Top Banner
All rights reserved This work by Wilolud Journals is li Continental J. Medical Re © Wilolud Journals, 2013 Printed in Nigeria Streptococcus pn Ukpai A. 1 Department of Medical Laboratory Sc Nigeria. 2 Department of M ABSTRACT Streptococcus pneumoniae is one colonises the nasopharyngeal cel disease condition is often preceded children less than 2 years, the elde may disseminate and cause life- bacteraemia/sepsis and meningitis. of age die annually in develo pneumococcal infections has been shows that global prevalence of pe varies between 18.2% to 22.1% and the use of pneumococcal polysacch vaccines are antibody dependent a elicit immunologic memory. In con as they only provide protection aga capsular switch in types not incorp understanding of the structure and way for the possibility of developin factors and the potential targets for KEY WORDS: Streptococcus pn protein-based pneumococcal vaccin Received for Publication: 15/03/13 Corresponding author:doyeneze@g INTRODUCTION Streptococcus pneumoniae is colonise individuals. The adherence and colon interactions between choline binding p and this interaction also facilitates the addition, pneumococcal surface pro apolactoferrin-mediated killing of Strep et al., 2004). This interaction is shown i The development of disease conditio individuals such as children less than pneumococci may disseminate and cau bacteraemia/sepsis and meningitis (Mc Kim et al., 2012). Report shows that a developing countries from pneumococc Treatment of pneumococcal infections studies shows that global prevalence of icensed under a Creative Commons Attribution 3.0 Unpor 27 esearch 7 (1): 27 -45, 2013 ISSN: 3 http://www.wilo doi:10.5707/cjmedres. neumoniae: VIRULENCE FACTORS AND THEIR RO PATHOGENESIS Eze 1 , Adam Mustapha 2 and Amos Nworie 1 ciences, College of Health Sciences, Ebonyi State Univer Microbiology, University of Maiduguri, Borno State, Niger e of the major bacteria cause of respiratory tract inf lls of asymptomatically healthy individuals. The deve d by nasopharyngeal colonisation. In susceptible individu erly above 65 years, and the immunocompromised, the p -threatening disease, including causing otitis media, Report shows that approximately 1 million children less oping countries from pneumococcal disease. The tr complicated by rising trend of antibiotic resistance. Sev enicillin-resistant and macrolide-resistant Streptococcus p nd 24.6% to 31.8% respectively. Current prevention strate haride vaccines and conjugates vaccines. The capsular pol and are poorly immunogenic in the target groups and al ontrast, the conjugate vaccines have shown high efficacy, ainst the capsular serotypes/groups present in the vaccine porated in the conjugate vaccines. However, recent adva d function of various pathogenicity and virulence factors ng protein vaccines. This review focuses on pneumococc protein-based pneumococcal vaccine production. neumoniae, Capsular polysaccharide vaccines, conjugat nes, virulence factors. 3 Accepted for Publication gmail.com es the nasopharyngeal cells and is carried asymptomatic nization of the nasopharyngeal cells is thought to be m protein A (CbpA) and human polymeric immunoglobin r e invasion of nasopharyngeal epithelial cells (Rosenow otein A (PspA) interacts with apolactoferrin interfe ptococcus pneumoniae in the nasopharynx and promotes c in figure 1.2. on is often preceded by nasopharyngeal colonisation. 2 years, the elderly above 65 years, and the immunoco use life-threatening disease, including causing otitis me cCullers and Tuomanen, 2001; Iannelli et al., 2004; Tuf approximately 1 million children less than 5 years of age cal disease (Swiatto et al., 2003). s has been complicated by rising trend of antibiotic res f penicillin-resistant and macrolide-resistant Streptococcus rted License : 2141 – 4211 oludjournal.com .2013.7.1.27.45 OLE IN sity, Abakaliki, ria. fections and elopment of uals such as pneumococci pneumonia, than 5 years reatment of veral studies pneumoniae egies include lysaccharide lso does not , but limited e resulting to ances in the have paved cal virulence te vaccines, n: 07/05/13 cally by healthy mediated by the receptor (hpIgR) et al., 1997). In ering with the carriage (Shaper . In susceptible ompromised, the edia, pneumonia, fari et al., 2011; e die annually in sistance. Several s pneumoniae
19

STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

Jan 12, 2023

Download

Documents

Ukpai Eze
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Continental J. Medical Research 7 (1): © Wilolud Journals, 2013 Printed in Nigeria

Streptococcus pneumoniae

Ukpai A. Eze1Department of Medical Laboratory Sciences, College of Health Sciences, Ebonyi State University, Abakaliki,

Nigeria.2Department of Microbiology, University of Maiduguri, Borno State, Nigeria.

ABSTRACT Streptococcus pneumoniae is one of the major bacteria colonises the nasopharyngeal cells of asymptomatically healthy individuals. The development of disease condition is often preceded by nasopharyngeal colonisation. In susceptible individuals such as children less than 2 years, the elderly above 65 years, and the immunocompromised, the pneumococci may disseminate and cause life-bacteraemia/sepsis and meningitis. Report shows that approximately 1 million cof age die annually in developing countries from pneumococcal disease. The treatment of pneumococcal infections has been complicated by rising trend of antibiotic resistance. Several studies shows that global prevalence of penicilvaries between 18.2% to 22.1% and 24.6% to 31.8% respectively. Current prevention strategies include the use of pneumococcal polysaccharide vaccines and conjugates vaccines. The capsular polysavaccines are antibody dependent and are poorly immunogenic in the target groups and also does not elicit immunologic memory. In contrast, the conjugate vaccines have shown high efficacy, but limited as they only provide protection against the capscapsular switch in types not incorporated in the conjugate vaccines. understanding of the structure and function of various pathogenicity and virulence factors have pway for the possibility of developing protein vaccines. This review focuses on pneumococcal virulence factors and the potential targets for KEY WORDS: Streptococcus pneumoniaeprotein-based pneumococcal vaccines, virulence factors. Received for Publication: 15/03/13Corresponding author:[email protected]

INTRODUCTION Streptococcus pneumoniae is colonises the nasopharyngeal cells and is carried asymptomatically by healthy individuals. The adherence and colonization of the nasopharyngeal cells is thought to be mediated by the interactions between choline binding protein A (CbpA) and human polymand this interaction also facilitates the invasion of nasopharyngeal epithelial cells (Rosenow addition, pneumococcal surface protein A (PspA) interacts with apolactoferrin interfering with the apolactoferrin-mediated killing of Streptococcus pneumoniaeet al., 2004). This interaction is shown in figure 1.2. The development of disease condition is often preceded by nasopharyngeal colonisation. In susceptible individuals such as children less than 2 years, the elderly above 65 years, and the immunocompromised, the pneumococci may disseminate and cause lifebacteraemia/sepsis and meningitis (McCullers and Tuomanen, 2001; Iannelli Kim et al., 2012). Report shows that approximately 1 million children less than 5 yearsdeveloping countries from pneumococcal disease (Swiatto Treatment of pneumococcal infections has been complicated by rising trend of antibiotic resistance. Several studies shows that global prevalence of penicillin

is licensed under a Creative Commons Attribution 3.0 Unported License

27

Continental J. Medical Research 7 (1): 27 -45, 2013 ISSN: 2141 © Wilolud Journals, 2013 http://www.wiloludjournal.com

doi:10.5707/cjmedres.2013.7.1.

Streptococcus pneumoniae: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS

Ukpai A. Eze1, Adam Mustapha2 and Amos Nworie1

Department of Medical Laboratory Sciences, College of Health Sciences, Ebonyi State University, Abakaliki, Department of Microbiology, University of Maiduguri, Borno State, Nigeria.

is one of the major bacteria cause of respiratory tract infections and colonises the nasopharyngeal cells of asymptomatically healthy individuals. The development of disease condition is often preceded by nasopharyngeal colonisation. In susceptible individuals such as

an 2 years, the elderly above 65 years, and the immunocompromised, the pneumococci -threatening disease, including causing otitis media, pneumonia,

bacteraemia/sepsis and meningitis. Report shows that approximately 1 million children less than 5 years of age die annually in developing countries from pneumococcal disease. The treatment of pneumococcal infections has been complicated by rising trend of antibiotic resistance. Several studies shows that global prevalence of penicillin-resistant and macrolide-resistant Streptococcus pneumoniaevaries between 18.2% to 22.1% and 24.6% to 31.8% respectively. Current prevention strategies include the use of pneumococcal polysaccharide vaccines and conjugates vaccines. The capsular polysavaccines are antibody dependent and are poorly immunogenic in the target groups and also does not elicit immunologic memory. In contrast, the conjugate vaccines have shown high efficacy, but limited as they only provide protection against the capsular serotypes/groups present in the vaccine resulting to capsular switch in types not incorporated in the conjugate vaccines. However, recent advances in the understanding of the structure and function of various pathogenicity and virulence factors have pway for the possibility of developing protein vaccines. This review focuses on pneumococcal virulence

and the potential targets for protein-based pneumococcal vaccine production.

Streptococcus pneumoniae, Capsular polysaccharide vaccines, conjugate vaccines, based pneumococcal vaccines, virulence factors.

3 Accepted for Publication: [email protected]

is colonises the nasopharyngeal cells and is carried asymptomatically by healthy individuals. The adherence and colonization of the nasopharyngeal cells is thought to be mediated by the interactions between choline binding protein A (CbpA) and human polymeric immunoglobin receptor (hpIgR) and this interaction also facilitates the invasion of nasopharyngeal epithelial cells (Rosenow addition, pneumococcal surface protein A (PspA) interacts with apolactoferrin interfering with the

Streptococcus pneumoniae in the nasopharynx and promotes carriage (Shaper ., 2004). This interaction is shown in figure 1.2.

The development of disease condition is often preceded by nasopharyngeal colonisation. In susceptible individuals such as children less than 2 years, the elderly above 65 years, and the immunocompromised, the pneumococci may disseminate and cause life-threatening disease, including causing otitis media, pneumonia, bacteraemia/sepsis and meningitis (McCullers and Tuomanen, 2001; Iannelli et al., 2004; Tufari

., 2012). Report shows that approximately 1 million children less than 5 years of age die annually in developing countries from pneumococcal disease (Swiatto et al., 2003).

Treatment of pneumococcal infections has been complicated by rising trend of antibiotic resistance. Several studies shows that global prevalence of penicillin-resistant and macrolide-resistant Streptococcus pneumoniae

Creative Commons Attribution 3.0 Unported License

ISSN: 2141 – 4211 http://www.wiloludjournal.com

.2013.7.1.27.45

VIRULENCE FACTORS AND THEIR ROLE IN

Department of Medical Laboratory Sciences, College of Health Sciences, Ebonyi State University, Abakaliki, Department of Microbiology, University of Maiduguri, Borno State, Nigeria.

cause of respiratory tract infections and colonises the nasopharyngeal cells of asymptomatically healthy individuals. The development of disease condition is often preceded by nasopharyngeal colonisation. In susceptible individuals such as

an 2 years, the elderly above 65 years, and the immunocompromised, the pneumococci threatening disease, including causing otitis media, pneumonia,

hildren less than 5 years of age die annually in developing countries from pneumococcal disease. The treatment of pneumococcal infections has been complicated by rising trend of antibiotic resistance. Several studies

Streptococcus pneumoniae varies between 18.2% to 22.1% and 24.6% to 31.8% respectively. Current prevention strategies include the use of pneumococcal polysaccharide vaccines and conjugates vaccines. The capsular polysaccharide vaccines are antibody dependent and are poorly immunogenic in the target groups and also does not elicit immunologic memory. In contrast, the conjugate vaccines have shown high efficacy, but limited

ular serotypes/groups present in the vaccine resulting to However, recent advances in the

understanding of the structure and function of various pathogenicity and virulence factors have paved way for the possibility of developing protein vaccines. This review focuses on pneumococcal virulence

accines, conjugate vaccines,

Accepted for Publication: 07/05/13

is colonises the nasopharyngeal cells and is carried asymptomatically by healthy individuals. The adherence and colonization of the nasopharyngeal cells is thought to be mediated by the

eric immunoglobin receptor (hpIgR) and this interaction also facilitates the invasion of nasopharyngeal epithelial cells (Rosenow et al., 1997). In addition, pneumococcal surface protein A (PspA) interacts with apolactoferrin interfering with the

in the nasopharynx and promotes carriage (Shaper

The development of disease condition is often preceded by nasopharyngeal colonisation. In susceptible individuals such as children less than 2 years, the elderly above 65 years, and the immunocompromised, the

atening disease, including causing otitis media, pneumonia, ., 2004; Tufari et al., 2011;

of age die annually in

Treatment of pneumococcal infections has been complicated by rising trend of antibiotic resistance. Several Streptococcus pneumoniae

Page 2: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: varies between 18.2% to 22.1% and 24.6% to 31.8% respectively (Felmingham, 2002; Jacoobs most common serotypes resistant to penicillin and macrolides include 6A, 6B, 9V, 14, 19A, 19F and 23F (Whitney et al., 2000). Current prevention strategies include pneumococcal 23which covers the serotypes implicated in approximately 85% of disease in the US and Europe, 62.9% in Taiwan, 72.9% in Japan, 58% in Brazil and 79.9 % in India (Lee addition, a 7-valent, 9-valent, 10-valent and 13been introduced (Tafuri et al., 2011; Kim dependent and are poorly immunogenic in the target groups; young children, elderly people and immunocompromised individuals, and also does not elicit immunologic memory (Briles Vidyashankara et al., 2012). The conjugate vaccines have shown high efficacy, but limited as protection against the capsular serotypes/groups present in the vaccine resulting to capsular switch in types not incorporated in the conjugate vaccines (Kim Therefore, to overcome the increasing frequency of antibioticon the understanding of the mechanisms of pneumococcal virulence factors and development ofvaccines is needed. This review focuses on the various surface components implicated in nasopharyngeal colonisation and adherence, including PspA, CbpA/PspC, Pneumococcal adherencepneumolysin and neuraminidase, and the potential targets for Pneumococcal serotype distribution and clinicalAsymptomatic pneumococcal colonisation of the nasopharynx is an important factor for disease initiation and provides the niche from where the bacteria can progress to the alveolar space, blood stream, middle ear or brain and cause pneumonia, septicaemia, otitis media and meningitis, respectively (MeCullers and Tuomanen, 2001). Pneumococcal carriage is more common in children compared to adults, reaching a prevalence of 55% at the age of three years (Bogaert et al., 2004). Some underlying medical codisease, including congenital immunodeficiency, Human Immunodeficiency Virus (HIV), leukaemia, lymphoma, sickle cell disease and tissue transplantation (CDC, 1997). Other predisposing conditions to pneumococcal disease include viral respiratory disease, chronic bronchitis, chronic obstructive pulmonary disease and smoking. The various clinical disease of In a review, Bogaert et al., (2004) reported that serotypesfound in nasopharyngeal colonisation. However, Syrjanen and colleagues reported that serotypes 6A, 6B, 23F, 19F, 11 and 14 were common isolates from nasopharyngeal samples from Finland (Syrjanen Netherlands, the common serotypes include 19F, 6B, 6A, 9V, and 23F (Bogaert serotypes 6B, 19F, 23F, 14 and 18C are common among children less than 2 years of age in Greece (Syrogiannopoulos et al., 2000), whereas serotypes 2004). In contrast, Soewignjo et al., (2001) reported that serogroups 6, 23, 15, 33, 19, 12, and 3 were common in Indonesia one series. In Kenya, the predominant serotypes were 13, 15, 14, 2004). The reasons for the observed differences are unclear. It has been speculated that living condition with poverty, crowding and exposure to patients with active diseases may partly account for their dominance in developing countries. Acute otitis media (AOM) has been reported as the most common disease condition associated with Streptococcus pneumoniae and remains the major predisposing factor for the development of antimicrobial resistant pneumococci (Hausdorff et albiofilms in the middle ear enhancing recurrence infection as well as promoting horizontal gene transfer (McEllistrem et al., 2007). Hausdorff found in 13-25% of middle ear fluid of those with otitis interna, 14 and 6B were isolated in 6of the isolates were 6A, 19A, and 9V. They also reported that serotypes 1, 3 and 5 are the major causes of acute otitis media in children less than 6 months old. In contrast, O’ Brien and Santosham, (2004) showed that serotypes 4, 6B, 9V, 18C, 19F and 23F are the common causes of invasive and middle ear infections in United States. In their research, Kilpi et al., (2001) reported that serpredominant serotypes associated with acute otitis media in Finland. This is similar to earlier report indicating that serotypes 19, 6, 3, 23, 11 and 18 are the common cause of acute otitis media in Finland (Luot1981). However, in another series Showal et al., (2006) showed that serotypes 1, 3, 5, 18C, 19A and 19F are the

is licensed under a Creative Commons Attribution 3.0 Unported License

28

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

varies between 18.2% to 22.1% and 24.6% to 31.8% respectively (Felmingham, 2002; Jacoobs et almost common serotypes resistant to penicillin and macrolides include 6A, 6B, 9V, 14, 19A, 19F and 23F

., 2000). Current prevention strategies include pneumococcal 23-valent polysaccharide vaccine cated in approximately 85% of disease in the US and Europe, 62.9% in Taiwan,

72.9% in Japan, 58% in Brazil and 79.9 % in India (Lee et al., 1991; Ferreira et al., 2006; Briles valent and 13-valent pneumococcal polysaccharide conjugate vaccines have

., 2011; Kim et al., 2012). The capsular polysaccharide vaccines are antibody unogenic in the target groups; young children, elderly people and

immunocompromised individuals, and also does not elicit immunologic memory (Briles ). The conjugate vaccines have shown high efficacy, but limited as they only provide

protection against the capsular serotypes/groups present in the vaccine resulting to capsular switch in types not incorporated in the conjugate vaccines (Kim et al., 2012).

