Journal List > Immune Netw > v.15(1) > 1033493

Fukuyama, Ikeda, Ohori, Sugita, Aso, Fujihashi, Briles, McGhee, and Fujihashi: A Molecular Mucosal Adjuvant To Enhance Immunity Against Pneumococcal Infection In The Elderly

Abstract

Streptococcus pneumoniae (the pneumococcus) causes a major upper respiratory tract infection often leading to severe illness and death in the elderly. Thus, it is important to induce safe and effective mucosal immunity against this pathogen in order to prevent pnuemocaccal infection. However, this is a very difficult task to elicit protective mucosal IgA antibody responses in older individuals. A combind nasal adjuvant consisting of a plasmid encoding the Flt3 ligand cDNA (pFL) and CpG oligonucleotide (CpG ODN) successfully enhanced S. pneumoniae-specific mucosal immunity in aged mice. In particular, a pneumococcal surface protein A-based nasal vaccine given with pFL and CpG ODN induced complete protection from S. pneumoniae infection. These results show that nasal delivery of a combined DNA adjuvant offers an attractive potential for protection against the pneumococcus in the elderly.

Abbreviations

CMIS

Common Mucosal Immune System

CpG ODN

CpG oligodeoxynucleotide

GCs

germinal centers

LP

Lamina propria

M cells

Microfold cells

NALT

Nasopharyngeal-associated lymphoid tissues

PPs

Peter's patches

pFL

Plasmid encoding Flt3 ligand cDNA

PPV

Pneumococcal polysaccharide vaccine

PspA

Pneumococcal surface protein A

pIgR

Polymeric Ig receptor

SIgA

Secretory IgA

UR

Upper respiratory

INTRODUCTION

In higher mammals, the mucosal immune system consists of an integrated network of tissues, lymphoid and mucous membrane-associated cells and effector antibody (Ab) molecules. Along with cytokines, chemokines and their receptors, these effector Ab molecules, which are primarily of the IgA isotype, are key players in mucosal immunity and appear to function in synergy with innate host factors (1,2). Thus, in order to induce antigen (Ag)-specific immune responses at these mucosal barriers, one must consider the common mucosal immune system (CMIS), which supports the concept of distinct mucosal IgA inductive and effector tissues (1,2). The Waldeyer's ring of tonsils and adenoids as nasopharyngeal-associated lymphoid tissues (NALT) serve as mucosal inductive sites. NALT is covered by a lymphoepithelium containing microfold (M) cells and well organized regions, the subepithelium with enriched Ag-presenting cells (APCs), a B cell zone with germinal centers (GCs), and adjacent T cell areas including an equal distribution of naïve and memory T cell phenotypes (1,2). Upon Ag-activation, memory B and T cell populations then emigrate from the mucosal inductive environment via lymphatic drainage, circulate through the bloodstream, and home to mucosal effector sites where abundant IgA producing plasma cells are present.
Effector sites for mucosal immune responses include the lymphoid cells in the lamina propria (LP) of the upper respiratory (UR), and reproductive tracts as well as secretory glandular tissues (1,2). Ag-specific mucosal effector cells include IgA-producing plasma cells as well as mature B and T lymphocytes. Secretory IgA (SIgA) is the primary Ig involved in protecting mucosal surfaces and is locally produced in effector tissues (1,2). In this regard, the majority of T and B cells in effector tissues are activated and express a memory phenotype (1,2). Although it has been shown that NALT shares common features with gastrointestinal (GI) inductive tissues such as Peyer's patches (PPs), their involution during immunosenescence, development during organogenesis and lymphocytes trafficking are distinctly regulated (3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20). Thus, the age-associated alteration including a reduction in PPs, intestinal Ag-specific SIgA Ab responses and lack of oral tolerance induction occur during the aging process (by 6~12 months in mice) (6,7,11,12). In contrast, NALT function remains intact during aging with notable signs of immunosenescence seen only in the elderly (2-year-old mice) (6,7,9).
In this short review, we summarize the potential for the development of a safe and effective nasal vaccine to prevent pneumococcal infection in the elderly.

