Elsevier

Vaccine

Volume 26, Issue 36, 26 August 2008, Pages 4676-4685
Vaccine

c-di-GMP is an effective immunomodulator and vaccine adjuvant against pneumococcal infection

https://doi.org/10.1016/j.vaccine.2008.06.099Get rights and content

Abstract

Cyclic diguanylate (c-di-GMP) is a unique bacterial intracellular signaling molecule capable of stimulating enhanced protective innate immunity against various bacterial infections. The effects of intranasal pretreatment with c-di-GMP, or intraperitoneal coadministration of c-di-GMP with the pneumolysin toxoid (PdB) or pneumococcal surface protein A (PspA) before pneumococcal challenge, were investigated in mice. We found that c-di-GMP had no significant direct short-term effect on the growth rate of Streptococcus pneumoniae either in vitro or in vivo. However, intranasal pretreatment of mice with c-di-GMP resulted in a significant decrease in bacterial load in lungs and blood after serotypes 2 and 3 challenge, and a significant decrease in lung titers after serotype 4 challenge. Potential cellular mediators of these enhanced protective responses were identified in lungs and draining lymph nodes. Intraperitoneal coadministration of c-di-GMP with PdB or PspA before challenge resulted in significantly higher antigen-specific antibody titers and increased survival of mice, compared to that obtained with alum adjuvant. These findings demonstrate that local or systemic c-di-GMP administration stimulates innate and adaptive immunity against invasive pneumococcal disease. We propose that c-di-GMP can be used as an effective broad spectrum immunomodulator and vaccine adjuvant to prevent infectious diseases.

Introduction

Streptococcus pneumoniae is the leading cause of bacterial pneumonia, meningitis, and otitis media in the United States [1]. In spite of the availability of antimicrobials, the capsular polysaccharide (PS) vaccine, and the 7-valent protein–PS conjugate vaccine, pneumococcal disease continues to be responsible for high morbidity and mortality worldwide, especially in groups at high risk [2]. Consequently, global efforts are focused on exploring alternative pneumococcal vaccine strategies to address the shortcomings of existing formulations, without compromising efficacy. One of these approaches involves the development of vaccines based on pneumococcal proteins that contribute to pathogenesis and are common to all serotypes. To date, the most promising vaccine candidates are the pneumolysin toxoid (PdB), pneumococcal surface protein A (PspA), pneumococcal surface protein C (PspC, also referred to as choline binding protein A) and the 37-kDa metal-binding lipoprotein PsaA (reviewed by [2]).

We have shown that c-di-GMP (3′,5′-cyclic diguanylic acid or cyclic diguanylate or cGpGp) initially identified in the bacterium Acetobacter xylinum is an intracellular signaling molecule [3], [4], [5]. It is present in multiple bacterial species but not in eukaryotes [6], [7], [8], [9], [10], [11], and is now recognized to control many key functions in bacteria, including survival, adherence, colonization, and biofilm formation [8], [9], [12], [13], [14], [15]. Recent studies indicate that it might also modulate host cellular responses [14], resulting in an enhanced control of infection [16]. Consistent with this notion, we recently demonstrated that c-di-GMP increases MIG/CXCL9 (a chemoattractant for activated T cells) suggesting possible antitumor activity [17], and inhibits basal and growth factor-induced proliferation of human colon carcinoma cells [18].

Recently, c-di-GMP was shown to modulate the immune system to prevent and fight various lethal bacterial infections [17]. Intranasal (i.n.) or subcutaneous administration of c-di-GMP before intratracheal challenge with Klebsiella pneumoniae also resulted in significantly increased survival and reduction in bacterial counts in lung and blood [19]. The response was characterized by enhanced accumulation of neutrophils, αβ T cells, and activated NK and αβ T lymphocytes, associated with earlier and more vigorous expression of chemokines and type-1 cytokines [19]. Moreover, lung macrophages recovered from Klebsiella-infected mice pretreated with c-di-GMP expressed greater quantities of inducible nitric oxide synthase (iNOS) and nitric oxide ex vivo than those isolated from mice pretreated with control cGMP [19]. These findings demonstrate that c-di-GMP delivered locally or systemically stimulates protective innate immunity in the lung, decrease bacterial burden and enhances protective responses against infection.

In this study, we investigated the ability of c-di-GMP to enhance resistance against systemic pneumococcal infection, using established mouse models. We provide additional direct evidence that c-di-GMP is immunostimulatory, can protect against infection, and acts as an effective vaccine adjuvant against systemic disease.

Section snippets

Bacterial strains

The pneumococcal strains used in this study were D39, a virulent type 2 strain [20], strain T4, a type 4 encapsulated strain [21], and Xen10, a bioluminescent derivative of type 3 strain A66.1 [22] that has been engineered to express luciferase so infections can be followed using bioluminescent imaging (Xenogen Corp., Hopkinton, MA).