Therefore, to overcome the increasing frequency of antibiotic-resistant strains and capsular switch, emphasises on the understanding of the mechanisms of pneumococcal virulence factors and development of

is needed. This review focuses on the various surface components implicated in nasopharyngeal isation and adherence, including PspA, CbpA/PspC, Pneumococcal adherence-virulence factor A (PavA),

pneumolysin and neuraminidase, and the potential targets for protein-based pneumococcal vaccine production.

Pneumococcal serotype distribution and clinical disease Asymptomatic pneumococcal colonisation of the nasopharynx is an important factor for disease initiation and provides the niche from where the bacteria can progress to the alveolar space, blood stream, middle ear or brain

caemia, otitis media and meningitis, respectively (MeCullers and Tuomanen, 2001). Pneumococcal carriage is more common in children compared to adults, reaching a prevalence of 55% at the

., 2004). Some underlying medical conditions are associated with pneumococcal disease, including congenital immunodeficiency, Human Immunodeficiency Virus (HIV), leukaemia, lymphoma, sickle cell disease and tissue transplantation (CDC, 1997). Other predisposing conditions to

ease include viral respiratory disease, chronic bronchitis, chronic obstructive pulmonary disease and smoking. The various clinical disease of Streptococcus pneumoniae is presented in figure 1.1.

., (2004) reported that serotypes 6A, 6B, 14, 15, 18C, 19F, and 23F are frequently found in nasopharyngeal colonisation. However, Syrjanen and colleagues reported that serotypes 6A, 6B, 23F, 19F, 11 and 14 were common isolates from nasopharyngeal samples from Finland (Syrjanen Netherlands, the common serotypes include 19F, 6B, 6A, 9V, and 23F (Bogaert et al., 2004). Similarly, serotypes 6B, 19F, 23F, 14 and 18C are common among children less than 2 years of age in Greece

., 2000), whereas serotypes 6B, 14, 19F, and 23 are common in USA (Bogaert ., (2001) reported that serogroups 6, 23, 15, 33, 19, 12, and 3 were common

in Indonesia one series. In Kenya, the predominant serotypes were 13, 15, 14, 6b, and 19F (Bogaert The reasons for the observed differences are unclear. It has been speculated that living condition with

poverty, crowding and exposure to patients with active diseases may partly account for their dominance in

Acute otitis media (AOM) has been reported as the most common disease condition associated with and remains the major predisposing factor for the development of antimicrobial

et al., 2002). This may be attributed to the continuous development of biofilms in the middle ear enhancing recurrence infection as well as promoting horizontal gene transfer

., 2007). Hausdorff et al., (2002) in a global study showed that serotypes 19F and 25% of middle ear fluid of those with otitis interna, 14 and 6B were isolated in 6-18% while 5

of the isolates were 6A, 19A, and 9V. They also reported that serotypes 1, 3 and 5 are the major causes of acute en less than 6 months old. In contrast, O’ Brien and Santosham, (2004) showed that

serotypes 4, 6B, 9V, 18C, 19F and 23F are the common causes of invasive and middle ear infections in United ., (2001) reported that serotypes 19F, 23F, 6A, 6B, 14 and 11 were the

predominant serotypes associated with acute otitis media in Finland. This is similar to earlier report indicating that serotypes 19, 6, 3, 23, 11 and 18 are the common cause of acute otitis media in Finland (Luot1981). However, in another series Showal et al., (2006) showed that serotypes 1, 3, 5, 18C, 19A and 19F are the

Creative Commons Attribution 3.0 Unported License

et al., 2003). The most common serotypes resistant to penicillin and macrolides include 6A, 6B, 9V, 14, 19A, 19F and 23F

valent polysaccharide vaccine cated in approximately 85% of disease in the US and Europe, 62.9% in Taiwan,

., 2006; Briles et al., 1998). In valent pneumococcal polysaccharide conjugate vaccines have ., 2012). The capsular polysaccharide vaccines are antibody

unogenic in the target groups; young children, elderly people and immunocompromised individuals, and also does not elicit immunologic memory (Briles et al., 1998;

they only provide protection against the capsular serotypes/groups present in the vaccine resulting to capsular switch in types not

sistant strains and capsular switch, emphasises on the understanding of the mechanisms of pneumococcal virulence factors and development of protein

is needed. This review focuses on the various surface components implicated in nasopharyngeal virulence factor A (PavA),

based pneumococcal vaccine production.

Asymptomatic pneumococcal colonisation of the nasopharynx is an important factor for disease initiation and provides the niche from where the bacteria can progress to the alveolar space, blood stream, middle ear or brain

caemia, otitis media and meningitis, respectively (MeCullers and Tuomanen, 2001). Pneumococcal carriage is more common in children compared to adults, reaching a prevalence of 55% at the

nditions are associated with pneumococcal disease, including congenital immunodeficiency, Human Immunodeficiency Virus (HIV), leukaemia, lymphoma, sickle cell disease and tissue transplantation (CDC, 1997). Other predisposing conditions to

ease include viral respiratory disease, chronic bronchitis, chronic obstructive pulmonary is presented in figure 1.1.

6A, 6B, 14, 15, 18C, 19F, and 23F are frequently found in nasopharyngeal colonisation. However, Syrjanen and colleagues reported that serotypes 6A, 6B, 23F, 19F, 11 and 14 were common isolates from nasopharyngeal samples from Finland (Syrjanen et al., 2001). In

., 2004). Similarly, serotypes 6B, 19F, 23F, 14 and 18C are common among children less than 2 years of age in Greece

6B, 14, 19F, and 23 are common in USA (Bogaert et al., ., (2001) reported that serogroups 6, 23, 15, 33, 19, 12, and 3 were common

(Bogaert et al., The reasons for the observed differences are unclear. It has been speculated that living condition with

poverty, crowding and exposure to patients with active diseases may partly account for their dominance in

Acute otitis media (AOM) has been reported as the most common disease condition associated with and remains the major predisposing factor for the development of antimicrobial

ay be attributed to the continuous development of biofilms in the middle ear enhancing recurrence infection as well as promoting horizontal gene transfer

., (2002) in a global study showed that serotypes 19F and 23F were 18% while 5-10%

of the isolates were 6A, 19A, and 9V. They also reported that serotypes 1, 3 and 5 are the major causes of acute en less than 6 months old. In contrast, O’ Brien and Santosham, (2004) showed that

serotypes 4, 6B, 9V, 18C, 19F and 23F are the common causes of invasive and middle ear infections in United otypes 19F, 23F, 6A, 6B, 14 and 11 were the

predominant serotypes associated with acute otitis media in Finland. This is similar to earlier report indicating that serotypes 19, 6, 3, 23, 11 and 18 are the common cause of acute otitis media in Finland (Luotonen et al., 1981). However, in another series Showal et al., (2006) showed that serotypes 1, 3, 5, 18C, 19A and 19F are the

Page 3: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: most frequent serotypes associated to acute otitis mediatypeable pneumococci are not involved in the development of acute otitis media. Recombination exchanges at the capsular biosynthetic locus results in serotype and this have been shown among pneumococ(Coffey et al., 1998). In Israel, the most common serotypes associated with acute otitis media were 14, 19F, 23F, 19A, 6B, 6B, 11A, 33 A and 18C (Moschoioni infections are generally related to age, predisposing factors, the immune status of the host, and other virulence factors of the infecting pneumococcal strain and that the infecting serotype/type is of minor importance. Bacteraemia, pneumonia and meningitis, including other systemic diseases such as endocarditis and pleural empyema are termed invasive pneumococcal disease. According to Hausdorff 2, 5, 6, 14, 19, and 23 are the most pred9V, 3, 23F and 7F are highly associated with invasive pneumococcal disease (Kanasanterveyslaitos However, O’Brien and Santosham, (2004) demonstrated that serotypes 4, 6mostly associated with invasive pneumococcal disease in USA. In another series, Brueggemann et al., (2003) reported that serotypes 1, 4, 14, and 18C are the major pneumococcal types involved in invasive disease in England, whereas serotypes 14, 9, 6, 19, 23, 8 and 4 causes invasive disease in Scotland (Kyaw et al., 2003). In contrast, the most common serogroups associated with invasive disease in Bangladesh are 2, 1, 14, 5, 45, and 12A (Saha et al., 2009). In addition, invaand 18C in Sweden (Sandgren et al., 2004). Furthermore, in India pneumococcal serotypes 1, 6, 19, 7, 5, 15, 14, 4, 16, and 18 cause invasive disease (INCLEAN, 1999). In the HIV positive padisease is caused by serotypes 6A, 6B, 9N, 9V, 18C, 19A, 19F, and 23F (Fry et al., 2003). The variations in the serotype distribution in invasive pneumococcal disease may be attributed to the introduction of the current 23valent vaccine which includes the serotypes that cause 88% of invasive disease in USA and 96% of those in the UK. Pneumococcal virulence factors and potential targets for proteinStreptococcus pneumoniae possesses several virulent factors thatnasopharynx and development of invasive disease. Currently, there are 91 know capsular serotypes of Streptococcus pneumoniae with variations in the polysaccharide structure occurring between different capsular serotypes (Park et al., 2007). The representation of the cell membrane, cell wall and capsule of pneumoniae is shown in figure 1. 3. PiaA PiaA is one of the various iron acquisition systems in immunogenic, found in all pneumococcal serotypes and highly conserved between strains, and this makes it a potencial vaccine candidate (Jomaa et althe nasopharyngeal washing of a mice mmight be responsible for adherence and colonisation. Intranasal and intraperitoneal immunisation has been effective in protecting against systemic infection and pneumonia respectively (Brown e2006).

is licensed under a Creative Commons Attribution 3.0 Unported License

29

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

most frequent serotypes associated to acute otitis media in Israel. They further stated that 6A, 6B, 15A and nontypeable pneumococci are not involved in the development of acute otitis media. Recombination exchanges at the capsular biosynthetic locus results in serotype and this have been shown among pneumococ

., 1998). In Israel, the most common serotypes associated with acute otitis media were 14, 19F, 23F, 19A, 6B, 6B, 11A, 33 A and 18C (Moschoioni et al., 2010).It can be deduced that the outcome of clinical

related to age, predisposing factors, the immune status of the host, and other virulence factors of the infecting pneumococcal strain and that the infecting serotype/type is of minor importance.

Bacteraemia, pneumonia and meningitis, including other systemic diseases such as endocarditis and pleural empyema are termed invasive pneumococcal disease. According to Hausdorff et al., (2005), serotypes/groups 1, 2, 5, 6, 14, 19, and 23 are the most predominant types in invasive disease globally. In Finland, serotypes 14, 4, 9V, 3, 23F and 7F are highly associated with invasive pneumococcal disease (Kanasanterveyslaitos However, O’Brien and Santosham, (2004) demonstrated that serotypes 4, 6B, 9V, 14, 18C, 19F, and 23F were mostly associated with invasive pneumococcal disease in USA. In another series, Brueggemann et al., (2003) reported that serotypes 1, 4, 14, and 18C are the major pneumococcal types involved in invasive disease in

whereas serotypes 14, 9, 6, 19, 23, 8 and 4 causes invasive disease in Scotland (Kyaw et al., 2003). In contrast, the most common serogroups associated with invasive disease in Bangladesh are 2, 1, 14, 5, 45, and

., 2009). In addition, invasive pneumococcal disease is caused by serotypes 1, 4, 7F, 9V, 12F, ., 2004). Furthermore, in India pneumococcal serotypes 1, 6, 19, 7, 5, 15, 14,

4, 16, and 18 cause invasive disease (INCLEAN, 1999). In the HIV positive patients, invasive pneumococcal disease is caused by serotypes 6A, 6B, 9N, 9V, 18C, 19A, 19F, and 23F (Fry et al., 2003). The variations in the serotype distribution in invasive pneumococcal disease may be attributed to the introduction of the current 23

ent vaccine which includes the serotypes that cause 88% of invasive disease in USA and 96% of those in the

Pneumococcal virulence factors and potential targets for protein-based vaccines possesses several virulent factors that promote their establishment in the

nasopharynx and development of invasive disease. Currently, there are 91 know capsular serotypes of with variations in the polysaccharide structure occurring between different capsular

., 2007). The representation of the cell membrane, cell wall and capsule of

PiaA is one of the various iron acquisition systems in S. pneumoniae. RFLP analysis indicates that it is highly mmunogenic, found in all pneumococcal serotypes and highly conserved between strains, and this makes it a

et al., 2006). LeMessurier et al., (2006) demonstrated high level of PiaA in the nasopharyngeal washing of a mice model challenged with D39 and WCH16 strains and proposed that it might be responsible for adherence and colonisation. Intranasal and intraperitoneal immunisation has been effective in protecting against systemic infection and pneumonia respectively (Brown et al., 2001; Jomaa

Creative Commons Attribution 3.0 Unported License

in Israel. They further stated that 6A, 6B, 15A and non-typeable pneumococci are not involved in the development of acute otitis media. Recombination exchanges at the capsular biosynthetic locus results in serotype and this have been shown among pneumococcal isolates

., 1998). In Israel, the most common serotypes associated with acute otitis media were 14, 19F, ., 2010).It can be deduced that the outcome of clinical

related to age, predisposing factors, the immune status of the host, and other virulence factors of the infecting pneumococcal strain and that the infecting serotype/type is of minor importance.

Bacteraemia, pneumonia and meningitis, including other systemic diseases such as endocarditis and pleural ., (2005), serotypes/groups 1,

ominant types in invasive disease globally. In Finland, serotypes 14, 4, 9V, 3, 23F and 7F are highly associated with invasive pneumococcal disease (Kanasanterveyslaitos et al., 2005).

B, 9V, 14, 18C, 19F, and 23F were mostly associated with invasive pneumococcal disease in USA. In another series, Brueggemann et al., (2003) reported that serotypes 1, 4, 14, and 18C are the major pneumococcal types involved in invasive disease in

whereas serotypes 14, 9, 6, 19, 23, 8 and 4 causes invasive disease in Scotland (Kyaw et al., 2003). In contrast, the most common serogroups associated with invasive disease in Bangladesh are 2, 1, 14, 5, 45, and

sive pneumococcal disease is caused by serotypes 1, 4, 7F, 9V, 12F, ., 2004). Furthermore, in India pneumococcal serotypes 1, 6, 19, 7, 5, 15, 14,

tients, invasive pneumococcal disease is caused by serotypes 6A, 6B, 9N, 9V, 18C, 19A, 19F, and 23F (Fry et al., 2003). The variations in the serotype distribution in invasive pneumococcal disease may be attributed to the introduction of the current 23-

ent vaccine which includes the serotypes that cause 88% of invasive disease in USA and 96% of those in the

promote their establishment in the nasopharynx and development of invasive disease. Currently, there are 91 know capsular serotypes of

with variations in the polysaccharide structure occurring between different capsular ., 2007). The representation of the cell membrane, cell wall and capsule of Streptococcus

. RFLP analysis indicates that it is highly mmunogenic, found in all pneumococcal serotypes and highly conserved between strains, and this makes it a

., (2006) demonstrated high level of PiaA in odel challenged with D39 and WCH16 strains and proposed that it

might be responsible for adherence and colonisation. Intranasal and intraperitoneal immunisation has been t al., 2001; Jomaa et al.,

Page 4: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,:

Figure 1.1: Infections caused by S. pneumoniaepneumococcal diseases are preceded by nasopharyngeal carriage, after which the bacterium can spread to the middle ear causing otitis media or disseminate to the lungs causing pneumonia. During infecbacteria may penetrate into the blood circulation, from which they can spread through the whole body. Otitis media might also cause infection of the meninges. Some wellcontributions to virulence at that particular site in the human or murine body, are indicated in dotted boxes. Ami- AliA/B, oligopeptide ABC transporter; CbpA, choline binssding protein A; CiaRH, the Cia twocomponent system; ComDE, two-component system specific for sensing of competeCps, polysaccharide capsule; Iga, immunoglobulin A1 protease; LytA, autolysin A; LytB, autolysin B; NanA, neuraminidase A; NanB, neuraminidase B; PavA, pneumococcal adherence and virulence factor A; phosphocholine; PcpA, choline binding protein PcpA; Ply, pneumolysin; PsaA, pneumococcal surface adhesin A [Mn2+ uptake ABC transporter]; PspA, pneumococcal surface protein A; ZmpC, metalloprotease.Source: Modified from Hendriksen et al Neuraminidase Neuraminidase (nanA), an exoglycosidase that cleaves the terminal sialic acid moieties from glycolipids, mucins, glycoproteins and oligopolysaccharides, is found in virtually all respiratory pathogens. The nanA and nanB are the major neuraminidase found in gene is universally present in all pneumococcal strains (Pettigrew 96% of a series of isolates and nanC gene Whenviewed on transparent media, pneumococcal colonies can be transparent or opaque. The transparent phenotype is frequently linked to nasopharyngeal colonization and invasion of the brain microvascular endothelial cells whereas the opaque phenotype is common in the blood during sepsis in a mice model (LeMessurier et al., 2006). King et alisolated from nasopharynx using a microarray technique. There are evidences showing thdesialation of the cell surfaces of Neisseria meningitidisglycoconjugate receptors, including lactoferrin and IgA2 (King nasopharyngeal carriage of other bacterial species, thereby enhancing the adhesion of respiratory epithelia. However, NanA is not associated with sepsis because nanA knockout mutant was not attenuated after an intraperitoneal inoculation in a murine model ( Choline Binding Proteins Streptococcus pneumoniae has several surface proteins called choline binding proteins (CBPs) that are attached to the cell wall techoic and lipotechoic acids (Rosenow same portion of choline Binding Region (CBR) consisting of between 2 and 10 highly conserved 20 amino acid repeats which bind non-covalently to chop residues (Garcia be widely described include autolysin (LytA) and pneumococcal surface protein A, PspA (Talkington 1991; Yother and Briles, 1992). Subsequently, other CBPs such as CbpA or PspC (Rosenow Brooks-Walter et al., 1999), cell wall hydrolases (LytB and LytC), amidase (CbpD), and serine protease (CbpG)

is licensed under a Creative Commons Attribution 3.0 Unported License

30

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

S. pneumoniae. Black arrows indicate the progression of infection. All

pneumococcal diseases are preceded by nasopharyngeal carriage, after which the bacterium can spread to the middle ear causing otitis media or disseminate to the lungs causing pneumonia. During infection of the lungs, bacteria may penetrate into the blood circulation, from which they can spread through the whole body. Otitis media might also cause infection of the meninges. Some well-known examples of proteins, which have

that particular site in the human or murine body, are indicated in dotted boxes. AliA/B, oligopeptide ABC transporter; CbpA, choline binssding protein A; CiaRH, the Cia two

component system specific for sensing of competence stimulating peptide; Cps, polysaccharide capsule; Iga, immunoglobulin A1 protease; LytA, autolysin A; LytB, autolysin B; NanA, neuraminidase A; NanB, neuraminidase B; PavA, pneumococcal adherence and virulence factor A;

ine binding protein PcpA; Ply, pneumolysin; PsaA, pneumococcal surface adhesin A [Mn2+ uptake ABC transporter]; PspA, pneumococcal surface protein A; ZmpC, metalloprotease.

et al., (2009).

), an exoglycosidase that cleaves the terminal sialic acid moieties from glycolipids, mucins, glycoproteins and oligopolysaccharides, is found in virtually all respiratory pathogens. The nanA and nanB are the major neuraminidase found in S. pneumoniae, although nanC can also be present. However,

is universally present in all pneumococcal strains (Pettigrew et al., 2006) whereas nanB genegene was present in only 51% of the same series (Pettigrew

viewed on transparent media, pneumococcal colonies can be transparent or opaque. The transparent phenotype is frequently linked to nasopharyngeal colonization and invasion of the brain microvascular

ue phenotype is common in the blood during sepsis in a mice model et al., (2004) demonstrated that nanA was more abundant in pneumococci

isolated from nasopharynx using a microarray technique. There are evidences showing that nanA can cause Neisseria meningitidis and Haemophilus influenzae and also modify the

glycoconjugate receptors, including lactoferrin and IgA2 (King et al., 2004). This is thought to decrease the e of other bacterial species, thereby enhancing the adhesion of S. pneumoniae

respiratory epithelia. However, NanA is not associated with sepsis because nanA knockout mutant was not attenuated after an intraperitoneal inoculation in a murine model (Berry and Paton, 2000).

has several surface proteins called choline binding proteins (CBPs) that are attached to the cell wall techoic and lipotechoic acids (Rosenow et al., 1997; Garcia et al., 1998). CPB is found at the same portion of choline Binding Region (CBR) consisting of between 2 and 10 highly conserved 20 amino acid

covalently to chop residues (Garcia et al., 1998). The first two pneumococcal CBPs to described include autolysin (LytA) and pneumococcal surface protein A, PspA (Talkington

1991; Yother and Briles, 1992). Subsequently, other CBPs such as CbpA or PspC (Rosenow ), cell wall hydrolases (LytB and LytC), amidase (CbpD), and serine protease (CbpG)

Creative Commons Attribution 3.0 Unported License

. Black arrows indicate the progression of infection. All pneumococcal diseases are preceded by nasopharyngeal carriage, after which the bacterium can spread to the

tion of the lungs, bacteria may penetrate into the blood circulation, from which they can spread through the whole body. Otitis

known examples of proteins, which have that particular site in the human or murine body, are indicated in dotted boxes.

AliA/B, oligopeptide ABC transporter; CbpA, choline binssding protein A; CiaRH, the Cia two-nce stimulating peptide;

Cps, polysaccharide capsule; Iga, immunoglobulin A1 protease; LytA, autolysin A; LytB, autolysin B; NanA, neuraminidase A; NanB, neuraminidase B; PavA, pneumococcal adherence and virulence factor A; PCho,

ine binding protein PcpA; Ply, pneumolysin; PsaA, pneumococcal surface adhesin A [Mn2+ uptake ABC transporter]; PspA, pneumococcal surface protein A; ZmpC, metalloprotease.

), an exoglycosidase that cleaves the terminal sialic acid moieties from glycolipids, mucins, glycoproteins and oligopolysaccharides, is found in virtually all respiratory pathogens. The nanA and

, although nanC can also be present. However, nanA genewas present in

rew et al., 2006). viewed on transparent media, pneumococcal colonies can be transparent or opaque. The transparent

phenotype is frequently linked to nasopharyngeal colonization and invasion of the brain microvascular ue phenotype is common in the blood during sepsis in a mice model

., (2004) demonstrated that nanA was more abundant in pneumococci at nanA can cause

and also modify the ., 2004). This is thought to decrease the

S. pneumoniae to the respiratory epithelia. However, NanA is not associated with sepsis because nanA knockout mutant was not

has several surface proteins called choline binding proteins (CBPs) that are attached ., 1998). CPB is found at the

same portion of choline Binding Region (CBR) consisting of between 2 and 10 highly conserved 20 amino acid ., 1998). The first two pneumococcal CBPs to

described include autolysin (LytA) and pneumococcal surface protein A, PspA (Talkington et al., 1991; Yother and Briles, 1992). Subsequently, other CBPs such as CbpA or PspC (Rosenow et al., 1997;

), cell wall hydrolases (LytB and LytC), amidase (CbpD), and serine protease (CbpG)

Page 5: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: were demonstrated (Garcia et al., 1999; Gosink least 10 pneumococcal CBPs have been characterized (Kausmally, 2005). Pneumococcal Surface Protein A (PspAproteins associated with virulence. PspA was the first surface exposed protein that was discovered to bind choline residues (Hollinghead et al., 2000). PspA is attached to the cell wall throughphosphorylcholine (ChoP) moieties on techoic and lipotechoic acids and its Csequence homology with CBR of CbpA (Brooksextends beyond the polysaccharide capsule (Yother and Briles, 1992). The molecular weight of PspA varies between strains from 67-99 kDa and is found in all clinically important serotypes of 1990; Waltman et al., 1990; Jedrzejas pneumoniae during infection and influence complement deposition through alternative pathway, but it is still unclear how this happens (Ogunniyi et althe α-helical N-terminal region of PspA is immunogenic and further showed that immunisation with a 43 kDa fragment containing this portion protects a mice challenged by intravenous and intratracheal inoculation. In human clinical studies, prior presence of antibodies to PspA was shown to influence the susceptibility of pneumoniae infection (Talkington et alpneumococcal vaccine (Briles et al., 1996; Briles There is increasing evidence showing that PspA have a role in inhibiting complementand colleagues reported that PspA-negative strains are cleared rapidly from blood after intravenous inoculation, but levels of isogenic PspA-positive strain increased rapidly (Tu deficient in C3 or factor B are unable clear mutant strains lacking PspA while C5 deficient mice cleared the same mutant strains. In addition, Serum C3 was significantly reduced within strain whereas no significant reduction of serum C3 occurred in mice infected with a PspAal., 1999). Similarly, a profound increase in C3 binding was discovered when both both PspApneumococci were incubated with anticollegues demonstrated that mutant pneumococcal strains without PspA and pneumolysin were unable to spread from the lung into the blood in mice with normal complethat C3 deposition is inhibited by PspA modulating the alternative pathway and pneumolysin inhibition of the classical pathway, which helps in the establishment of demonstrated that PspA is found majorly in blood in both D39 and WCH16 strains using RNA microarray, and proposed that PspA is a major virulent factor for sepsis than nasopharyngeal carriage. The inhibition of complement clearance of pneumococci by PspA in the blood enhances its ability to cause sepsis. Since highly immunogenic, it is possible that it can be used the development of a protein based vaccine. CbpA, also called PspC (Pneumococcal Surface Protein C) and SpsA protein IgA) is exposed to the surface of the pneumococcal capsule and is encoded by a heterogeneous group of mosaic genes (Brooks-Walter et al., 1999). In 1997, Rosenow and colleagues demonstrated that there is a reduction in the ability of CbpA-negative epithelial cells and endothelial cells when compared to the parent strain, and an almost 100nasopharyngeal carriage in an infant rat model indica(Rosenow et al., 1997). Similarly, Balachandran CbpA were unable to colonise the nasopharynx of a mouse model, and were unable to invade and multiply in the lungs. In addition, using RNA microarray LeMessurier and colleagues demonstrated that CbpA is the major virulent factor for the development of pneumonia in both D39 and WCH16 strains as more concentrations were detected in the lung of an animal model (LeMessurier promote pneumococcal adherence to the brain microvascular endothelsubarachnoid space (Ring et al., 1998; Orihuela Zhang et al., (2000) reported that lack of CbpA decreased the ability of mutant pneumococcal strain to invade the nasopharyngeal cells by more than 90% whenthat a knockout mice lacking pIgR trafficking has a delayed onset of bacteraemia after intranasal inoculation. As a result, they proposed that pneumococcal CbpA interact with human pIgR and invthe pIgR. This is similar to the report of Koedel

is licensed under a Creative Commons Attribution 3.0 Unported License

31

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

., 1999; Gosink et al., 2000; Kausmally et al., 2005). Reports inleast 10 pneumococcal CBPs have been characterized (Kausmally, 2005).

PspA)and Choline binding protein A (CbpA) are major choline binding proteins associated with virulence. PspA was the first surface exposed protein that was discovered to bind

., 2000). PspA is attached to the cell wall through the interface between phosphorylcholine (ChoP) moieties on techoic and lipotechoic acids and its C-terminal CBR, which has 95sequence homology with CBR of CbpA (Brooks-Walter et al., 1999). The highly charged N-

saccharide capsule (Yother and Briles, 1992). The molecular weight of PspA varies 99 kDa and is found in all clinically important serotypes of S. pneumoniae

1990; Waltman et al., 1990; Jedrzejas et al., 2000). Reports have indicated that PspA is upduring infection and influence complement deposition through alternative pathway, but it is still

et al., 2002; Yutse et al., 2005). Talkington et al., (1991) demterminal region of PspA is immunogenic and further showed that immunisation with a 43 kDa

fragment containing this portion protects a mice challenged by intravenous and intratracheal inoculation. In ior presence of antibodies to PspA was shown to influence the susceptibility of

et al., 1996; McCool et al., 2002). PspA is a potential candidate for ., 1996; Briles et al., 1998).

e is increasing evidence showing that PspA have a role in inhibiting complement-mediated opsonisation. Tu negative strains are cleared rapidly from blood after intravenous inoculation,

strain increased rapidly (Tu et al., 1999). They also showed that mice deficient in C3 or factor B are unable clear mutant strains lacking PspA while C5 deficient mice cleared the same mutant strains. In addition, Serum C3 was significantly reduced within 30 minutes with PspAstrain whereas no significant reduction of serum C3 occurred in mice infected with a PspA-positive strain (Tu

., 1999). Similarly, a profound increase in C3 binding was discovered when both both PspAci were incubated with anti-PspA (Ren et al., 2003; Ren et al., 2004). Furthermore, Yutse and

collegues demonstrated that mutant pneumococcal strains without PspA and pneumolysin were unable to spread from the lung into the blood in mice with normal complement function (Yutse et al., 2005). They also proposed that C3 deposition is inhibited by PspA modulating the alternative pathway and pneumolysin inhibition of the classical pathway, which helps in the establishment of S. pneumoniae septicaemia. LeMessuriedemonstrated that PspA is found majorly in blood in both D39 and WCH16 strains using RNA microarray, and proposed that PspA is a major virulent factor for sepsis than nasopharyngeal carriage. The inhibition of

ococci by PspA in the blood enhances its ability to cause sepsis. Since immunogenic, it is possible that it can be used the development of a protein based vaccine.

CbpA, also called PspC (Pneumococcal Surface Protein C) and SpsA (Streptococcus pneumoniaeprotein IgA) is exposed to the surface of the pneumococcal capsule and is encoded by a heterogeneous group of

., 1999). In 1997, Rosenow and colleagues demonstrated that there is a negative S. pneumoniae mutant (CbpA-) to bind cytokine-activated human lung

epithelial cells and endothelial cells when compared to the parent strain, and an almost 100-fold decrease in nasopharyngeal carriage in an infant rat model indicating that CbpA plays a role as adhesin in

., 1997). Similarly, Balachandran et al., (2002) reported that S. pneumoniae CbpA were unable to colonise the nasopharynx of a mouse model, and were unable to invade and multiply in the lungs. In addition, using RNA microarray LeMessurier and colleagues demonstrated that CbpA is the major

velopment of pneumonia in both D39 and WCH16 strains as more concentrations were detected in the lung of an animal model (LeMessurier et al., 2006). Furthermore, CbpA has also been shown to promote pneumococcal adherence to the brain microvascular endothelium and subsequent invasion of the

., 1998; Orihuela et al., 2004).

., (2000) reported that lack of CbpA decreased the ability of mutant pneumococcal strain to invade the nasopharyngeal cells by more than 90% when compared to the parent strain. Furthermore, they demonstrated that a knockout mice lacking pIgR trafficking has a delayed onset of bacteraemia after intranasal inoculation. As a result, they proposed that pneumococcal CbpA interact with human pIgR and invades cells by subversion of the pIgR. This is similar to the report of Koedel et al., (2002) which shows that CbpA is necessary for

Creative Commons Attribution 3.0 Unported License

., 2005). Reports indicate that at

and Choline binding protein A (CbpA) are major choline binding proteins associated with virulence. PspA was the first surface exposed protein that was discovered to bind

the interface between terminal CBR, which has 95-95%

-terminal region saccharide capsule (Yother and Briles, 1992). The molecular weight of PspA varies

S. pneumoniae (Crain et al., have indicated that PspA is up-regulated in S.

during infection and influence complement deposition through alternative pathway, but it is still ., (1991) demonstrated that

terminal region of PspA is immunogenic and further showed that immunisation with a 43 kDa fragment containing this portion protects a mice challenged by intravenous and intratracheal inoculation. In

ior presence of antibodies to PspA was shown to influence the susceptibility of S. ., 2002). PspA is a potential candidate for

mediated opsonisation. Tu negative strains are cleared rapidly from blood after intravenous inoculation,

., 1999). They also showed that mice deficient in C3 or factor B are unable clear mutant strains lacking PspA while C5 deficient mice cleared the

30 minutes with PspA-negative positive strain (Tu et

., 1999). Similarly, a profound increase in C3 binding was discovered when both both PspA- mutant and ., 2004). Furthermore, Yutse and

collegues demonstrated that mutant pneumococcal strains without PspA and pneumolysin were unable to spread ., 2005). They also proposed

that C3 deposition is inhibited by PspA modulating the alternative pathway and pneumolysin inhibition of the septicaemia. LeMessurier et al., (2006)

demonstrated that PspA is found majorly in blood in both D39 and WCH16 strains using RNA microarray, and proposed that PspA is a major virulent factor for sepsis than nasopharyngeal carriage. The inhibition of

ococci by PspA in the blood enhances its ability to cause sepsis. Since PspA is immunogenic, it is possible that it can be used the development of a protein based vaccine.

ccus pneumoniae secretary protein IgA) is exposed to the surface of the pneumococcal capsule and is encoded by a heterogeneous group of

., 1999). In 1997, Rosenow and colleagues demonstrated that there is a activated human lung

fold decrease in ting that CbpA plays a role as adhesin in S. pneumoniae

mutants lacking CbpA were unable to colonise the nasopharynx of a mouse model, and were unable to invade and multiply in the lungs. In addition, using RNA microarray LeMessurier and colleagues demonstrated that CbpA is the major

velopment of pneumonia in both D39 and WCH16 strains as more concentrations were ., 2006). Furthermore, CbpA has also been shown to

ium and subsequent invasion of the

., (2000) reported that lack of CbpA decreased the ability of mutant pneumococcal strain to invade compared to the parent strain. Furthermore, they demonstrated

that a knockout mice lacking pIgR trafficking has a delayed onset of bacteraemia after intranasal inoculation. As ades cells by subversion of

., (2002) which shows that CbpA is necessary for S.