Novel vaccines and delivery systems for the prevention of pnuemonoccal infection

In order to prevent pneumococcucal infection, nasal immunization strategies have been employed using novel vaccines and delivery systems. It has been shown that PspA plus IL-12 as nasal adjuvant enhanced PspA-specific IgG and IgA Ab responses with increased protection from nasal carriage (21). In this regard, the safety of nasal application of IL-12 was evaluated by using ex vivo human bronchoalveolar lavage (BAL) cells (22). These studies suggest that IL-12 could be used in clinical trials for pneumococcal protein-based vaccines although additional safety evaluations may be required. In addition to classical admixtures of adjuvants and pneumococcal Ags, PspA-fusion proteins have also been used as nasal vaccines. Nasal immunization with recombinant fusion proteins consisting of PspA and FlaB (flagellin of Vibrio vulnificus) resulted in cross-protective mucosal immunity (23). Others showed that mice vaccinated with human Fcγ receptor type I (Fcγ RI)-specific humanized single-chain Ab linked to PspA showed complement- and lactoferrin-mediated anti-pneumococcal innate immunity (24).
In more recent developments, nano-technology has been introduced in order to improve pneumococcal vaccines. Thus, nasal immunization with chitosan-DNA nanoparticles expressing pneumococcal surface antigen A (PsaA) elicited protective immunity against nasal colonization by S. pneumoniae (25). More recently, nanogels containing pneumococcal surface protein A (PspA) alone induced PspA-specific mucosal IgA Ab and Th17 responses which contributed to protection from S. pneumoniae infection in the lower respiratory tract (26). Further, Lactobacillus casei and Salmonella enterica serovar Typhimurium-derived outer membrane vesicles were employed as a nasal delivery system for pneumococcal surface protein Ags and successfully elicited protective immunity against challenge with S. pneumoniae (27,28). Similarly, a bacteria-based, nasal delivery approach was performed using live attenuated S. pneumoniae (29,30,31). Nasal delivery of S. pneumoniae which possessed a mutation in pep27, an autolysis-inducing factor, or deletion of ftsY, the single recognition pathway component, resulted in serotype-independent protection (30,31). Most recently, the live attenuated SPY1 vaccine strain induced both specific Ab and Th17 responses for protection form S. pneumoniae infection (29). Despite these advanced vaccine strategies which provide significant protection from pneumococcal infection, their effectiveness and safety have never been evaluated for the elderly.

Secretory IgA Abs play an important role in immunity to Streptococcus pneumoniae

The highest incidence of influenza and pneumococcal diseases occur in humans over 65 years of age. Thus, severe illness and mortality caused by pneumococci sharply increases in the elderly (32). The development of effective vaccines for the elderly remains a largely unmet goal. The effectiveness of the currently licensed pneumococcal polysaccharide vaccine (PPV) for adults consists of capsular polysaccharides derived from 23 serotypes (PPV23); however, its effectiveness becomes markedly reduced at ages above 75 years (33,34). It has been previously reported that older adults have a less effective Ab response to PPV23 than do their younger counterparts (35,36,37). In this regard, one should strongly consider developing a new generation of vaccines that could induce pneumococcal-specific IgA immunity in the respiratory tract since it has been shown that IgA plays a central role in immune defense, controlling the spread of respiratory pathogens including S. pneumoniae. Indeed, nasal immunization with PspA and cholera toxin as adjuvant elicited PspA-specific mucosal IgA and systemic IgG Ab responses and provided protection against carriage of S. pneumoniae and pneumococcal infection (38,39,40). Further, it was reported that S. pneumoniae capsular polysaccharide-specific polymeric IgA Abs more efficiently initiated complement-mediated killing of the organism (41). Others demonstrated that polymeric Ig receptor deficient (pIgR-/-) mice which lack the ability to actively secrete polymeric SIgA Abs onto mucosal surfaces, failed to provide protection against nasal challenge with S. pneumoniae despite being given a mucosal vaccine which induced SIgA Abs in normal mice (42,43,44).
Our studies provide more direct evidence that PspA-specific SIgA Abs play a key role in the prevention of bacterial colonization of the nasal mucosa using IgA deficient (IgA-/-) mice. Thus, although IgA-/- mice given nasal PspA plus plasmid encoding Flt3 ligand cDNA (pFL) revealed significantly higher levels of PspA-specific IgG Ab responses, and high numbers of S. pneumoniae CFUs were detected in the nasal mucosa. In contrast, vaccinated, wild type mice showed essentially no bacteria in the nasal cavity. Further, a nasal vaccine consisting of PspA plus pFL effectively reduced pre-existing S. pneumoniae in the nasal cavity (45). The importance of IL-17 synthesis for protection against pneumococcal infection (46,47,48). Although IgA-/- mice given nasal PspA plus pFL exhibited increased levels of IL-17 producing CD4+ T cells as did IgA+/+ mice, this mutant mouse strain failed to prevent colonization by S. pneumoniae. These results show that a PspA-based vaccine induced specific SIgA Abs plays a necessary role in the regulation of S. pneumoniae colonization in the nasal cavity.

