dendritic cell–derived hepcidin sequesters iron from the microbiota to promote mucosal healing

    Science

    (2020)
  • V. Braun et al.

    Sideromycins: tools and antibiotics

    Biometals

    (2009)
  • R. Caruso et al.

    Host–microbiota interactions in inflammatory bowel disease

    Nat. Rev. Immunol.

    (2020)
  • M. Caza et al.

    Shared and distinct mechanisms of iron acquisition by bacterial and fungal pathogens of humans

    Front. Cell. Infect. Microbiol.

    (2013)
  • M. Constante et al.

    Dietary heme induces gut dysbiosis, aggravates colitis, and potentiates the development of adenomas in mice

    Front. Microbiol.

    (2017)
  • B.D. Corbin et al.

    Metal chelation and inhibition of bacterial growth in tissue abscesses

    Science

    (2008)
  • J.H. Crosa

    Genetics and molecular biology of siderophore-mediated iron transport in bacteria

    Microbiol. Mol. Biol. Rev.

    (1989)
  • N.K. Das et al.

    Microbial metabolite signaling is required for systemic iron homeostasis

    Cell Metabol.

    (2020)
  • L. Dethlefsen et al.

    An ecological and evolutionary perspective on human–microbe mutualism and disease

    Nature

    (2007)
  • B. Dogan et al.

    Inflammation-associated adherent-invasive Escherichia coli are enriched in pathways for use of propanediol and iron and M-cell translocation

    Inflamm. Bowel Dis.

    (2014)
  • H. Drakesmith et al.

    Hepcidin and the iron-infection Axis

    Science

    (2012)
  • M. Ferrante et al.

    Outcome after proctocolectomy with ileal pouch-anal anastomosis for ulcerative colitis

    Inflamm. Bowel Dis.

    (2008)
  • M.A. Fischbach et al.

    The pathogen-associated iroA gene cluster mediates bacterial evasion of lipocalin 2

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2006)
  • T.H. Flo et al.

    Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron

    Nature

    (2004)
  • E.C. Garcia et al.

    Redundancy and specificity of Escherichia coli iron acquisition systems during urinary tract infection

    Infect. Immun.

    (2011)
  • P. Gionchetti et al.

    The role of antibiotics and probiotics in pouchitis

    Ann. Gastroenterol.

    (2012)
  • C. Gottschick et al.

    The urinary microbiota of men and women and its changes in women during bacterial vaginosis and antibiotic treatment

    Microbiome

    (2017)
  • H. Haas et al.

    An iron-mimicking, trojan horse-entering fungi—has the time come for molecular imaging of fungal infections?

    PLoS Pathog.

    (2015)
  • C.W. Hesseltine et al.

    Coprogen, a new growth factor for coprophilic fungi

    J. Am. Chem. Soc.

    (1952)
  • V.I. Holden et al.

    Klebsiella pneumoniae siderophores induce inflammation, bacterial dissemination, and HIF-1α stabilization during pneumonia

    mBio

    (2016)
  • M.I. Hood et al.

    Nutritional immunity: transition metals at the pathogen–host interface

    Nat. Rev. Microbiol.

    (2012)
  • C. Huttenhower et al.

    Structure, function and diversity of the healthy human microbiome

    Nature

    (2012)
  • T. Jaeggi et al.

    Iron fortification adversely affects the gut microbiome, increases pathogen abundance and induces intestinal inflammation in Kenyan infants

    Gut

    (2015)
  • G.P. Jarosik et al.

    Acquisition of iron by Gardnerella vaginalis

    Infect. Immun.

    (1998)
  • Cited by (22)

    • Discovery of chromene compounds as inhibitors of PvdQ acylase of Pseudomonas aeruginosa

      2022, Microbes and Infection
      Citation Excerpt :

      It is important to note that the inhibitor as it is presented in this work does not actively kill P. aeruginosa cells but impairs their ability to survive in the host environment. This is also due to a tightly regulated iron homeostasis of the host [29]. We could observe that the supplementation of pyoverdine could recover the virulence in the pvdQ deficient mutant (Fig. S4).

    • Baseline iron status and presence of anaemia determine the course of systemic Salmonella infection following oral iron supplementation in mice

      2021, EBioMedicine
      Citation Excerpt :

      Salmonella Typhimurium (S.Tm) is a facultative intracellular, Gram-negative pathogen replicating within macrophages and it is highly dependent on iron for achieving full virulence [17–19]. Because of the central role of iron in host-pathogen interaction, immune system driven restriction of iron from invading microbes has been identified as a central anti-microbial defence strategy for which the term nutritional immunity has been coined [20–22]. The idea is to reduce the accessibility of iron for microbes, and those specific processes may differ according to the localization and tissue specific preferences of different pathogens [23,24].

    • Prebiotics and iron bioavailability? Unveiling the hidden association - A review

      2021, Trends in Food Science and Technology
      Citation Excerpt :

      However, minor amounts of iron are continuously being lost from the body through the surface of skin, gastrointestinal and genitourinary tracts. These losses are though negligible and have been estimated to be less than 1 mg per day (Gerner, Nuccio, & Raffatellu, 2020). It should be considered that losses of iron through excessive bleeding such as chronic ulcers may be substantial.

    View all citing articles on Scopus
    View full text