Page 6: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: pneumoniae to invade blood-brain barrier by biand crossing the capillary endothelium by transcytosis. However, Brock pIgR-mediated invasion is only found in Detroitmediated pneumococcal invasion is strain and cell specific. CbpA is highly polymorphic (Iannelli and there is substantial strain to strain variation in 2006). In addition to a virulence role in adherence and invasion, CbpA is also involved in complement by binding to factor H (Dave the attachment of factor B to complement component 3b (C3b) and therefore, prevents host cells from being attacked by its own complement pathway (Dave binding pneumococcal CbpA, factor H is then able to bind C3b deposited on the the complement pathway at this crucial phase. In addition, CbpA could decrease alternative pathway activity by modulating the rate of C3 degradation to iC3b, causing dissociation of factor B (Bf) from the C3 convertase reducing C3b deposition, or inhibiting C3bBb formation by preferentially binding C3b. CbpA is a potential vaccine candidate as a result of its role in carriage and pathogenesis. Furthermore, the CbpA is extends beyond the capsule and therefore, carriage can beantibodies. Reports of immunization studies indicate that CbpA can be highly protective against in mice model following intra-peritoneal challenge (Brooks Pneumolysin The toxigenic potential of pneumolysin (Ply) in the pathogenesis of pneumococcal disease has been demonstrated and it is found in practically all clinical isolates of toxigenic potential lies on its interaction of the cell membranes which results in transmembrane pore formation allowing the influx of water, ions and some organic macromolecules, culminating to cell lysis (Boulnois 1991). In addition, it has been proposed that Ply may also induce upregulation of nitric oxide production by macrophages causing damage to the host’s cells (Braun been shown to cause activation of the classical pathwayantibody through binding of C1 to Ply domain which is common to the Fc portion of IgG (Paton Mitchell et al., 1991). Jounblat et al., (2003) demonstrated that the growth of blood is as a result of complement activating property of Ply. Various studies have shown the association of Ply and pneumococcal septicaemia, and reduction in replication and survival in the blood of an animal model was found in Ply-negative mutant strains (Benton The alteration of the plasma membrane integrity by ply has been shown to stimulate the production and release of cytokine IL-8 and TNFα by neutrophils (Cockeran mediated the production of TNFα, IL-deduced that the induction of inflammatory responses might be a contributing factor to the severity opneumococcal disease and the high mortality rate. Furthermore, Ply has been demonstrated to inhibit the beating of the cilia lining the respiratory epithelium increasing the accumulation of the bacteria in the lung which may result to pneumonia (Boulnois et alpathogenesis, and further research is needed to unlock these other roles. Zinc Metalloproteinases The pneumococcus produces large proteases on its surface which are mainly zincmetalloproteinases and previously sequenced genomes showed that four can be found in an isolate (Chiavolini al., 2003). These are frequently associated with invasive infections and virulence in serotype 4 pneumococci, but not associated with virulence in seserotype 19F (Chiavolini et al., 2003). The IgA1 protease and ZmpC are the only zinc metalloproteinases that are sufficiently characterized. The Immunoglobulin A1 protease allow the pneumocoexplains its substantial diversity as it is required to cleave structurally diverse substrates (Poulsen To cleave structurally diverse substrates, pneumococcal strains show variation in the num

is licensed under a Creative Commons Attribution 3.0 Unported License

32

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

brain barrier by binding to the human polymeric immunoglobin receptor (pIgR) and crossing the capillary endothelium by transcytosis. However, Brock et al., (2002) demonstrated that CpbA

mediated invasion is only found in Detroit-562 cell line and therefore, suggested thmediated pneumococcal invasion is strain and cell specific. CbpA is highly polymorphic (Iannelli and there is substantial strain to strain variation in S. pneumoniae and this influences virulence (Kerr

In addition to a virulence role in adherence and invasion, CbpA is also involved in S. pneumoniaecomplement by binding to factor H (Dave et al., 2001; Kerr et al., 2006). Factor is a serum protein that impedes

lement component 3b (C3b) and therefore, prevents host cells from being attacked by its own complement pathway (Dave et al., 2001; Jarva et al., 2003). It can be deduced that by binding pneumococcal CbpA, factor H is then able to bind C3b deposited on the cell surface, thereby arresting the complement pathway at this crucial phase. In addition, CbpA could decrease alternative pathway activity by modulating the rate of C3 degradation to iC3b, causing dissociation of factor B (Bf) from the C3 convertase

ing C3b deposition, or inhibiting C3bBb formation by preferentially binding C3b.

CbpA is a potential vaccine candidate as a result of its role in carriage and pathogenesis. Furthermore, the CbpA is extends beyond the capsule and therefore, carriage can be abolished by either opsonic or neutralizing antibodies. Reports of immunization studies indicate that CbpA can be highly protective against

peritoneal challenge (Brooks-Walter et al., 1999; Ogunniyi et al., 2001

The toxigenic potential of pneumolysin (Ply) in the pathogenesis of pneumococcal disease has been demonstrated and it is found in practically all clinical isolates of S. pneumoniae (Paton et altoxigenic potential lies on its interaction of the cell membranes which results in transmembrane pore formation allowing the influx of water, ions and some organic macromolecules, culminating to cell lysis (Boulnois

, it has been proposed that Ply may also induce upregulation of nitric oxide production by macrophages causing damage to the host’s cells (Braun et al., 1999; Braun et al., 2000). Furthermore, Ply has been shown to cause activation of the classical pathway of complement system in the absence of specific antibody through binding of C1 to Ply domain which is common to the Fc portion of IgG (Paton

., (2003) demonstrated that the growth of S. pneumoniae blood is as a result of complement activating property of Ply. Various studies have shown the association of Ply and pneumococcal septicaemia, and reduction in replication and survival in the blood of an animal model was

ant strains (Benton et al., 1995; Berry et al., 1999).

The alteration of the plasma membrane integrity by ply has been shown to stimulate the production and release by neutrophils (Cockeran et al., 2002). In addition, reports indicate that Ply

-1β, and IL-6 by blood monocytes (Houldsworth et al., 1994). It can be deduced that the induction of inflammatory responses might be a contributing factor to the severity opneumococcal disease and the high mortality rate. Furthermore, Ply has been demonstrated to inhibit the beating of the cilia lining the respiratory epithelium increasing the accumulation of the bacteria in the lung which may

et al., 1991). It is likely that Ply has multiple roles in pneumococcal pathogenesis, and further research is needed to unlock these other roles.

The pneumococcus produces large proteases on its surface which are mainly zinc lloproteinases and previously sequenced genomes showed that four can be found in an isolate (Chiavolini

., 2003). These are frequently associated with invasive infections and virulence in serotype 4 pneumococci, but not associated with virulence in serotype 3 pneumococci and only two are important for virulence in

., 2003). The IgA1 protease and ZmpC are the only zinc metalloproteinases that

allow the pneumococcus to evade host mucosal immunoglobulin which explains its substantial diversity as it is required to cleave structurally diverse substrates (Poulsen To cleave structurally diverse substrates, pneumococcal strains show variation in the number and sequence of

Creative Commons Attribution 3.0 Unported License

nding to the human polymeric immunoglobin receptor (pIgR) ., (2002) demonstrated that CpbA-

562 cell line and therefore, suggested that CpbA-pIgR-mediated pneumococcal invasion is strain and cell specific. CbpA is highly polymorphic (Iannelli et al., 2002)

e and this influences virulence (Kerr et al.,

S. pneumoniae evasion of ., 2006). Factor is a serum protein that impedes

lement component 3b (C3b) and therefore, prevents host cells from being ., 2003). It can be deduced that by

cell surface, thereby arresting the complement pathway at this crucial phase. In addition, CbpA could decrease alternative pathway activity by modulating the rate of C3 degradation to iC3b, causing dissociation of factor B (Bf) from the C3 convertase

CbpA is a potential vaccine candidate as a result of its role in carriage and pathogenesis. Furthermore, the CbpA abolished by either opsonic or neutralizing

antibodies. Reports of immunization studies indicate that CbpA can be highly protective against S. pneumoniae ., 2001).

The toxigenic potential of pneumolysin (Ply) in the pathogenesis of pneumococcal disease has been et al., 1993). Its

toxigenic potential lies on its interaction of the cell membranes which results in transmembrane pore formation allowing the influx of water, ions and some organic macromolecules, culminating to cell lysis (Boulnois et al.,

, it has been proposed that Ply may also induce upregulation of nitric oxide production by ., 2000). Furthermore, Ply has

of complement system in the absence of specific antibody through binding of C1 to Ply domain which is common to the Fc portion of IgG (Paton et al., 1984;

in the lung and blood is as a result of complement activating property of Ply. Various studies have shown the association of Ply and pneumococcal septicaemia, and reduction in replication and survival in the blood of an animal model was

The alteration of the plasma membrane integrity by ply has been shown to stimulate the production and release ., 2002). In addition, reports indicate that Ply

., 1994). It can be deduced that the induction of inflammatory responses might be a contributing factor to the severity of pneumococcal disease and the high mortality rate. Furthermore, Ply has been demonstrated to inhibit the beating of the cilia lining the respiratory epithelium increasing the accumulation of the bacteria in the lung which may

., 1991). It is likely that Ply has multiple roles in pneumococcal

lloproteinases and previously sequenced genomes showed that four can be found in an isolate (Chiavolini et ., 2003). These are frequently associated with invasive infections and virulence in serotype 4 pneumococci,

rotype 3 pneumococci and only two are important for virulence in ., 2003). The IgA1 protease and ZmpC are the only zinc metalloproteinases that

ccus to evade host mucosal immunoglobulin which explains its substantial diversity as it is required to cleave structurally diverse substrates (Poulsen et al., 1998).

ber and sequence of

Page 7: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: repeat regions to facilitate (Poulsen microarray DNA CGH experiments where failure to hybridize indicates substantial divergence from the probe on the microarray (Hakenbeck et al., 2001). The Zinc metalloproteinase C (ZmpC) (Oggioni et al., 2003). Reports indicate the absence of found in 26% of pneumococcal isolates from patients with pneumonia (Chiavolini not detected in isolates from nasal or conjunctival swab (Oggioni that ZmpC is associated with pneumococcal virulence and pathogenicity in the lung Two-component signal transduction systems The two-components signal transduction systems (TCS) consists of a membrane associated sensor, histidine kinase (HK) which on receipt of a specific stimulus phosphorylates an aspartate residue in a cytoplasmic cognate response regulator (RR) which invariabal., 2006). It regulates various cellular processes in bacteria such as osmoregulation, sporulation, genetic competence and chemotaxis. The TCS allow pneumococci to respond to changes inet al., 2006). Currently, thirteen TCS and one orphan response regulator have been found in the pneumococcus (Paterson et al., 2006). The most studied TCS in an important virulence factor. ComD is associated with pneumonia and bacteriaemia in serotype 2 strain D39, serotype 3 strain 0100993 and serotype 4 strain TIGR4 (Paterson contribution of TCS to virulence varies depending microarray analysis have given new insights into gene regulation of TCS, the understanding of their interaction remain largely unclear. Pneumococcal pilus A pneumococcal pilus (PI-1 and PI-2) is encofor three pilus subunits (RrgA, RrgB and RrgC) and can be found in some but not all pneumococcal isolates (Paterson and Mitchell, 2006) where it has a role in adherence and may be involved al., 2006; Gianfaldoni et al., 2007). PIdocumented as absent from serotypes 1, 7F, 8 and 12B and its presence appears to be a clonal property (Moschioni et al., 2008) where it is particularly associated with ST156 and ST162 (Sjostrom contrast, Turner et al., (2011) reported that PIserotypes, including 4, 6A, 6B, 9, 14, 19F, 19A, 23F,common in in 19F (91%) and 23F (77%). In addition, Moschioni media clinical isolates from Israel detected PI1 in 6A, 6B, 14, 19F, 23F, 33A and 11A, whereas PI2010). Pillus antigens should be properly evaluated as they are currently regarded as potential candidate for inclusion in a protein-based pneumococcal vaccine. Pyruvate oxidase (SpxB) SpxB encodes a pyruvate oxidase that catalyses the reduction of pyruvate, free phosphate and oxygen to form carbon dioxide, acetylphosphate and hydrogen peroxide (Johnston major virulence factor in S. pneumoniae (Spellerberg highly expressed in environments rich in both oxygen and carbon (IV) oxide, conditions which are perculiar to the nasopharynx. The formation of hydrogen peroxide inhibits the growth of competing microflora, including Haemophilus influenzae, Neisseria meningitides and Moraxella catarhalis, and may also enhance pneumococcal adherence to nasopharyngeal epithelium and progression to th2004). In addition, production of hydrogen peroxide during infection promotes inflammation and cell damage, and induces TLR-2 and TLR-4 independent apoptosis in the brain epithelial cells. This results to nedamage and brain cell mitochondrial damage, and SpxB is implicated in pneumococcal meningitis. Furthermore, SpxB also enhances pneumococcal resistance to hydrogen peroxide and this allows it to grow in high hydrogen peroxide concentrations than many LeMessurier et al., (2006) demonstrated that brain in WCH16 strain intraperitoneal challenge in a mouse model. The presence of SpxB might be the cause of increased nasopharyngeal carriage of Streptococcus pneumoniae

is licensed under a Creative Commons Attribution 3.0 Unported License

33

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

repeat regions to facilitate (Poulsen et al., 1998). This high level of diversity has been demonstrated in microarray DNA CGH experiments where failure to hybridize indicates substantial divergence from the probe

., 2001).

The Zinc metalloproteinase C (ZmpC) specifically cleaves human matrix metalloproteinase 9 (MMP., 2003). Reports indicate the absence of ZmpC in R6 genome (Chiavolini et al

found in 26% of pneumococcal isolates from patients with pneumonia (Chiavolini et al., 2003) whereas it was not detected in isolates from nasal or conjunctival swab (Oggioni et al., 2003). Oggioni et al., (2003) proposed

is associated with pneumococcal virulence and pathogenicity in the lung

component signal transduction systems components signal transduction systems (TCS) consists of a membrane associated sensor, histidine

kinase (HK) which on receipt of a specific stimulus phosphorylates an aspartate residue in a cytoplasmic cognate response regulator (RR) which invariably results in alteration of levels of gene transcription (Paterson

., 2006). It regulates various cellular processes in bacteria such as osmoregulation, sporulation, genetic competence and chemotaxis. The TCS allow pneumococci to respond to changes in their environment (Paterson

., 2006). Currently, thirteen TCS and one orphan response regulator have been found in the pneumococcus ., 2006). The most studied TCS in S. pneumoniae is the competence gene (comDE

nt virulence factor. ComD is associated with pneumonia and bacteriaemia in serotype 2 strain D39, serotype 3 strain 0100993 and serotype 4 strain TIGR4 (Paterson et al., 2006; Hendriksen et alcontribution of TCS to virulence varies depending on S. pneumoniae strain and site of infection. Although microarray analysis have given new insights into gene regulation of TCS, the understanding of their interaction

2) is encoded by the rlrA islet (LeMieux et al., 2006), which includes genes for three pilus subunits (RrgA, RrgB and RrgC) and can be found in some but not all pneumococcal isolates (Paterson and Mitchell, 2006) where it has a role in adherence and may be involved in virulence (Barocchi

., 2007). PI-1 is associated with serotypes 4, 6B, 9V and 14 while it has been documented as absent from serotypes 1, 7F, 8 and 12B and its presence appears to be a clonal property

08) where it is particularly associated with ST156 and ST162 (Sjostrom ., (2011) reported that PI-1 was present in 35.2 % of pneumococcal isolates occurring in 10

serotypes, including 4, 6A, 6B, 9, 14, 19F, 19A, 23F, 33C and non-typeable pneumococci (NT) and PIcommon in in 19F (91%) and 23F (77%). In addition, Moschioni et al., (2010) in a research involving otitis media clinical isolates from Israel detected PI-1 in 30.1 % and PI-2 in 7 % of the isolates tested. They found PI1 in 6A, 6B, 14, 19F, 23F, 33A and 11A, whereas PI-2 was detected in serotype 19F only (Moschoioni 2010). Pillus antigens should be properly evaluated as they are currently regarded as potential candidate for

based pneumococcal vaccine.

encodes a pyruvate oxidase that catalyses the reduction of pyruvate, free phosphate and oxygen to form carbon dioxide, acetylphosphate and hydrogen peroxide (Johnston et al., 2004). SpxB has been dmajor virulence factor in S. pneumoniae (Spellerberg et al., 1996). Pericone et al., (2000) reported that highly expressed in environments rich in both oxygen and carbon (IV) oxide, conditions which are perculiar to

he formation of hydrogen peroxide inhibits the growth of competing microflora, including Haemophilus influenzae, Neisseria meningitides and Moraxella catarhalis, and may also enhance pneumococcal adherence to nasopharyngeal epithelium and progression to the lungs (Pericone et al., 2000; Orihuela 2004). In addition, production of hydrogen peroxide during infection promotes inflammation and cell damage,

4 independent apoptosis in the brain epithelial cells. This results to nedamage and brain cell mitochondrial damage, and SpxB is implicated in pneumococcal meningitis. Furthermore, SpxB also enhances pneumococcal resistance to hydrogen peroxide and this allows it to grow in high hydrogen

other bacteria colonizing the nasopharynx (Pericone ., (2006) demonstrated that SpxB gene is found in high concentration in the nasopharynx and

brain in WCH16 strain intraperitoneal challenge in a mouse model. The presence of SpxB might be the cause of Streptococcus pneumoniae globally.