Novel vaccine adjuvant development for use in the elderly

The results obtained in IgA-/- mice were very similar to the immune responses seen in aged mice which failed to elicit Ag-specific SIgA Ab responses in the mucosal lymphoid tissues despite significant IgG Ab induction (9). Thus, it is essential that we continue to explore new avenues for effective pneumococcal vaccine development for the elderly which include the induction of specific SIgA Ab responses. Adjuvant systems have provided significant improvement in the development of vaccines for another respiratory pathogen, i.e., the influenza virus vaccines for the elderly (49,50,51). Thus, a combination of alum and poly I:C as an adjuvant enhanced the effectiveness of an influenza virus-like particle vaccine in aged mice (51). Further, an H5N1 vaccine with MF59 adjuvant induced a rapid rise in broadly cross-reactive Abs as well as long-lived human memory B cells (49). More recently, the AS03 adjuvant system (Squalene, DL-α-tocopherol and polysorbate 80, GlaxoSmithKline) improved the immune response to inactivated 2009 H1N1 influenza vaccine in both healthy (18~64 years) and older adults (> 65 years) (50).
CpG oligodeoxynucleotide (CpG ODN) as vaccine adjuvant has been shown to restore Ag-specific immune responses to ovalbumin (OVA), diphtheria toxoid, hepatitis B, polysaccharide of S. pneumoniae, amyloid β and tumor cells in aged mice and rats (52,53,54,55,56,57,58). When 3 month old (young adult) and 18 month-old (moderately aged) mice were orally immunized with OVA plus CpG ODN as adjuvant, both groups of mice showed high and equivalent levels of OVA-specific systemic IgG and mucosal IgA Ab responses (59). Furthermore, we have shown that a combination of nasal pFL and CpG ODN selectively increased the frequencies of CD11c+ dendritic cells (DCs) in the nasal mucosa of young adult mice. Thus, absolute cell numbers of CD8- B220+ plasmacytoid DC (pDC) and CD8+ B220- non-pDC populations were preferentially increased by use of the combined adjuvant regimen (60). Although native cholera toxin (the most potent nasal adjuvant) failed to induce OVA-specific SIgA Ab responses in 2-year-old mice (9), nasal immunization with 100 µg of OVA and a combination of 50 µg of pFL and 10 µg of CpG ODN in aged mice resulted in significant SIgA Ab responses in external secretions, which were essentially equivalent to those observed in young adult mice (60). These results clearly indicate that a combination of pFL and CpG ODN can be used as a potent nasal adjuvant system for the induction of mucosal Ab responses in aged mice (60,61). In addition, it has been shown that the effect of aging on IL-2 production was abrogated by exogenous IL-2 delivery (62). Thus, mucosal IL-2 treatment reversed age-impaired mucosal immune responses by enhancing mucosal immunity or abrogating unresponsiveness in aged mice (63). Furthermore, recent studies showed that keratinocyte growth factor or IL-7 treatment prevented thymic atrophy (64,65). IL-15 treatment also restored impaired DC function in mesenteric lymph nodes of aged mice (66). Taken together, we hypothesize that mucosal delivery of DC-targeting pFL and CpG ODN as a mucosal adjuvant, would elicit Ag-specific SIgA Ab responses in the elderly against S. pneumonia infection.

A nasal combined adjuvant elicits protective siga immunity to S. pneumoniae in aged mice