Mice

I.n. challenge studies with D39 and T4 were carried out using 6–8-week old female Balb/cByJ mice (Jackson Laboratory, Bar Harbor, ME) at St. Jude Children's

c-di-GMP has no direct effect on growth of c-di-GMP

In two independent experimental, we found no effect in either promoting or inhibiting growth of S. pneumoniae Xen10 was observed as a result of incubation with c-di-GMP (data not shown).

Pretreatment of S. pneumoniae with c-di-GMP has no direct effect on growth

In experiments to test the effect of pre-incubating S. pneumoniae with c-di-GMP, we found that in contrast to the reduced early control at 0–4 h seen in pretreated mice, mice given c-di-GMP at the time of challenge showed responses matching those of the control group. No significant differences were observed in

Discussion

In this study, we report that pretreatment of mice either i.n. or i.p. with c-di-GMP, but not control nucleotide (cGMP) or saline, can induce a significant protective immune response against i.n. or i.p. challenge with virulent pneumococci in various murine infection models. Our current findings are consistent with previous studies with Staphylococcus aureus and K. pneumoniae, as well as recent in vivo and in vitro studies showing that c-di-GMP is immunomodulatory and immunostimulatory [16],

Acknowledgments

The authors wish to acknowledge excellent technical support from Amy R. Iverson at St. Jude Children's Research Hospital. D.K.R.K. is a recipient of a Burroughs Wellcome Career Award in the Biomedical Sciences. This research was supported by the Army Research Office Grant to DKRK, National Health and Medical Research Council of Australia Program Grant 284214, and Welcome Trust Grant #075846 to ACK and Paul Kaye and MRC Grant #G981148.

Conflicts of interest: D.K.R. Karaolis has three related

References (50)

  • P. Ross et al.

    Regulation of cellulose synthesis in Acetobacter xylinum by cyclic diguanylic acid

    Nature

    (1987)
  • D.A. D’Argenio et al.

    Cyclic di-GMP as a bacterial second messenger

    Microbiology

    (2004)
  • H.A. Jones et al.

    HmsT, a protein essential for expression of the haemin storage (Hms+) phenotype of Yersinia pestis

    Microbiology

    (1999)
  • U. Römling et al.

    C-di-GMP: the dawning of a novel bacterial signalling system

    Mol Microbiol

    (2005)
  • D.A. D’Argenio et al.

    Autolysis and autoaggregation in Pseudomonas aeruginosa colony morphology mutants

    J Bacteriol

    (2002)
  • G.B. Hecht et al.

    Identification of a novel response regulator required for the swarmer-to-stalked-cell transition in Caulobacter crescentus

    J Bacteriol

    (1995)
  • D.K.R. Karaolis et al.

    c-di-GMP (3′–5′-cyclic diguanylic acid) inhibits Staphylococcus aureus cell–cell interactions and biofilm formation

    Antimicrob Agents Chemother

    (2005)
  • U. Römling et al.

    AgfD, the checkpoint of multicellular and aggregative behaviour in Salmonella typhimurium regulates at least two independent pathways

    Mol Microbiol

    (2000)
  • E. Brouillette et al.

    3′,5′-cyclic diguanylic acid reduces the virulence of biofilm-forming Staphylococcus aureus strains in a mouse model of mastitis infection

    Antimicrob Agents Chemother

    (2005)
  • D.K.R. Karaolis et al.

    Bacterial c-di-GMP is an immunostimulatory molecule

    J Immunol

    (2007)
  • D.K.R. Karaolis et al.

    Cyclic-di-GMP stimulates protective innate immunity in bacterial pneumonia

    Infect Immun

    (2007)
  • O.T. Avery et al.

    Studies on the chemical nature of the substance inducing transformation of pneumococcal types: induction of transformation by a desoxyribonucleic acid fraction isolated from pneumococcus type III

    J Exp Med

    (1944)
  • H. Tettelin et al.

    Complete genome sequence of a virulent isolate of Streptococcus pneumoniae

    Science

    (2001)
  • K.P. Francis et al.

    Visualizing pneumococcal infections in the lungs of live mice using bioluminescent Streptococcus pneumoniae transformed with a novel gram-positive lux transposing

    Infect Immun

    (2001)
  • M. Hyodo et al.

    Organic synthesis, chemical properties, and biological activities of cyclic bus(3′–5′) diguanylic acid (c-di-GMP) and its analogy

    J Synch Org Chem Jon

    (2006)
  • Cited by (74)

    View all citing articles on Scopus
    View full text