Creative Commons Attribution 3.0 Unported License

., 1998). This high level of diversity has been demonstrated in microarray DNA CGH experiments where failure to hybridize indicates substantial divergence from the probe

fically cleaves human matrix metalloproteinase 9 (MMP-9) et al., 2003). It was

) whereas it was ., (2003) proposed

components signal transduction systems (TCS) consists of a membrane associated sensor, histidine kinase (HK) which on receipt of a specific stimulus phosphorylates an aspartate residue in a cytoplasmic

ly results in alteration of levels of gene transcription (Paterson et ., 2006). It regulates various cellular processes in bacteria such as osmoregulation, sporulation, genetic

their environment (Paterson ., 2006). Currently, thirteen TCS and one orphan response regulator have been found in the pneumococcus

comDE) system and is nt virulence factor. ComD is associated with pneumonia and bacteriaemia in serotype 2 strain D39,

et al., 2007). The strain and site of infection. Although

microarray analysis have given new insights into gene regulation of TCS, the understanding of their interaction

., 2006), which includes genes for three pilus subunits (RrgA, RrgB and RrgC) and can be found in some but not all pneumococcal isolates

in virulence (Barocchi et 1 is associated with serotypes 4, 6B, 9V and 14 while it has been

documented as absent from serotypes 1, 7F, 8 and 12B and its presence appears to be a clonal property 08) where it is particularly associated with ST156 and ST162 (Sjostrom et al., 2007). In

1 was present in 35.2 % of pneumococcal isolates occurring in 10 typeable pneumococci (NT) and PI-1 was

., (2010) in a research involving otitis d. They found PI-

2 was detected in serotype 19F only (Moschoioni et al., 2010). Pillus antigens should be properly evaluated as they are currently regarded as potential candidate for

encodes a pyruvate oxidase that catalyses the reduction of pyruvate, free phosphate and oxygen to form ., 2004). SpxB has been described as a

., (2000) reported that SpxB is highly expressed in environments rich in both oxygen and carbon (IV) oxide, conditions which are perculiar to

he formation of hydrogen peroxide inhibits the growth of competing microflora, including Haemophilus influenzae, Neisseria meningitides and Moraxella catarhalis, and may also enhance pneumococcal

., 2000; Orihuela et al., 2004). In addition, production of hydrogen peroxide during infection promotes inflammation and cell damage,

4 independent apoptosis in the brain epithelial cells. This results to neuronal damage and brain cell mitochondrial damage, and SpxB is implicated in pneumococcal meningitis. Furthermore, SpxB also enhances pneumococcal resistance to hydrogen peroxide and this allows it to grow in high hydrogen

other bacteria colonizing the nasopharynx (Pericone et al., 2003). gene is found in high concentration in the nasopharynx and

brain in WCH16 strain intraperitoneal challenge in a mouse model. The presence of SpxB might be the cause of

Page 8: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: Pneumococcal surface antigen A Pneumococcal surface antigen (PsaA) is the third gene in the lipoprotein receptor-associated antigen family (Johnston bacteria involved in in the transport of Mnhave shown the presence of PsaA gene in all the 90 pneumococcal serotypes using PCR technique (Morrison al., 2000). Manganese acts as a cofactor for bacterial proteins involved in glycolysis, gluconeogenesis, sugar and amino acid metabolism, peptide cleavage, nucleic acid degradation, signal transduction, and oxidative stress defence, and has been shown to contribute to bacterial survival and growth during infection (Jedrzejas, 2004). In addition, Mutants that lack PsaA failed to establish coin vivo models, which may be as a result of low concentrations of Mn2+ in these locations (McAllister 2004). PsaA gene was found in high level the blood of a mice model challenged with D39 been suggested to be involved in pneumococcal septicaemia (LeMessurier also found in Streptococcus mitis, Streptococcus oralisfunction in these bacteria is still unclear. Intranasal immunisation of mice with PsaA has been reported to protect against pneumococcal carriage in the nasopharynx (Briles cholera toxin B subunit-PsaA fusion protein protected mice against pneumococcal colonisation after intranasal immunisation (Pimenta et al., 2006). These studies indicate that PsaA is a potential musal vaccine candidate. Other virulence factors PsaR has been shown to contribute to the survival of pneumococci during bacteraemia. PsaR is mediate its activity through Psa operon, PcpA, and has been reported to be responsible for the regulation of Manganese which is physiology and virulence (Hendriksen et alvirulence of a serotype 3 in bacteraemia model of infection after 24 hours of infection. Similarly, PsaR does not contribute to nasopharyngeal colonisation, but it is involved in invasive disease where it has strainimpact during both pneumonia and bacteraemia (Hendriksen activated by codY and is required for adnutritional regulation and adherence (Hendriksen pathogenesis remains unclear. GlnA gene, a glutamate synthase, catalyses the gene is the major glutamine/glutamate ABC transporter. Recently, it was described that are involved in pneumococcal adhesion to human nasopharyngeal cells, which is a prehost (Kloosterman et al., 2006). In a mouse model, GlnA was shown to be associated with colonisation and GlnP for bacterial survival in the lungs, and a glnA2008b). In addition, glnP double mutant was unable to reach the systemic circulation in a mouse model, indicating that glnP is involved the dissemination of possible that a vaccine against glnP may be able to eliminate pnoligopeptide ABC transporter, Ami/Ali permease is associated with adherence, through direct interaction with human receptors or upregulating pneumococcal adhesins (Cundell al., 2007; Hendriksen et al., 2008b). It is essential for a successful nasopharyngeal colonisation in a mice model, and was not involved in invasive disease. Autolysins are also implicated in pneumococcal virulence. They are group of enzymes capable of dbacterial peptidoglycan leading to cell lysis. Several autolysins have been identified in autolysin A (LytA), autolysin B (LytB) and Autolysin C, LytC (Garcia LytA is the major pneumococcal autolysin and causes the autolysis of bacterial cell wall peptidoglycan, and also mediate the release of pneumolysin (Martner factor A (PavA), a fibronectin adhesion have been demonstraand adherence to nasopharyngeal cells, invasion, and Modulation of meningeal inflammation, but its mechanism of action still remain unclear (Homes further understanding of these factors will help to clarify pneumococcal factors which favour adherence.

is licensed under a Creative Commons Attribution 3.0 Unported License

34

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

) is the third gene in the psa operon, a lipoprotein which is related to the associated antigen family (Johnston et al., 2004). It is part of the transport system in

bacteria involved in in the transport of Mn2+ and Zn2+ into the cell (Jedrzejas, 2004). Morrison andgene in all the 90 pneumococcal serotypes using PCR technique (Morrison

., 2000). Manganese acts as a cofactor for bacterial proteins involved in glycolysis, gluconeogenesis, sugar peptide cleavage, nucleic acid degradation, signal transduction, and oxidative stress

defence, and has been shown to contribute to bacterial survival and growth during infection (Jedrzejas, 2004). In addition, Mutants that lack PsaA failed to establish colonisation, cause pneumonia, bateraemia or otitis media in

models, which may be as a result of low concentrations of Mn2+ in these locations (McAllister gene was found in high level the blood of a mice model challenged with D39 and WCH16, and has

been suggested to be involved in pneumococcal septicaemia (LeMessurier et al., 2006). However, Streptococcus oralis and Streptococcus anginosus (Jado et al

function in these bacteria is still unclear. Intranasal immunisation of mice with PsaA has been reported to protect against pneumococcal carriage in the nasopharynx (Briles et al., 2000; Johnson et al

PsaA fusion protein protected mice against pneumococcal colonisation after intranasal ., 2006). These studies indicate that PsaA is a potential musal vaccine candidate.

to the survival of pneumococci during bacteraemia. PsaR is mediate its , and PrtA and is majorly found in TIGR4 and D39 pneumococcal strains. It

has been reported to be responsible for the regulation of Manganese which is required for pneumococcal et al., 2009). Lau et al., (2001) demonstrated that PsaR is required for full

virulence of a serotype 3 in bacteraemia model of infection after 24 hours of infection. Similarly, PsaR does not ontribute to nasopharyngeal colonisation, but it is involved in invasive disease where it has strain

impact during both pneumonia and bacteraemia (Hendriksen et al., 2009). Choline binding protein (PcaA) is activated by codY and is required for adherence to nasopharyngeal cells, indicating a relationship between nutritional regulation and adherence (Hendriksen et al., 2008a), but the exact role of of PcaA in pneumococcal

gene, a glutamate synthase, catalyses the formation of glutamine from glutamate and ammonia, and gene is the major glutamine/glutamate ABC transporter. Recently, it was described that GlnP are involved in pneumococcal adhesion to human nasopharyngeal cells, which is a pre-requisite to invade the

., 2006). In a mouse model, GlnA was shown to be associated with colonisation and GlnP for bacterial survival in the lungs, and a glnA-glnP double mutant lost its virulence (Hendriksen

glnP double mutant was unable to reach the systemic circulation in a mouse model, indicating that glnP is involved the dissemination of S. pneumoniae from the lungs to the blood stream. It is possible that a vaccine against glnP may be able to eliminate pneumococcal septicaemia Furthermore, the oligopeptide ABC transporter, Ami/Ali permease is associated with adherence, through direct interaction with human receptors or upregulating pneumococcal adhesins (Cundell et al., 1995; Kerr et al., 2004; Anderton

., 2008b). It is essential for a successful nasopharyngeal colonisation in a mice model, and was not involved in invasive disease.

Autolysins are also implicated in pneumococcal virulence. They are group of enzymes capable of dbacterial peptidoglycan leading to cell lysis. Several autolysins have been identified in S. pneumoniaeautolysin A (LytA), autolysin B (LytB) and Autolysin C, LytC (Garcia et al., 1999; Hendriksen

ococcal autolysin and causes the autolysis of bacterial cell wall peptidoglycan, and also mediate the release of pneumolysin (Martner et al., 2008). Similarly, pneumococcal adherence and virulence factor A (PavA), a fibronectin adhesion have been demonstrated to be involved in pneumococcal colonization and adherence to nasopharyngeal cells, invasion, and Modulation of meningeal inflammation, but its mechanism of action still remain unclear (Homes et al., 2001; Pracht et al., 2005; Rajam et al., 2008). It isfurther understanding of these factors will help to clarify pneumococcal factors which favour adherence.

Creative Commons Attribution 3.0 Unported License

operon, a lipoprotein which is related to the ., 2004). It is part of the transport system in

into the cell (Jedrzejas, 2004). Morrison and colleagues gene in all the 90 pneumococcal serotypes using PCR technique (Morrison et

., 2000). Manganese acts as a cofactor for bacterial proteins involved in glycolysis, gluconeogenesis, sugar peptide cleavage, nucleic acid degradation, signal transduction, and oxidative stress

defence, and has been shown to contribute to bacterial survival and growth during infection (Jedrzejas, 2004). In lonisation, cause pneumonia, bateraemia or otitis media in

models, which may be as a result of low concentrations of Mn2+ in these locations (McAllister et al., and WCH16, and has

., 2006). However, PsaA gene is et al., 2001), but its

function in these bacteria is still unclear. Intranasal immunisation of mice with PsaA has been reported to et al., 2002) and a

PsaA fusion protein protected mice against pneumococcal colonisation after intranasal ., 2006). These studies indicate that PsaA is a potential musal vaccine candidate.

to the survival of pneumococci during bacteraemia. PsaR is mediate its and is majorly found in TIGR4 and D39 pneumococcal strains. It

required for pneumococcal ., (2001) demonstrated that PsaR is required for full

virulence of a serotype 3 in bacteraemia model of infection after 24 hours of infection. Similarly, PsaR does not ontribute to nasopharyngeal colonisation, but it is involved in invasive disease where it has strain-specific

., 2009). Choline binding protein (PcaA) is herence to nasopharyngeal cells, indicating a relationship between

., 2008a), but the exact role of of PcaA in pneumococcal

formation of glutamine from glutamate and ammonia, and GlnP and GlnA genes site to invade the

., 2006). In a mouse model, GlnA was shown to be associated with colonisation and glnP double mutant lost its virulence (Hendriksen et al.,

glnP double mutant was unable to reach the systemic circulation in a mouse model, from the lungs to the blood stream. It is

eumococcal septicaemia Furthermore, the oligopeptide ABC transporter, Ami/Ali permease is associated with adherence, through direct interaction with

., 2004; Anderton et ., 2008b). It is essential for a successful nasopharyngeal colonisation in a mice model,

Autolysins are also implicated in pneumococcal virulence. They are group of enzymes capable of degrading S. pneumoniae including,

., 1999; Hendriksen et al., 2009). ococcal autolysin and causes the autolysis of bacterial cell wall peptidoglycan, and also

., 2008). Similarly, pneumococcal adherence and virulence ted to be involved in pneumococcal colonization

and adherence to nasopharyngeal cells, invasion, and Modulation of meningeal inflammation, but its mechanism ., 2008). It is hoped that

further understanding of these factors will help to clarify pneumococcal factors which favour adherence.

Page 9: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,:

Figure 1.2: Interaction between S pneumoniae viscosity of the mucus and exposes the Ncan interact with pneumococcal surfacehost epithelial cells upregulate the plateletaffinity via its cell-wall phosphocholine (ChoP) for PAFr. Moreover, a second cholineshows increased affinity for immobilised sialic acid and lactoIg receptor (pIgR), which increases migration through the mucosal barrier (transcytosis). Pneumococcal IgA1protease cleaves opsonising IgA, which results ithe physical proximity of ChoP to the PAFr. Adapted from:

Figure 1.3: A representsation of the cell membrane, cell wall, and capsule of (Musher, 2005) CONCLUSION A thorough understanding of pneumococcal components necessary for pathogenicity and virulence is required to combat pneumococcal disease. Until recently, the capsule has been the major focus of pneumococcal vaccine. However, recent advances in the understanding of the structure and function of various pathogenicity and virulence factors have paved way for the possibility of developing protein vaccines. The introduction of proteinbased pneumococcal vaccine comprising surface

is licensed under a Creative Commons Attribution 3.0 Unported License

35

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

S pneumoniae and epithelial cells. Neuraminidase (NanA) decreases the

viscosity of the mucus and exposes the N-acetyl-glycosamine (GlcNAc) receptors on the epithelial cells, which can interact with pneumococcal surface-associated proteins such as PsaA. In response to cytokine stimulation, host epithelial cells upregulate the platelet-activating-factor receptors (PAFr). The pneumococcus has increased

wall phosphocholine (ChoP) for PAFr. Moreover, a second choline-binding protein, CbpA, ed affinity for immobilised sialic acid and lacto-N-neotreatose, and binds directly to the polymeric

Ig receptor (pIgR), which increases migration through the mucosal barrier (transcytosis). Pneumococcal IgA1protease cleaves opsonising IgA, which results in a change (neutralisation) of surface charge and increases the physical proximity of ChoP to the PAFr. Adapted from: (Bogaert et al., 2004).