Since a combination of pFL and CpG ODN was a potent nasal adjuvant in aged mice, we next examined whether this double adjuvant system could successfully induce bacterial antigen i.e., PspA-specific SIgA Ab responses in the UR tract mucosa for prevention of both S. pneumoniae carriage and infection. Nasal immunization with PspA plus a combination of pFL and CpG ODN elicited elevated levels of PspA-specific SIgA Ab responses in external secretions and plasma in both young adult and aged mice (61). Significant levels of PspA-specific CD4+ T cell proliferative and PspA-induced Th1- and Th2- but not Th17-type cytokine responses were noted in NALT and cervical lymph nodes of aged mice (61). In addition, increased numbers of mature-type CD8 or CD11b-expressing DCs were detected in mucosal tissues of aged mice (61). Importantly, aged mice given PspA plus a combination of pFL and CpG ODN showed protective immunity against nasal S. pneumoniae colonization (61). In contrast, both aged and young adult mice given nasal PspA alone fail to provide sufficient protection after nasal challenge. Thus, high numbers of S. pneumoniae CFUs were seen the NWs and NPs of both groups of mice. Further, aged mice given PspA plus pFL or CpG ODN revealed high numbers of bacterial CFUs in both NWs and NPs. The numbers of S. pneumoniae CFUs were essentially the same as those mice given PspA alone. (61). These results demonstrate that nasal delivery of a combined DNA adjuvant offers an attractive possibility for protection against S. pneumoniae in the elderly.

CLOSING REMARKS

Our focus has targeted the NALT for development of nasal vaccines containing a novel adjuvant combination which stimulates mucosal DCs in order to elicit pathogen-specific SIgA Ab responses in the elderly. Our studies have shown that vaccine/adjuvant-triggered NALT DCs from aged mice possess normal APC functions and thus fully support Ag-specific CD4+ T cell responses. In particular, nasal immunization with PspA given with pFL and CpG ODN elicited increased levels of Ag-specific SIgA Abs for subsequent protection from S. pneunomiae infection in aged mice. The elderly have already experienced numerous UR infections and have received prior vaccines. Thus, although they possess specific Abs, their titers are both too low and too clonally restricted to effectively combat reinfection. In the future, it would be of great benefit to the aged population if one could use pFL and CpG ODN alone, in order to immediately enhance pre-existing mucosal immunity which had developed in response to past respiratory infections and/or deliberate (annual) vaccination. Thus, subsequent nasal vaccine boosting could then more effectively expand pathogen-specific memory T and B cell responses to provide prolonged protection.

Abbreviations

CMIS

Common Mucosal Immune System

CpG ODN

CpG oligodeoxynucleotide

GCs

germinal centers

LP

Lamina propria

M cells

Microfold cells

NALT

Nasopharyngeal-associated lymphoid tissues

PPs

Peter's patches

pFL

Plasmid encoding Flt3 ligand cDNA

PPV

Pneumococcal polysaccharide vaccine

PspA

Pneumococcal surface protein A

pIgR

Polymeric Ig receptor

SIgA

Secretory IgA

UR

Upper respiratory

ACKNOWLEDGEMENTS

This work was supported by National Institutes of Health (NIH) grant AG025873.

Notes

The authors have no financial conflict of interest.

References

1. Fujihashi K, Boyaka PN, McGhee JR. Host defenses at mucosal surfaces. In : Rich RT, Fleisher TA, Shearer WT, Schroeder HW, Frew AJ, Weyand CM, editors. Clinical Immunology. Philadelphia: Elsevier;2013. p. 239–251.
2. Kiyono H, Kunisawa J, McGhee JR, Mestecky J. The Mucosal Immune System. In : Paul WE, editor. Fundamental Immunology. Philadelphia: Lippincott Williams & Wilkins;2008. p. 983–1030.
3. Campbell DJ, Butcher EC. Rapid acquisition of tissue-specific homing phenotypes by CD4+ T cells activated in cutaneous or mucosal lymphoid tissues. J Exp Med. 2002; 195:135–141.
crossref
4. Campbell JJ, Brightling CE, Symon FA, Qin S, Murphy KE, Hodge M, Andrew DP, Wu L, Butcher EC, Wardlaw AJ. Expression of chemokine receptors by lung T cells from normal and asthmatic subjects. J Immunol. 2001; 166:2842–2848.
crossref
5. Csencsits KL, Walters N, Pascual DW. Cutting edge: dichotomy of homing receptor dependence by mucosal effector B cells: α(E) versus L-selectin. J Immunol. 2001; 167:2441–2445.
crossref
6. Fujihashi K, Kiyono H. Mucosal immunosenescence: new developments and vaccines to control infectious diseases. Trends Immunol. 2009; 30:334–343.
crossref
7. Fujihashi K, McGhee JR. Mucosal immunity and tolerance in the elderly. Mech Ageing Dev. 2004; 125:889–898.
crossref
8. Fukuyama S, Hiroi T, Yokota Y, Rennert PD, Yanagita M, Kinoshita N, Terawaki S, Shikina T, Yamamoto M, Kurono Y, Kiyono H. Initiation of NALT organogenesis is independent of the IL-7R, LTβR, and NIK signaling pathways but requires the Id2 gene and CD3-CD4+CD45+ cells. Immunity. 2002; 17:31–40.
crossref
9. Hagiwara Y, McGhee JR, Fujihashi K, Kobayashi R, Yoshino N, Kataoka K, Etani Y, Kweon MN, Tamura S, Kurata T, Takeda Y, Kiyono H, Fujihashi K. Protective mucosal immunity in aging is associated with functional CD4+ T cells in nasopharyngeal-associated lymphoreticular tissue. J Immunol. 2003; 170:1754–1762.
crossref
10. Kantele A, Zivny J, Hakkinen M, Elson CO, Mestecky J. Differential homing commitments of antigen-specific T cells after oral or parenteral immunization in humans. J Immunol. 1999; 162:5173–5177.
11. Kato H, Fujihashi K, Kato R, Dohi T, Fujihashi K, Hagiwara Y, Kataoka K, Kobayashi R, McGhee JR. Lack of oral tolerance in aging is due to sequential loss of Peyer's patch cell interactions. Int Immunol. 2003; 15:145–158.
crossref
12. Koga T, McGhee JR, Kato H, Kato R, Kiyono H, Fujihashi K. Evidence for early aging in the mucosal immune system. J Immunol. 2000; 165:5352–5359.
crossref
13. Kunisawa J, Nochi T, Kiyono H. Immunological commonalities and distinctions between airway and digestive immunity. Trends Immunol. 2008; 29:505–513.
crossref
14. Macpherson AJ, McCoy KD, Johansen FE, Brandtzaeg P. The immune geography of IgA induction and function. Mucosal Immunol. 2008; 1:11–22.
crossref
15. Pascual DW, Riccardi C, Csencsits-Smith K. Distal IgA immunity can be sustained by αEβ7+ B cells in L-selectin-/- mice following oral immunization. Mucosal Immunol. 2008; 1:68–77.
crossref
16. Rennert PD, Browning JL, Mebius R, Mackay F, Hochman PS. Surface lymphotoxin α/β complex is required for the development of peripheral lymphoid organs. J Exp Med. 1996; 184:1999–2006.
crossref
17. Yoshida H, Honda K, Shinkura R, Adachi S, Nishikawa S, Maki K, Ikuta K, Nishikawa SI. IL-7 receptor alpha+ CD3(-) cells in the embryonic intestine induces the organizing center of Peyer's patches. Int Immunol. 1999; 11:643–655.
crossref
18. Kurebayashi S, Ueda E, Sakaue M, Patel DD, Medvedev A, Zhang F, Jetten AM. Retinoid-related orphan receptor γ (RORγ) is essential for lymphoid organogenesis and controls apoptosis during thymopoiesis. Proc Natl Acad Sci U S A. 2000; 97:10132–10137.
crossref
19. Sun Z, Unutmaz D, Zou YR, Sunshine MJ, Pierani A, Brenner-Morton S, Mebius RE, Littman DR. Requirement for RORγ in thymocyte survival and lymphoid organ development. Science. 2000; 288:2369–2373.
crossref