Figure 1.3: A representsation of the cell membrane, cell wall, and capsule of S.pneumoniae. Adapted from:

A thorough understanding of pneumococcal components necessary for pathogenicity and virulence is required to combat pneumococcal disease. Until recently, the capsule has been the major focus of pneumococcal vaccine.

nt advances in the understanding of the structure and function of various pathogenicity and virulence factors have paved way for the possibility of developing protein vaccines. The introduction of proteinbased pneumococcal vaccine comprising surface-exposed proteins associated with nasopharyngeal colonisation

Creative Commons Attribution 3.0 Unported License

and epithelial cells. Neuraminidase (NanA) decreases the glycosamine (GlcNAc) receptors on the epithelial cells, which

ytokine stimulation, factor receptors (PAFr). The pneumococcus has increased

binding protein, CbpA, neotreatose, and binds directly to the polymeric

Ig receptor (pIgR), which increases migration through the mucosal barrier (transcytosis). Pneumococcal n a change (neutralisation) of surface charge and increases

Adapted from:

A thorough understanding of pneumococcal components necessary for pathogenicity and virulence is required to combat pneumococcal disease. Until recently, the capsule has been the major focus of pneumococcal vaccine.

nt advances in the understanding of the structure and function of various pathogenicity and virulence factors have paved way for the possibility of developing protein vaccines. The introduction of protein-

ed proteins associated with nasopharyngeal colonisation

Page 10: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: and adherence such as neuraminidase, PsaA, and CbpA will increase vaccine coverage as these pneumococcal virulence proteins appear to be conserved in most serotypes. REFERENCES Anderton, J. M. G., Rajam, G., RomeroS. and Ades, E. W. (2007) E-cadherin is a receptor for the common protein pneumococcal surface adhesion A (PsaA) of Streptococcus pneumoniae. Microbiology and Pathology Balachandran, P., Brooks-Walter, A., ViRole of pneumococcal surface protein C in nasopharyngeal carriage and pneumonia and its ability to elicit protection against Streptococcus pneumoniae Balachandran, P., Hollingshead, S. K., Paton, J. C. and Briles, D. E. (2001) The autolytic enzyme LytA of Streptococcus pneumoniae is not responsible for releasing pneumolysin. 3116. Barocchi, M. A., Ries, J., Zogaj, X., Hemsley, C., Albiger, B., Kanth, A., Dahlberg, S., Fernebro, J., Moschioni, M., Masignani, V., Hultenby, K., Taddei, A. R., Beiter, K., Wartha, F., Von Euler, A., Covacci, A., Holden, D. W., Normark, S., Rappuoli, R. and Henriquesand host inflammatory responses. Proceedings of the National Academy of Sciences of the United States of America.103, pp. 2857-2862. Benton, K.A., Everson, M. P. and Briles, D. E. (1995) A pneumolysinpneumoniae causes chronic bacteraemia rather than sepsis in mice. Berry, A. M. and Paton, J. C. (1996) Sequence heterogeneity of PsaA, a 37essential for virulence of Streptococcus pneumoniae Berry, A. M., Ogunniyi, A. D., Miller, D.C. and Paton, J. C. (1999) Comparative virulence of pneumoniae strains with insertion-duplication, point, and detection mand Immunity. 67, pp. 981-985. Berry, A. M., Paton, J. C. and Hansman, D. (1992) Effect of insertional inactivation of the genes encoding pneumolysin and autolysin on the virulence of pp. 87-93. Bogaert, D, Groot De, R. and Hermans, P. W. M. (2004) pneumococcal disease. Lancet Infectious Disease Boulnois, G. J., Paton, J. C., Mitchell, T., J. and Andrew, P. W. (1991) Structure and function of pneumolysin, the multifunctional thiol-activated toxin of 2616. Braun, J. S., Sublett, J. E., Freyer, D., Mitchell, T. J., Cleveland, J. L., Tuomanen, E. I. and Weber, J. R. (2000) Pneumococcal pneumolysin and H2O2

Investigation. 190, pp. 19-27. Braun, J., Novak, R., Gao, G., Murray, P. and Shenep, J. (1999) Pneumolysin, a protein toxin of pneumoniae, induces nitric oxide production from macrophages. Briles, D. E., Ades, E., Paton, J. C., Sampson, J. S., Carlone, G. M., Hueband Hollingshead, S. K. (2000) Intranasal immunisation of mice with a mixture of the pneumococcal proteins PsaA and PsaP is highly protective against nasopharyngeal carriage of Immunity. 68, pp. 796-800.

is licensed under a Creative Commons Attribution 3.0 Unported License

36

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

adherence such as neuraminidase, PsaA, and CbpA will increase vaccine coverage as these pneumococcal virulence proteins appear to be conserved in most serotypes.

Anderton, J. M. G., Rajam, G., Romero-Steiner, S., Summer, S., Kowalczyk, A. P., Carlone, G. M., Sampson, J. cadherin is a receptor for the common protein pneumococcal surface adhesion A

Microbiology and Pathology. 42, pp. 225-236.

Walter, A., Virolainen-Julkunen, A., Hollingshead, S. K. and Briles, D. E. (2002) Role of pneumococcal surface protein C in nasopharyngeal carriage and pneumonia and its ability to elicit

Streptococcus pneumoniae. Infection and Immunity. 70, pp. 2526-2534.

Balachandran, P., Hollingshead, S. K., Paton, J. C. and Briles, D. E. (2001) The autolytic enzyme LytA of is not responsible for releasing pneumolysin. Journal of Bacteriology

Zogaj, X., Hemsley, C., Albiger, B., Kanth, A., Dahlberg, S., Fernebro, J., Moschioni, M., Masignani, V., Hultenby, K., Taddei, A. R., Beiter, K., Wartha, F., Von Euler, A., Covacci, A., Holden, D. W., Normark, S., Rappuoli, R. and Henriques-Normark, B. (2006) A pneumococcal pilus influences virulence

Proceedings of the National Academy of Sciences of the United States of

Benton, K.A., Everson, M. P. and Briles, D. E. (1995) A pneumolysin-negative mutant of causes chronic bacteraemia rather than sepsis in mice. Infection and Immunity. 63, pp. 448

Berry, A. M. and Paton, J. C. (1996) Sequence heterogeneity of PsaA, a 37-kilodalton putative adhesion Streptococcus pneumoniae. Infection and Immunity. 60, pp. 5255-5262.

Berry, A. M., Ogunniyi, A. D., Miller, D.C. and Paton, J. C. (1999) Comparative virulence of duplication, point, and detection mutations in pneumolysin gene.

Berry, A. M., Paton, J. C. and Hansman, D. (1992) Effect of insertional inactivation of the genes encoding pneumolysin and autolysin on the virulence of Streptococcus pneumoniae type 3. Microbial Pathogenesis

Bogaert, D, Groot De, R. and Hermans, P. W. M. (2004) Streptococcus pneumoniae colonisation: the key to Lancet Infectious Disease. 4, pp. 144-154.

Boulnois, G. J., Paton, J. C., Mitchell, T., J. and Andrew, P. W. (1991) Structure and function of pneumolysin, activated toxin of Streptococcus pneumoniae.Molecular Microbiology

reyer, D., Mitchell, T. J., Cleveland, J. L., Tuomanen, E. I. and Weber, J. R. (2000) 2 mediate brain cell apaoptosis during meningitis. Journal of Clinical

ay, P. and Shenep, J. (1999) Pneumolysin, a protein toxin of , induces nitric oxide production from macrophages. Infection and Immunity. 67, pp. 3750

Briles, D. E., Ades, E., Paton, J. C., Sampson, J. S., Carlone, G. M., Huebner, R. C., Virolainen, A., Swiatlo, E. and Hollingshead, S. K. (2000) Intranasal immunisation of mice with a mixture of the pneumococcal proteins PsaA and PsaP is highly protective against nasopharyngeal carriage of Streptococcus pneumoniae

Creative Commons Attribution 3.0 Unported License

adherence such as neuraminidase, PsaA, and CbpA will increase vaccine coverage as these pneumococcal

Carlone, G. M., Sampson, J. cadherin is a receptor for the common protein pneumococcal surface adhesion A

Julkunen, A., Hollingshead, S. K. and Briles, D. E. (2002) Role of pneumococcal surface protein C in nasopharyngeal carriage and pneumonia and its ability to elicit

Balachandran, P., Hollingshead, S. K., Paton, J. C. and Briles, D. E. (2001) The autolytic enzyme LytA of Journal of Bacteriology. 183, pp. 3108-

Zogaj, X., Hemsley, C., Albiger, B., Kanth, A., Dahlberg, S., Fernebro, J., Moschioni, M., Masignani, V., Hultenby, K., Taddei, A. R., Beiter, K., Wartha, F., Von Euler, A., Covacci, A., Holden, D.

2006) A pneumococcal pilus influences virulence Proceedings of the National Academy of Sciences of the United States of

ve mutant of Streptococcus . 63, pp. 448-455.

kilodalton putative adhesion 5262.

Berry, A. M., Ogunniyi, A. D., Miller, D.C. and Paton, J. C. (1999) Comparative virulence of Streptococcus utations in pneumolysin gene. Infection

Berry, A. M., Paton, J. C. and Hansman, D. (1992) Effect of insertional inactivation of the genes encoding Microbial Pathogenesis. 12,

colonisation: the key to

Boulnois, G. J., Paton, J. C., Mitchell, T., J. and Andrew, P. W. (1991) Structure and function of pneumolysin, Molecular Microbiology. 2, pp. 2611-

reyer, D., Mitchell, T. J., Cleveland, J. L., Tuomanen, E. I. and Weber, J. R. (2000) Journal of Clinical

ay, P. and Shenep, J. (1999) Pneumolysin, a protein toxin of Streptococcus . 67, pp. 3750-3756.

ner, R. C., Virolainen, A., Swiatlo, E. and Hollingshead, S. K. (2000) Intranasal immunisation of mice with a mixture of the pneumococcal proteins

Streptococcus pneumoniae. Infection and

Page 11: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: Briles, D. E., King, J. D., Gray, M. A., McDaniel, L. S., Swiatlo, E. and Benton, K. A. (1996) PspA, a proteineliciting pneumococcal protein: immunogeni Briles, D. E., Tart, R. C., Swiatlo, E., Dillard, J. P., Smith, P., Benton, K. A., Ralph, B. A., BrooksCrain, M. J., Hollingshead, S. K. and McDaniels, L. S. (1998) Pneumococcal diversity: considerations for new vaccine strategies with emphasis on pneumococcal surface protein A (PspA). pp. 645-657. Brocks, S. C., McGraw, P. A., Wright, P. F. and Crowe, J. E. J. (2002) The human polymeric immunoglobulin receptor facilitates invasion of epithelial cells by manner. Infection and Immunity. 70, pp. 5091 Brooks, W. A., Briles, D. E. and Hollingshead, S. K. (1999) Theencodes a polymorphic protein, PspC, which elicipneumococcal bacteraemia. Infection and Immunity Brown, J. S., Gillliland, S. M. and Holden, D. W. (2001) A encoding an ABC transporter involved in iron uptake and virulence. Brueggemann, A. B. and Spratt, B. G. (2003) Geographic Distribution and Clonal Diversity of pneumoniae Serotype 1 Isolates. Journal of Clinical Centre for Disease Control and Prevention (1997) Prevention of pneumococcal diseases: Recommendations of the Advisory Committee on Immunisation Practices. (ACIP). Chiavolini, D., Memmi, G., Maggi, T., Iazinc metalloproteinases of Streptococcus pneumoniae Microbiology.3, pp. 14. Cockeran, R., Durandt, C., Feldman, C., Mitchell, T. J.,release of interleukin-8 by human neutrophils Coffey, T. J., Enright, M. C., Daniels, M., Morona, J. K., Morona, R., Hryniewicz, W., Paton, JB. G. (1998) Recombinational exchanges at the capsular polysaccharide biosynthetic locus lead to frequent serotype changes among natural isolates of Crain, M. J., Waltman, W.D 2nd, Turner, J. S., Yother, J., Talkington, D. F., McDaniel, L. S., Gray, B. M. and Briles, D. E. (1990) Pneumococcal surface protein A (PspA) is serologically highly variable and is expressed by all clinically important capsular serotypes of 3299. Cundell, D.R., Pearce, B. J., Sandros, J., Naughton, A. M. and Masure, H. R. (Streptococcus pneumoniae affects adherence to eukaryotic cells. Dave, S., Brooks-Walter, A., Pangburn, M. K. and McDaniel, L. S. (2001) PspC, a pneumococcal surface protein, binds human factor H. Infection and Immunity Eskola, J., Kilpi, T. and Palmu, A. (2001) Efficacy of a pnmedia.New England Journal of Medicine. Farrell, D. J., Klugman, K. P. and Pichichero, M. (2007) Increased Antimicrobial Resistance Among Nonvaccine Serotypes of Streptococcus pneumoniae Valent Pneumococcal Vaccine in the United States. 128.

is licensed under a Creative Commons Attribution 3.0 Unported License

37

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

Briles, D. E., King, J. D., Gray, M. A., McDaniel, L. S., Swiatlo, E. and Benton, K. A. (1996) PspA, a proteineliciting pneumococcal protein: immunogenicity of isolated native PspA in mice. Vaccine. 4, pp. 858

Briles, D. E., Tart, R. C., Swiatlo, E., Dillard, J. P., Smith, P., Benton, K. A., Ralph, B. A., BrooksCrain, M. J., Hollingshead, S. K. and McDaniels, L. S. (1998) Pneumococcal diversity: considerations for new

s on pneumococcal surface protein A (PspA). Clinical Microbiology Review

Brocks, S. C., McGraw, P. A., Wright, P. F. and Crowe, J. E. J. (2002) The human polymeric immunoglobulin receptor facilitates invasion of epithelial cells by Streptococcus pneumoniae in a strain-specific and cell type

. 70, pp. 5091-5095.

Brooks, W. A., Briles, D. E. and Hollingshead, S. K. (1999) ThepspC gene of Streptococcus pneumoniaeencodes a polymorphic protein, PspC, which elicits cross-reactive antibodies to PspA and provides immunity to

Infection and Immunity. 67, pp. 6533-6542.

Brown, J. S., Gillliland, S. M. and Holden, D. W. (2001) A Streptococcus pneumoniae pathogenicity island nsporter involved in iron uptake and virulence. Molecular Microbiology. 40, pp. 572

Brueggemann, A. B. and Spratt, B. G. (2003) Geographic Distribution and Clonal Diversity of Journal of Clinical Microbiology.41, pp. 4966-4970.

Centre for Disease Control and Prevention (1997) Prevention of pneumococcal diseases: Recommendations of the Advisory Committee on Immunisation Practices. (ACIP).MMWR. 46, pp. 1-24.

Chiavolini, D., Memmi, G., Maggi, T., Iannelli, F., Pozzi, G. and Oggioni, M. R. (2003) The three extraStreptococcus pneumoniae have a different impact on virulence in mice.

Cockeran, R., Durandt, C., Feldman, C., Mitchell, T. J., R, A. (2002) Pneumolysin activates the synthesis and 8 by human neutrophils in vitro. Journal of Infectious Diseases. 186, pp. 562

Coffey, T. J., Enright, M. C., Daniels, M., Morona, J. K., Morona, R., Hryniewicz, W., Paton, JB. G. (1998) Recombinational exchanges at the capsular polysaccharide biosynthetic locus lead to frequent serotype changes among natural isolates of Streptococcus pneumoniae. Molecular Microbiology. 27, pp. 73

, Turner, J. S., Yother, J., Talkington, D. F., McDaniel, L. S., Gray, B. M. and Briles, D. E. (1990) Pneumococcal surface protein A (PspA) is serologically highly variable and is expressed by all clinically important capsular serotypes of Streptococcus pneumoniae. Infection and Immunity

Cundell, D.R., Pearce, B. J., Sandros, J., Naughton, A. M. and Masure, H. R. (1995) Peptide permeases from affects adherence to eukaryotic cells. Infection and Immunity. 63, pp. 2493

Walter, A., Pangburn, M. K. and McDaniel, L. S. (2001) PspC, a pneumococcal surface Infection and Immunity. 69, pp. 3435-3437.

Eskola, J., Kilpi, T. and Palmu, A. (2001) Efficacy of a pneumococcal conjugate vaccine against acute otitis New England Journal of Medicine.344, pp. 403-409.

Farrell, D. J., Klugman, K. P. and Pichichero, M. (2007) Increased Antimicrobial Resistance Among Streptococcus pneumoniae in the Pediatric Population After the Introduction of 7

Valent Pneumococcal Vaccine in the United States. Paediatric and Infectious Disease Journal

Creative Commons Attribution 3.0 Unported License

Briles, D. E., King, J. D., Gray, M. A., McDaniel, L. S., Swiatlo, E. and Benton, K. A. (1996) PspA, a protein-. 4, pp. 858-867.

Briles, D. E., Tart, R. C., Swiatlo, E., Dillard, J. P., Smith, P., Benton, K. A., Ralph, B. A., Brooks-Walter, A., Crain, M. J., Hollingshead, S. K. and McDaniels, L. S. (1998) Pneumococcal diversity: considerations for new

Clinical Microbiology Review. 11,

Brocks, S. C., McGraw, P. A., Wright, P. F. and Crowe, J. E. J. (2002) The human polymeric immunoglobulin specific and cell type

Streptococcus pneumoniae reactive antibodies to PspA and provides immunity to

pathogenicity island . 40, pp. 572-585.

Brueggemann, A. B. and Spratt, B. G. (2003) Geographic Distribution and Clonal Diversity of Streptococcus

Centre for Disease Control and Prevention (1997) Prevention of pneumococcal diseases: Recommendations of

nnelli, F., Pozzi, G. and Oggioni, M. R. (2003) The three extra-cellular have a different impact on virulence in mice. BMC

R, A. (2002) Pneumolysin activates the synthesis and . 186, pp. 562-565.

Coffey, T. J., Enright, M. C., Daniels, M., Morona, J. K., Morona, R., Hryniewicz, W., Paton, J. C. and Spratt, B. G. (1998) Recombinational exchanges at the capsular polysaccharide biosynthetic locus lead to frequent

. 27, pp. 73-83.

, Turner, J. S., Yother, J., Talkington, D. F., McDaniel, L. S., Gray, B. M. and Briles, D. E. (1990) Pneumococcal surface protein A (PspA) is serologically highly variable and is expressed by

Infection and Immunity. 58, pp. 3293-

1995) Peptide permeases from 63, pp. 2493-2498.