20. Yokota Y, Mansouri A, Mori S, Sugawara S, Adachi S, Nishikawa S, Gruss P. Development of peripheral lymphoid organs and natural killer cells depends on the helix-loop-helix inhibitor Id2. Nature. 1999; 397:702–706.
crossref
21. Arulanandam BP, Lynch JM, Briles DE, Hollingshead S, Metzger DW. Intranasal vaccination with pneumococcal surface protein A and interleukin-12 augments antibody-mediated opsonization and protective immunity against Streptococcus pneumoniae infection. Infect Immun. 2001; 69:6718–6724.
crossref
22. Wright AK, Christopoulou I, El BS, Limer J, Gordon SB. rhIL-12 as adjuvant augments lung cell cytokine responses to pneumococcal whole cell antigen. Immunobiology. 2011; 216:1143–1147.
crossref
23. Nguyen CT, Kim SY, Kim MS, Lee SE, Rhee JH. Intranasal immunization with recombinant PspA fused with a flagellin enhances cross-protective immunity against Streptococcus pneumoniae infection in mice. Vaccine. 2011; 29:5731–5739.
crossref
24. Bitsaktsis C, Iglesias BV, Li Y, Colino J, Snapper CM, Hollingshead SK, Pham G, Gosselin DR, Gosselin EJ. Mucosal immunization with an unadjuvanted vaccine that targets Streptococcus pneumoniae PspA to human Fcγ receptor type I protects against pneumococcal infection through complement- and lactoferrin-mediated bactericidal activity. Infect Immun. 2012; 80:1166–1180.
crossref
25. Xu J, Dai W, Wang Z, Chen B, Li Z, Fan X. Intranasal vaccination with chitosan-DNA nanoparticles expressing pneumococcal surface antigen a protects mice against nasopharyngeal colonization by Streptococcus pneumoniae. Clin Vaccine Immunol. 2011; 18:75–81.
crossref
26. Kong IG, Sato A, Yuki Y, Nochi T, Takahashi H, Sawada S, Mejima M, Kurokawa S, Okada K, Sato S, Briles DE, Kunisawa J, Inoue Y, Yamamoto M, Akiyoshi K, Kiyono H. Nanogel-based PspA intranasal vaccine prevents invasive disease and nasal colonization by Streptococcus pneumoniae. Infect Immun. 2013; 81:1625–1634.
crossref
27. Hernani Mde L, Ferreira PC, Ferreira DM, Miyaji EN, Ho PL, Oliveira ML. Nasal immunization of mice with Lactobacillus casei expressing the pneumococcal surface protein C primes the immune system and decreases pneumococcal nasopharyngeal colonization in mice. FEMS Immunol Med Microbiol. 2011; 62:263–272.
crossref
28. Muralinath M, Kuehn MJ, Roland KL, Curtiss R III. Immunization with Salmonella enterica serovar Typhimurium-derived outer membrane vesicles delivering the pneumococcal protein PspA confers protection against challenge with Streptococcus pneumoniae. Infect Immun. 2011; 79:887–894.
crossref
29. Xu X, Wang H, Liu Y, Wang Y, Zeng L, Wu K, Wang J, Ma F, Xu W, Yin Y, Zhang X. Mucosal immunization with the live attenuated vaccine SPY1 induces humoral and Th2-Th17-regulatory T cell cellular immunity and protects against pneumococcal infection. Infect Immun. 2015; 83:90–100.
crossref
30. Rosch JW, Iverson AR, Humann J, Mann B, Gao G, Vogel P, Mina M, Murrah KA, Perez AC, Edward SW, Tuomanen EI, McCullers JA. A live-attenuated pneumococcal vaccine elicits CD4+ T-cell dependent class switching and provides serotype independent protection against acute otitis media. EMBO Mol Med. 2014; 6:141–154.
crossref
31. Kim EH, Choi SY, Kwon MK, Tran TD, Park SS, Lee KJ, Bae SM, Briles DE, Rhee DK. Streptococcus pneumoniae pep27 mutant as a live vaccine for serotype-independent protection in mice. Vaccine. 2012; 30:2008–2019.
crossref
32. Janssens JP, Krause KH. Pneumonia in the very old. Lancet Infect Dis. 2004; 4:112–124.
crossref
33. Schenkein JG, Nahm MH, Dransfield MT. Pneumococcal vaccination for patients with COPD: current practice and future directions. Chest. 2008; 133:767–774.
34. Shapiro ED, Berg AT, Austrian R, Schroeder D, Parcells V, Margolis A, Adair RK, Clemens JD. The protective efficacy of polyvalent pneumococcal polysaccharide vaccine. N Engl J Med. 1991; 325:1453–1460.
crossref