Walter, A., Pangburn, M. K. and McDaniel, L. S. (2001) PspC, a pneumococcal surface

eumococcal conjugate vaccine against acute otitis

Farrell, D. J., Klugman, K. P. and Pichichero, M. (2007) Increased Antimicrobial Resistance Among the Pediatric Population After the Introduction of 7-

Paediatric and Infectious Disease Journal.26(2), pp.123-

Page 12: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: Felmingham, D. (2002) Evolving resistance patterns in communityresults from the PROTEKT global surveillance study. ProspectiveResistant Organism Tracking and Epidemiology for the Ketolide Telithromycin. Ferreira, D. M., Miyaji, E. N., Oliveira, M. L. S., Darrieux, M., Areas, A. P. N., Ho, L. P. and Leite, L. C. C. (2006) DNA vaccines expressing pneumococcal surface protein A (PspA) elicit protection levels comparable to recombinant protein. Journal of Medical Microbiology Fry, A. M., Facklam, R. R., Whitney, C. G., Plikaytis, B. D. and Schuchat, A. (2003). Multistate evaluation of invasive pneumococcal diseases in adults with human immunodeficiency virus infection: serotype and antimicrobial resistance patterns in theGarcia, J. L., SanchezVersatility of choline-binding domains. Garcia, P., Gonzalez, M. P., Garcia, E., Lopez, R. and Garcia, J. (199hydrolase essential for cell separation. Molecular Microbiology Gianfaldoni, C., Censini, S., Hilleringmann, M., Moschioni, M., Facciotti, C., Pansegrau, W., Masignani, V., Covacci, A., Rappuoli, R., Barocchi, M. A. and Ruggiero, P. (2007) Protect Mice against Lethal Challenge. Glover, D. T, Hollingshead, S. K. and Briles, D. E. (2008) elicits protection against lung infection and fatal sepsis. Gosink, K. K., Mann, E. R., Gulielmo, C., Tuomanen, E. I. and Masure, H. R. (2000) Role of novel choline binding proteins in Streptococcus pneumoniae Greenwood, B. (1999). The epidemiology of pneumococcal infection in children in the developing world.Trans R Soc Lond B Biol Sci.354(1384), pp. 777 Hakenbeck, R., Balmelle, N., Weber, B., Gardes, C., Keck, W. and De Saizieu, A. (2001) Mosaic genes and mosaic chromosomes: Intra- and interspecies genomic variation of Immunity.69, pp. 2477-2486. Hammerschmidt, S. (2006) Adherence molecules ofMicrobiology. 9, pp. 12-20. Hammerschmidt, S., Taley, S. R., Brandtzaeg, P. and Chlatwal, G. S. (1997) SpsA, a novel pneumococcal surface protein with specific binding to secretory immunoglobulin A and secretoryMicrobiology. 25, pp. 1113-1124. Hausdorff, W. P., Bryant, J., Paradiso, P. R. and Siber, G. R. (2000). Which pneumococcal serogroups cause the most invasive disease: implications for conjugate vaccine formulation and use, part I. Disease. 30(1), pp. 100-121. Hausdorff, W. P., Feikin, D.R. and Klugman, K. P. (2005) Epidemiological differences among pneumococcal serotypes. Lancet Infectious Disease. 5, pp. 83 Hausdorff, W. P., Yothers, G., R., D., Kilpi, T., PLopez, E. L., Mason, E. O., Syriopoulou, V., Wynne, B. and Bryant, J. (2002) Multinational study of pneumococcal serotypes causing otitis media in children. 1016. Hausdorff, W. P., Yothers, G., R., D., Kilpi, T., Pelton, S., Cohen, R., Jacobs, M. R., Kaplan, S. L., Levy, C., Lopez, E. L., Mason, E. O., Syriopoulou, V., Wynne, B. and Bryant, J. (2002) Multinational study of

is licensed under a Creative Commons Attribution 3.0 Unported License

38

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

(2002) Evolving resistance patterns in community-acquired respiratorytract pathogens: first results from the PROTEKT global surveillance study. ProspectiveResistant Organism Tracking and Epidemiology for the Ketolide Telithromycin.Journal of Infection. 44(Suppl A), pp. 3-10.

Ferreira, D. M., Miyaji, E. N., Oliveira, M. L. S., Darrieux, M., Areas, A. P. N., Ho, L. P. and Leite, L. C. C. (2006) DNA vaccines expressing pneumococcal surface protein A (PspA) elicit protection levels comparable to

ical Microbiology. 55, pp. 375-378.

Fry, A. M., Facklam, R. R., Whitney, C. G., Plikaytis, B. D. and Schuchat, A. (2003). Multistate evaluation of invasive pneumococcal diseases in adults with human immunodeficiency virus infection: serotype and

bial resistance patterns in theGarcia, J. L., Sanchez-Beato, A. R., Medrano, F. J. and Lopez, R. (1998) binding domains. Microbial Drug Resistance. 4, pp. 25-36.

Garcia, P., Gonzalez, M. P., Garcia, E., Lopez, R. and Garcia, J. (1999) LytB, a novel pneumococcal murein Molecular Microbiology. 31, pp. 1275-1281.

Gianfaldoni, C., Censini, S., Hilleringmann, M., Moschioni, M., Facciotti, C., Pansegrau, W., Masignani, V., , Barocchi, M. A. and Ruggiero, P. (2007) Streptococcus pneumoniae Pilus Subunits

Protect Mice against Lethal Challenge. Infection and Immunity.75, pp. 1059-1062.

Glover, D. T, Hollingshead, S. K. and Briles, D. E. (2008) Streptococcus pneumoniae surfaceelicits protection against lung infection and fatal sepsis. Infection and Immunity. 76, pp. 2767-2776.

Gosink, K. K., Mann, E. R., Gulielmo, C., Tuomanen, E. I. and Masure, H. R. (2000) Role of novel choline pneumoniae virulence. Infection and Immunity. 68, pp. 5690-5695.

Greenwood, B. (1999). The epidemiology of pneumococcal infection in children in the developing world..354(1384), pp. 777-785.

Weber, B., Gardes, C., Keck, W. and De Saizieu, A. (2001) Mosaic genes and and interspecies genomic variation of Streptococcus pneumoniae

Hammerschmidt, S. (2006) Adherence molecules of pathogenic pneumococci. Current Opinion in

Hammerschmidt, S., Taley, S. R., Brandtzaeg, P. and Chlatwal, G. S. (1997) SpsA, a novel pneumococcal surface protein with specific binding to secretory immunoglobulin A and secretory component.

Hausdorff, W. P., Bryant, J., Paradiso, P. R. and Siber, G. R. (2000). Which pneumococcal serogroups cause the most invasive disease: implications for conjugate vaccine formulation and use, part I. Clinical Infectious

Hausdorff, W. P., Feikin, D.R. and Klugman, K. P. (2005) Epidemiological differences among pneumococcal . 5, pp. 83-93.

Hausdorff, W. P., Yothers, G., R., D., Kilpi, T., Pelton, S., Cohen, R., Jacobs, M. R., Kaplan, S. L., Levy, C., Lopez, E. L., Mason, E. O., Syriopoulou, V., Wynne, B. and Bryant, J. (2002) Multinational study of pneumococcal serotypes causing otitis media in children. Paediatric Infectious Disease Journal

Hausdorff, W. P., Yothers, G., R., D., Kilpi, T., Pelton, S., Cohen, R., Jacobs, M. R., Kaplan, S. L., Levy, C., Lopez, E. L., Mason, E. O., Syriopoulou, V., Wynne, B. and Bryant, J. (2002) Multinational study of

Creative Commons Attribution 3.0 Unported License

acquired respiratorytract pathogens: first results from the PROTEKT global surveillance study. ProspectiveResistant Organism Tracking and

Ferreira, D. M., Miyaji, E. N., Oliveira, M. L. S., Darrieux, M., Areas, A. P. N., Ho, L. P. and Leite, L. C. C. (2006) DNA vaccines expressing pneumococcal surface protein A (PspA) elicit protection levels comparable to

Fry, A. M., Facklam, R. R., Whitney, C. G., Plikaytis, B. D. and Schuchat, A. (2003). Multistate evaluation of invasive pneumococcal diseases in adults with human immunodeficiency virus infection: serotype and

Beato, A. R., Medrano, F. J. and Lopez, R. (1998)

9) LytB, a novel pneumococcal murein

Gianfaldoni, C., Censini, S., Hilleringmann, M., Moschioni, M., Facciotti, C., Pansegrau, W., Masignani, V., Pilus Subunits

surface protein PcpA 2776.

Gosink, K. K., Mann, E. R., Gulielmo, C., Tuomanen, E. I. and Masure, H. R. (2000) Role of novel choline 5695.

Greenwood, B. (1999). The epidemiology of pneumococcal infection in children in the developing world.Philos

Weber, B., Gardes, C., Keck, W. and De Saizieu, A. (2001) Mosaic genes and Streptococcus pneumoniae.Infection and

Current Opinion in

Hammerschmidt, S., Taley, S. R., Brandtzaeg, P. and Chlatwal, G. S. (1997) SpsA, a novel pneumococcal component. Molecular

Hausdorff, W. P., Bryant, J., Paradiso, P. R. and Siber, G. R. (2000). Which pneumococcal serogroups cause the inical Infectious

Hausdorff, W. P., Feikin, D.R. and Klugman, K. P. (2005) Epidemiological differences among pneumococcal

elton, S., Cohen, R., Jacobs, M. R., Kaplan, S. L., Levy, C., Lopez, E. L., Mason, E. O., Syriopoulou, V., Wynne, B. and Bryant, J. (2002) Multinational study of

Paediatric Infectious Disease Journal.21, pp. 1008-

Hausdorff, W. P., Yothers, G., R., D., Kilpi, T., Pelton, S., Cohen, R., Jacobs, M. R., Kaplan, S. L., Levy, C., Lopez, E. L., Mason, E. O., Syriopoulou, V., Wynne, B. and Bryant, J. (2002) Multinational study of

Page 13: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: pneumococcal serotypes causing otitis media in children. 1016. Hendriksen, W. T., Bootsma, H. J., Diepen van, A., Estevao, S., Kuipers, O. P., Groot dW. M. (2009) Strain-specific impact of PsaR ofvirulence. Microbiology. 155, pp. 1569- Hendriksen, W. T., Bootsma, H. J., Estevao, S., Hoogenboezem, T., Jong de, A., Groot and Hermans, P. W. M. (2008a) CodY of and colonization. Journal Bacteriology. Hendriksen, W. T., Kloosterman, T. G., Bootsma, H. J., Estavao, S., GrP. W. W. (2008b) Site-specific contributions of glutamateto virulence of Streptococcus pneumoniae Hendriksen, W. T., Kloosterman, T. G., Bootsma, H. J., Estevao, S., Groot De, R., O’Callaghan, C. and Andrew, P. W. (2004) The role of pneumolysin in pneumococcal pneumonia and meningitis. Clinical and Experimental Immunology Henrichsen, J. (1995) Six newly recognized types of Microbiology. 33(10), pp. 2759-62. Hollingshead, S. K., Becker, R. and Briles, D. E. (2000). Diversity of PspA: mosaic genes and evidence for past recombination in Streptococcus pneumoniae Hollingshead, S. K., Becker, R. and Briles, D. E. (2000) Diversity of PspA: Mosaic genes and evidence for past recombination in Streptococcus pneumoniae Homes, A. R., McNab, R., Millsap, K. W., Rhode, M., Hammerschmidt, S., Mawdley, J. L. and Jenkinson, H. F. (2001) The PavA gene of Streptococcus pneumoniaevirulence. Molecular Microbiology. 41, pp. 1395 Houldsworth, S., Andrew, P. W. and Mitchell, T. J. (1994) Pneumolysin stimulates production of tumour necrosis factor alpha and interleukin-1 beta by human mononuclear phagocytes. 1501-1503. Iannelli, F., Oggioni, M. R. and Pozzi, G. (2002) Allelic variation in the highly polymorphic locus Streptococcus pneumoniae.Gene. 284, pp. 63 INCLEN (1999). Prospective multicentre hospital surveillance of International Clinical Epidemiology Network (INCLEN). Jacobs, M. R. (2004). Streptococcus pneumoniaeMedicine.117(Suppl 3A), pp. 3S-15S. Jacobs, M. R., Felmingham, D., Appelbaum, P. C. and Gruneberg, R. N. (2003). The Alexander Project 19982000: susceptibility of pathogens isolated from community acquired respiratory tract infection to commonly used antimicrobial agents. Journal of Antimicrobial Chemotherapy Jarva, H., Jokiranta, T. S., Wurzner, R. and Meri, S. (2003) Complement resistance mechanisms of Streptococci. Molecular Immunology. 40, pp. 95-107. Jedrzejas, M. J. (2004) Extracellular virulence factors for pp. 891-914.

is licensed under a Creative Commons Attribution 3.0 Unported License

39

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

pneumococcal serotypes causing otitis media in children. Paediatric Infectious Disease Journal.

Hendriksen, W. T., Bootsma, H. J., Diepen van, A., Estevao, S., Kuipers, O. P., Groot de, R. and Hermans, P. specific impact of PsaR ofStreptococcus pneumoniae on global gene expression and

-1579.

Hendriksen, W. T., Bootsma, H. J., Estevao, S., Hoogenboezem, T., Jong de, A., Groot De, R., Kuipers, O. P. 2008a) CodY of Streptococcus pneumoniae: link between nutritional gene regulation

.190, pp. 590-601.

Hendriksen, W. T., Kloosterman, T. G., Bootsma, H. J., Estavao, S., Groot De, R., Kuipers, O. P. and Hermans, specific contributions of glutamate-dependent regulator GlnR and GlnR-

Streptococcus pneumoniae. Infection and Immunity. 76, pp. 1230-1238.

osterman, T. G., Bootsma, H. J., Estevao, S., Groot De, R., Hirst, R. A., Kadioglu, A., O’Callaghan, C. and Andrew, P. W. (2004) The role of pneumolysin in pneumococcal pneumonia and

Clinical and Experimental Immunology. 138, pp. 195-201.

ichsen, J. (1995) Six newly recognized types of Streptococcus pneumoniae. Journal of Clinical

Hollingshead, S. K., Becker, R. and Briles, D. E. (2000). Diversity of PspA: mosaic genes and evidence for past Streptococcus pneumoniae. Infection and Immunity. 68(10), pp. 5889-5900.

Hollingshead, S. K., Becker, R. and Briles, D. E. (2000) Diversity of PspA: Mosaic genes and evidence for past Streptococcus pneumoniae. Infection and Immunity.68, pp. 5889-5900.

Homes, A. R., McNab, R., Millsap, K. W., Rhode, M., Hammerschmidt, S., Mawdley, J. L. and Jenkinson, H. F. Streptococcus pneumoniae encodes a fibronectin-binding protein that is essential for

. 41, pp. 1395-1408.

Houldsworth, S., Andrew, P. W. and Mitchell, T. J. (1994) Pneumolysin stimulates production of tumour 1 beta by human mononuclear phagocytes. Infection and Immunity

Iannelli, F., Oggioni, M. R. and Pozzi, G. (2002) Allelic variation in the highly polymorphic locus . 284, pp. 63-71.

INCLEN (1999). Prospective multicentre hospital surveillance of Streptococcus

International Clinical Epidemiology Network (INCLEN). Lancet. 353(9160), pp. 1216-21.

Streptococcus pneumoniae: epidemiology and patterns of resistance. American Journal of

am, D., Appelbaum, P. C. and Gruneberg, R. N. (2003). The Alexander Project 19982000: susceptibility of pathogens isolated from community acquired respiratory tract infection to commonly

Journal of Antimicrobial Chemotherapy. 52(2), pp. 229-46.

Jarva, H., Jokiranta, T. S., Wurzner, R. and Meri, S. (2003) Complement resistance mechanisms of Streptococci. 107.

Jedrzejas, M. J. (2004) Extracellular virulence factors for Streptococcus pneumoniae. Frontiers in Bioscience

Creative Commons Attribution 3.0 Unported License

Paediatric Infectious Disease Journal.21, pp. 1008-

e, R. and Hermans, P. on global gene expression and

De, R., Kuipers, O. P. : link between nutritional gene regulation

oot De, R., Kuipers, O. P. and Hermans, -regulated genes

Hirst, R. A., Kadioglu, A., O’Callaghan, C. and Andrew, P. W. (2004) The role of pneumolysin in pneumococcal pneumonia and

Journal of Clinical

Hollingshead, S. K., Becker, R. and Briles, D. E. (2000). Diversity of PspA: mosaic genes and evidence for past

Hollingshead, S. K., Becker, R. and Briles, D. E. (2000) Diversity of PspA: Mosaic genes and evidence for past

Homes, A. R., McNab, R., Millsap, K. W., Rhode, M., Hammerschmidt, S., Mawdley, J. L. and Jenkinson, H. F. binding protein that is essential for

Houldsworth, S., Andrew, P. W. and Mitchell, T. J. (1994) Pneumolysin stimulates production of tumour Infection and Immunity. 62, pp.