35. Romero-Steiner S, Musher DM, Cetron MS, Pais LB, Groover JE, Fiore AE, Plikaytis BD, Carlone GM. Reduction in functional antibody activity against Streptococcus pneumoniae in vaccinated elderly individuals highly correlates with decreased IgG antibody avidity. Clin Infect Dis. 1999; 29:281–288.
crossref
36. Rubins JB, Alter M, Loch J, Janoff EN. Determination of antibody responses of elderly adults to all 23 capsular polysaccharides after pneumococcal vaccination. Infect Immun. 1999; 67:5979–5984.
crossref
37. Rubins JB, Puri AK, Loch J, Charboneau D, MacDonald R, Opstad N, Janoff EN. Magnitude, duration, quality, and function of pneumococcal vaccine responses in elderly adults. J Infect Dis. 1998; 178:431–440.
crossref
38. Briles DE, Hollingshead SK, King J, Swift A, Braun PA, Park MK, Ferguson LM, Nahm MH, Nabors GS. Immunization of humans with recombinant pneumococcal surface protein A (rPspA) elicits antibodies that passively protect mice from fatal infection with Streptococcus pneumoniae bearing heterologous PspA. J Infect Dis. 2000; 182:1694–1701.
crossref
39. Wu HY, Nahm MH, Guo Y, Russell MW, Briles DE. Intranasal immunization of mice with PspA (pneumococcal surface protein A) can prevent intranasal carriage, pulmonary infection, and sepsis with Streptococcus pneumoniae. J Infect Dis. 1997; 175:839–846.
crossref
40. Yamamoto M, Briles DE, Yamamoto S, Ohmura M, Kiyono H, McGhee JR. A nontoxic adjuvant for mucosal immunity to pneumococcal surface protein A. J Immunol. 1998; 161:4115–4121.
41. Janoff EN, Fasching C, Orenstein JM, Rubins JB, Opstad NL, Dalmasso AP. Killing of Streptococcus pneumoniae by capsular polysaccharide-specific polymeric IgA, complement, and phagocytes. J Clin Invest. 1999; 104:1139–1147.
crossref
42. Ferreira DM, Darrieux M, Silva DA, Leite LC, Ferreira JM Jr, Ho PL, Miyaji EN, Oliveira ML. Characterization of protective mucosal and systemic immune responses elicited by pneumococcal surface protein PspA and PspC nasal vaccines against a respiratory pneumococcal challenge in mice. Clin Vaccine Immunol. 2009; 16:636–645.
crossref
43. Park SM, Ko HJ, Shim DH, Yang JY, Park YH, Curtiss R III, Kweon MN. MyD88 signaling is not essential for induction of antigen-specific B cell responses but is indispensable for protection against Streptococcus pneumoniae infection following oral vaccination with attenuated Salmonella expressing PspA antigen. J Immunol. 2008; 181:6447–6455.
crossref
44. Sun K, Johansen FE, Eckmann L, Metzger DW. An important role for polymeric Ig receptor-mediated transport of IgA in protection against Streptococcus pneumoniae nasopharyngeal carriage. J Immunol. 2004; 173:4576–4581.
crossref
45. Fukuyama Y, King JD, Kataoka K, Kobayashi R, Gilbert RS, Oishi K, Hollingshead SK, Briles DE, Fujihashi K. Secretory-IgA antibodies play an important role in the immunity to Streptococcus pneumoniae. J Immunol. 2010; 185:1755–1762.
crossref
46. Lu YJ, Gross J, Bogaert D, Finn A, Bagrade L, Zhang Q, Kolls JK, Srivastava A, Lundgren A, Forte S, Thompson CM, Harney KF, Anderson PW, Lipsitch M, Malley R. Interleukin-17A mediates acquired immunity to pneumococcal colonization. PLoS Pathog. 2008; 4:e1000159.
crossref
47. Zhang Z, Clarke TB, Weiser JN. Cellular effectors mediating Th17-dependent clearance of pneumococcal colonization in mice. J Clin Invest. 2009; 119:1899–1909.
crossref
48. Zygmunt BM, Rharbaoui F, Groebe L, Guzman CA. Intranasal immunization promotes th17 immune responses. J Immunol. 2009; 183:6933–6938.
crossref
49. Galli G, Hancock K, Hoschler K, DeVos J, Praus M, Bardelli M, Malzone C, Castellino F, Gentile C, McNally T, Del GG, Banzhoff A, Brauer V, Montomoli E, Zambon M, Katz J, Nicholson K, Stephenson I. Fast rise of broadly cross-reactive antibodies after boosting long-lived human memory B cells primed by an MF59 adjuvanted prepandemic vaccine. Proc Natl Acad Sci U S A. 2009; 106:7962–7967.
crossref
50. Jackson LA, Chen WH, Stapleton JT, Dekker CL, Wald A, Brady RC, Edupuganti S, Winokur P, Mulligan MJ, Keyserling HL, Kotloff KL, Rouphael N, Noah DL, Hill H, Wolff MC. Immunogenicity and safety of varying dosages of a monovalent 2009 H1N1 influenza vaccine given with and without AS03 adjuvant system in healthy adults and older persons. J Infect Dis. 2012; 206:811–820.
crossref
51. Schneider-Ohrum K, Giles BM, Weirback HK, Williams BL, DeAlmeida DR, Ross TM. Adjuvants that stimulate TLR3 or NLPR3 pathways enhance the efficiency of influenza virus-like particle vaccines in aged mice. Vaccine. 2011; 29:9081–9092.
crossref
52. Maletto B, Ropolo A, Moron V, Pistoresi-Palencia MC. CpG-DNA stimulates cellular and humoral immunity and promotes Th1 differentiation in aged BALB/c mice. J Leukoc Biol. 2002; 72:447–454.
53. Maletto BA, Ropolo AS, Liscovsky MV, Alignani DO, Glocker M, Pistoresi-Palencia MC. CpG oligodeoxinucleotides functions as an effective adjuvant in aged BALB/c mice. Clin Immunol. 2005; 117:251–261.
crossref
54. Manning BM, Enioutina EY, Visic DM, Knudson AD, Daynes RA. CpG DNA functions as an effective adjuvant for the induction of immune responses in aged mice. Exp Gerontol. 2001; 37:107–126.
crossref
55. Qin W, Jiang J, Chen Q, Yang N, Wang Y, Wei X, Ou R. CpG ODN enhances immunization effects of hepatitis B vaccine in aged mice. Cell Mol Immunol. 2004; 1:148–152.
56. Sen G, Chen Q, Snapper CM. Immunization of aged mice with a pneumococcal conjugate vaccine combined with an unmethylated CpG-containing oligodeoxynucleotide restores defective immunoglobulin G antipolysaccharide responses and specific CD4+-T-cell priming to young adult levels. Infect Immun. 2006; 74:2177–2186.
crossref
57. Sharma S, Dominguez AL, Hoelzinger DB, Lustgarten J. CpG-ODN but not other TLR-ligands restore the antitumor responses in old mice: the implications for vaccinations in the aged. Cancer Immunol Immunother. 2008; 57:549–561.
crossref
58. Subramanian S, vya Shree AN. Enhanced Th2 immunity after DNA prime-protein boost immunization with amyloid beta (1-42) plus CpG oligodeoxynucleotides in aged rats. Neurosci Lett. 2008; 436:219–222.
crossref
59. Alignani D, Maletto B, Liscovsky M, Ropolo A, Moron G, Pistoresi-Palencia MC. Orally administered OVA/CpG-ODN induces specific mucosal and systemic immune response in young and aged mice. J Leukoc Biol. 2005; 77:898–905.
crossref
60. Fukuiwa T, Sekine S, Kobayashi R, Suzuki H, Kataoka K, Gilbert RS, Kurono Y, Boyaka PN, Krieg AM, McGhee JR, Fujihashi K. A combination of Flt3 ligand cDNA and CpG ODN as nasal adjuvant elicits NALT dendritic cells for prolonged mucosal immunity. Vaccine. 2008; 26:4849–4859.
crossref
61. Fukuyama Y, King JD, Kataoka K, Kobayashi R, Gilbert RS, Hollingshead SK, Briles DE, Fujihashi K. A combination of Flt3 ligand cDNA and CpG oligodeoxynucleotide as nasal adjuvant elicits protective secretory-IgA immunity to Streptococcus pneumoniae in aged mice. J Immunol. 2011; 186:2454–2461.
crossref
62. Haynes L, Linton PJ, Eaton SM, Tonkonogy SL, Swain SL. Interleukin 2, but not other common gamma chain-binding cytokines, can reverse the defect in generation of CD4 effector T cells from naive T cells of aged mice. J Exp Med. 1999; 190:1013–1024.
crossref
63. Fayad R, Zhang H, Quinn D, Huang Y, Qiao L. Oral administration with papillomavirus pseudovirus encoding IL-2 fully restores mucosal and systemic immune responses to vaccinations in aged mice. J Immunol. 2004; 173:2692–2698.
crossref
64. Henson SM, Pido-Lopez J, Aspinall R. Reversal of thymic atrophy. Exp Gerontol. 2004; 39:673–678.
crossref
65. Min D, Panoskaltsis-Mortari A, Kuro O, Hollander GA, Blazar BR, Weinberg KI. Sustained thymopoiesis and improvement in functional immunity induced by exogenous KGF administration in murine models of aging. Blood. 2007; 109:2529–2537.
crossref
66. Moretto MM, Lawlor EM, Khan IA. Aging mice exhibit a functional defect in mucosal dendritic cell response against an intracellular pathogen. J Immunol. 2008; 181:7977–7984.
crossref
TOOLS
Similar articles