Iannelli, F., Oggioni, M. R. and Pozzi, G. (2002) Allelic variation in the highly polymorphic locus pspC of

American Journal of

am, D., Appelbaum, P. C. and Gruneberg, R. N. (2003). The Alexander Project 1998-2000: susceptibility of pathogens isolated from community acquired respiratory tract infection to commonly

Jarva, H., Jokiranta, T. S., Wurzner, R. and Meri, S. (2003) Complement resistance mechanisms of Streptococci.

rontiers in Bioscience. 9,

Page 14: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: Jedrzejas, M. J., Hollingshead, S. K., Lebowitz, J., Chantalat, L., Briles, D. E. and Lamani, E. (2000).Production and characterization of the functional fragment of pneumococcal surface protein A. 373(1), pp. 116-125. Johnston, J. W., Myers, L. E., Ochs, M. M., Benjamin, W. H. J., Briles, D. E. and Hollingshead, S. K. (2004) Lipoprotein PsaA in virulence of Streptococcus superoxide. Infection and Immunity. 72, pp. 5858 Jomaa, M., Terry, S., Hale, C., Jones, X., Dougan, G. and Brown, J. (2006) Immunisation with iron uptake ABC transporter proteins PiaA and PiuA prevents respiratory infection with pp. 5133-5139. Jounblat, R., Kadioglu, A., Mitchell, T. J. and Andrew, P. W. (2003). Pneumococcal behaviuor and host responses during bronchopneumonia are affected differentlactivities of pneumolysin. Infection and Immunity Kadioglu, A., Ginges, N. A., Grattan, K., Kerr, A., Mitchell, T. J. and Andrew, P. W. (2000) Host cellular immune response to pneumococcal lung infection in mice. Kansanterveyslaitos, ed. Infectious diseases in FinInstitute B 13/2005. 2005, National Public Health Institute: Helsinki. 76 s. [Accessed 10 December, 2012]. Kausmally, L., Johnsborg, O., Lunde, M., Knutsen, E. and Havarstein, L. S. (2005) Choline(CbpD) in Streptococcus pneumoniae pp. 4338-4345. Kerr, A. R., Andrian, P. V., Estevao, S., Groot De, R., Alloing, G., Claverys, J. P., Mitchel, T. J. and Hermans P. W. (2004) The Ami-Ali A/Ali B permease of colonisation but not in invasive disease. Kerr, A. R., Paterson, G. K., Mccluskey, J., Iannelli, F., Oggioni, contribution of PspC to pneumococcal virulence varies between strains and is accomplished by both complement evasion and complement-independent mechanisms. Kilpi, T., Herva, E., Kaijalainen, T., Syrjänen, R. and Takala, A. K. (2001) Bacteriology of acute otitis media in a cohort of Finnish children followed for the first two years of life. pp. 654-662. Kim, E. Y., Choi, S. Y., Kwon, M. Y., Tran, D. H.T., Park, S. S. Lee, K. J., Bae, S. M., Briles, D. E. and Rhee, D. K. (2012) Streptococcus pneumoniaepepmice. Vaccine. 30, pp. 2008-2019. King, S. J., Hippe, K. R., Gould, J. M., Bae, D., Peterson, C, Cline, R. T., Fasching, C., Janoff, E. N. and Weiser, J. N. (2004) Phase variable desialylation of host proteins that bind to and protects airway. Molecular Microbiology Kloosterman, T. G., Hendriksen, W. T., Bijlsma, J. J., Bootsma, H. J., Hijum van, S. A., Kok, J., Hermans, P. W. M. and Kuipers, O. P. (2006) Regulation ofglutamine and glutamate metabolism by GlnR and GlnA in Streptococcus pneumoniae. Journal Biological Chemistry Koedel, U., Scheld, W. M. and Pfister, H. W. (2002) Pathogenesis and Pathophysiology of Pneumococcal Meningitis.Lancet Infectious Diseases.2

is licensed under a Creative Commons Attribution 3.0 Unported License

40

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

Jedrzejas, M. J., Hollingshead, S. K., Lebowitz, J., Chantalat, L., Briles, D. E. and Lamani, E. (2000).Production functional fragment of pneumococcal surface protein A. Arch Biochem Biophys

Johnston, J. W., Myers, L. E., Ochs, M. M., Benjamin, W. H. J., Briles, D. E. and Hollingshead, S. K. (2004) Streptococcus pneumoniae: surface accessibility and role in protection from . 72, pp. 5858-5867.

Jomaa, M., Terry, S., Hale, C., Jones, X., Dougan, G. and Brown, J. (2006) Immunisation with iron uptake ABC PiuA prevents respiratory infection with Streptococcus pneumoniae

Jounblat, R., Kadioglu, A., Mitchell, T. J. and Andrew, P. W. (2003). Pneumococcal behaviuor and host responses during bronchopneumonia are affected differently by the cytolytic and complement

Infection and Immunity. 71(4), pp. 1813-1819.

Kadioglu, A., Ginges, N. A., Grattan, K., Kerr, A., Mitchell, T. J. and Andrew, P. W. (2000) Host cellular immune response to pneumococcal lung infection in mice. Infection and Immunity. 68, pp. 492-501.

Infectious diseases in Finland 1995-2004. Publications of the National Public Health Institute B 13/2005. 2005, National Public Health Institute: Helsinki. 76 s. [Accessed 10 December, 2012].

Kausmally, L., Johnsborg, O., Lunde, M., Knutsen, E. and Havarstein, L. S. (2005) Choline-binding protein D is essential for competence induced lysis. Journal of Bacteriology

Kerr, A. R., Andrian, P. V., Estevao, S., Groot De, R., Alloing, G., Claverys, J. P., Mitchel, T. J. and Hermans Ali A/Ali B permease of Streptococcus pneumoniae is involved in nasopharyngeal

colonisation but not in invasive disease. Infection and Immunity. 72, pp. 3902-3906.

Kerr, A. R., Paterson, G. K., Mccluskey, J., Iannelli, F., Oggioni, M. R., Pozzi, G. and Mitchell, T. J. (2006) The contribution of PspC to pneumococcal virulence varies between strains and is accomplished by both

independent mechanisms. Infection and Immunity.74, pp.5319

Herva, E., Kaijalainen, T., Syrjänen, R. and Takala, A. K. (2001) Bacteriology of acute otitis media in a cohort of Finnish children followed for the first two years of life. Paediatric Infectious Disease Journal

., Kwon, M. Y., Tran, D. H.T., Park, S. S. Lee, K. J., Bae, S. M., Briles, D. E. and Rhee, Streptococcus pneumoniaepep27 mutant as a live vaccine for serotype-independent protection in

R., Gould, J. M., Bae, D., Peterson, C, Cline, R. T., Fasching, C., Janoff, E. N. and Weiser, J. N. (2004) Phase variable desialylation of host proteins that bind to Streptococcus pneumoniaein vivo

Molecular Microbiology. 54, pp. 159-171.

Kloosterman, T. G., Hendriksen, W. T., Bijlsma, J. J., Bootsma, H. J., Hijum van, S. A., Kok, J., Hermans, P. 2006) Regulation ofglutamine and glutamate metabolism by GlnR and GlnA in

Biological Chemistry.281, pp. 25097-25109.

Koedel, U., Scheld, W. M. and Pfister, H. W. (2002) Pathogenesis and Pathophysiology of Pneumococcal 2, pp. 721-736.

Creative Commons Attribution 3.0 Unported License

Jedrzejas, M. J., Hollingshead, S. K., Lebowitz, J., Chantalat, L., Briles, D. E. and Lamani, E. (2000).Production Arch Biochem Biophys.

Johnston, J. W., Myers, L. E., Ochs, M. M., Benjamin, W. H. J., Briles, D. E. and Hollingshead, S. K. (2004) : surface accessibility and role in protection from

Jomaa, M., Terry, S., Hale, C., Jones, X., Dougan, G. and Brown, J. (2006) Immunisation with iron uptake ABC Streptococcus pneumoniae. Vaccine. 24,

Jounblat, R., Kadioglu, A., Mitchell, T. J. and Andrew, P. W. (2003). Pneumococcal behaviuor and host y by the cytolytic and complement-activating

Kadioglu, A., Ginges, N. A., Grattan, K., Kerr, A., Mitchell, T. J. and Andrew, P. W. (2000) Host cellular 501.

. Publications of the National Public Health Institute B 13/2005. 2005, National Public Health Institute: Helsinki. 76 s. [Accessed 10 December, 2012].

binding protein D Journal of Bacteriology. 187,

Kerr, A. R., Andrian, P. V., Estevao, S., Groot De, R., Alloing, G., Claverys, J. P., Mitchel, T. J. and Hermans , is involved in nasopharyngeal

M. R., Pozzi, G. and Mitchell, T. J. (2006) The contribution of PspC to pneumococcal virulence varies between strains and is accomplished by both

5319-5324.

Herva, E., Kaijalainen, T., Syrjänen, R. and Takala, A. K. (2001) Bacteriology of acute otitis media in Paediatric Infectious Disease Journal. 20,

., Kwon, M. Y., Tran, D. H.T., Park, S. S. Lee, K. J., Bae, S. M., Briles, D. E. and Rhee, independent protection in

R., Gould, J. M., Bae, D., Peterson, C, Cline, R. T., Fasching, C., Janoff, E. N. and Streptococcus pneumoniaein vivo

Kloosterman, T. G., Hendriksen, W. T., Bijlsma, J. J., Bootsma, H. J., Hijum van, S. A., Kok, J., Hermans, P. 2006) Regulation ofglutamine and glutamate metabolism by GlnR and GlnA in

Koedel, U., Scheld, W. M. and Pfister, H. W. (2002) Pathogenesis and Pathophysiology of Pneumococcal

Page 15: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: Kyaw, M. H., Christie, P., Clarke, S. C., Mooney, J. D., Ahmed, S., Jones, I. G. and Campbell, H. (2003) Invasive pneumococcal disease in Scotland, 1999patients and disease in relation to future Lau, G. W., Haataja, W. S., Lonetto, M., Kensit, S. E., Marra, A., Bryant, A. P., McDevitt, D. A. and Holden, D. W. (2001) A functional genomic analysis of type 3 Microbiology. 40, pp. 555-571. Lee, C. J., Banks, S. D. and Li, J. P. (1991) Virulence, Immunity and Vaccine related to pneumoniae. Critical Reviews in Microbiology LeMessurier, K. S., Ogunniyi, A. D. and Paton, J. C. (2006) Differential expression of key pneumococcal virulence genes in vivo.Microbiology. 152, pp. 305 Luotonen, J., Herva, E., Karma, P., Timonen, M., Leinonen, M. and Mäkelä, P. H. (1981) The bacteriology of acute otitis media in children: with special reference to and antigen methods. Scandinavian Journal Infectious Disease Martner, A., Dahlgren, C., Paton, J. C. and Wold, A. E. (2008) Pneumolysin pneumoniae autolysis is a potent activator of intracellular oxygen radical production in neutrophils. and Immunity. 76, pp. 4079-4087. McAllister, L. J., Tseng, H. J., Ogunniyi, A. D., Jennings, M. P., McEwan, A. G.Molecular analysis of the psa permease complex of 889-901. McCool, T. L., Cate, T. R., Moy, G. and Weiser, J. N. (2002) The immune response to pneumococcal proteins during experimental human carriage. Journal of Experimental Medicine McCuller, J. A. and Tuomanen, E. I. (2001) Molecular pathogenesis of pneumococcal pneumonia.Bioscience.1, pp. D877-889. Mcellistrem, M. C., Ransford, J. V. and pneumococcal phase variants of a clinically relevant serotype 3 clone. 97-101. Mitchell, T. J., Andrew, P. W., Saunders, F. K., Smith, A. N. and Boand antibody binding by pneumolysin via a region of toxin homologous to a human acuteMolecular Microbiology. 5, pp. 1883-1888. Morrison, K. E., Lake D., Crook, J., Carlone, G. M., Ades, E., FacklaConfirmation of psaA in all 90 serotypes of identification and diagnosis. Journal of Clinical Microbiology Moschioni, M., De Angelis, G., Melchiorre, S., Masignani, V., Leibovitz, E., Barocchi, M. A. and Dagan, R. (2010) Prevalence of pilus-encoding islets among acute otitis media Israel. Clinical Microbiology and Infectious Diseases Moschioni, M., Donati, C., Muzzi, A., Masignani, V., Censini, S., Hanage, W. P., Bishop, C. J., Reis, J. N., Normark, S., Henriques-Normark, B., Covacci, A., Rappuoli, R. and Barocchi, M. A. (2008). Streptococcus pneumoniae contains 3 rlrA pilus variants t896. Musher, D. M. (2005) Streptococcus pneumoniae in Mandell, Douglas and Bennett's Principles and Practice of Infectious Diseases, Philadelphia, Elsevier Churchill Livingstone.

is licensed under a Creative Commons Attribution 3.0 Unported License

41

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

Kyaw, M. H., Christie, P., Clarke, S. C., Mooney, J. D., Ahmed, S., Jones, I. G. and Campbell, H. (2003) Invasive pneumococcal disease in Scotland, 1999-2001: Use of record linkeage to explore associations between patients and disease in relation to future vaccination policy. Clinical Infectious Diseases.37, pp. 1283

Lau, G. W., Haataja, W. S., Lonetto, M., Kensit, S. E., Marra, A., Bryant, A. P., McDevitt, D. A. and Holden, D. W. (2001) A functional genomic analysis of type 3 Streptococcus pneumoniae virulence.

Lee, C. J., Banks, S. D. and Li, J. P. (1991) Virulence, Immunity and Vaccine related to Critical Reviews in Microbiology. 18(2), pp. 89-114.

nniyi, A. D. and Paton, J. C. (2006) Differential expression of key pneumococcal . 152, pp. 305-311.

Luotonen, J., Herva, E., Karma, P., Timonen, M., Leinonen, M. and Mäkelä, P. H. (1981) The bacteriology of s media in children: with special reference to Streptococcus pneumonia as studied by bacteriological

Scandinavian Journal Infectious Disease. 13, pp. 177-183.

Martner, A., Dahlgren, C., Paton, J. C. and Wold, A. E. (2008) Pneumolysin released during autolysis is a potent activator of intracellular oxygen radical production in neutrophils.

McAllister, L. J., Tseng, H. J., Ogunniyi, A. D., Jennings, M. P., McEwan, A. G. and Paton, J. C. (2004) Molecular analysis of the psa permease complex of Streptococcus pneumoniae. Molecular Microbiology

McCool, T. L., Cate, T. R., Moy, G. and Weiser, J. N. (2002) The immune response to pneumococcal proteins Journal of Experimental Medicine. 195, pp. 359-365.

McCuller, J. A. and Tuomanen, E. I. (2001) Molecular pathogenesis of pneumococcal pneumonia.

Mcellistrem, M. C., Ransford, J. V. and Khan, S. A. (2007) Characterization of in vitro biofilmpneumococcal phase variants of a clinically relevant serotype 3 clone. Journal of Clinical Microbiology.

Mitchell, T. J., Andrew, P. W., Saunders, F. K., Smith, A. N. and Boulnois, G. J. (1991) Complement activation and antibody binding by pneumolysin via a region of toxin homologous to a human acute

1888.

Morrison, K. E., Lake D., Crook, J., Carlone, G. M., Ades, E., Facklam, R. and Sampson, J.S. (2000) Confirmation of psaA in all 90 serotypes of Streptococcus pneumoniae by PCR and potential of this assay for

Journal of Clinical Microbiology. 38, pp. 434-437.

hiorre, S., Masignani, V., Leibovitz, E., Barocchi, M. A. and Dagan, R. encoding islets among acute otitis media Streptococcus pneumoniae isolates from

Clinical Microbiology and Infectious Diseases. 16 (9), pp. 1501-1504.

Moschioni, M., Donati, C., Muzzi, A., Masignani, V., Censini, S., Hanage, W. P., Bishop, C. J., Reis, J. N., Normark, B., Covacci, A., Rappuoli, R. and Barocchi, M. A. (2008). Streptococcus

pneumoniae contains 3 rlrA pilus variants that are clonally related. Journal of Infectious Disease

Streptococcus pneumoniae in Mandell, Douglas and Bennett's Principles and Practice of Philadelphia, Elsevier Churchill Livingstone.

Creative Commons Attribution 3.0 Unported License

Kyaw, M. H., Christie, P., Clarke, S. C., Mooney, J. D., Ahmed, S., Jones, I. G. and Campbell, H. (2003) 2001: Use of record linkeage to explore associations between

1283-1291.

Lau, G. W., Haataja, W. S., Lonetto, M., Kensit, S. E., Marra, A., Bryant, A. P., McDevitt, D. A. and Holden, D. virulence. Molecular

Lee, C. J., Banks, S. D. and Li, J. P. (1991) Virulence, Immunity and Vaccine related to Streptococcus

nniyi, A. D. and Paton, J. C. (2006) Differential expression of key pneumococcal

Luotonen, J., Herva, E., Karma, P., Timonen, M., Leinonen, M. and Mäkelä, P. H. (1981) The bacteriology of as studied by bacteriological

released during Streptococcus autolysis is a potent activator of intracellular oxygen radical production in neutrophils. Infection

and Paton, J. C. (2004) Molecular Microbiology. 53, pp.

McCool, T. L., Cate, T. R., Moy, G. and Weiser, J. N. (2002) The immune response to pneumococcal proteins

McCuller, J. A. and Tuomanen, E. I. (2001) Molecular pathogenesis of pneumococcal pneumonia.Frontiers of

biofilm-associated Journal of Clinical Microbiology.45, pp.

ulnois, G. J. (1991) Complement activation and antibody binding by pneumolysin via a region of toxin homologous to a human acute-phase protein.

m, R. and Sampson, J.S. (2000) by PCR and potential of this assay for

hiorre, S., Masignani, V., Leibovitz, E., Barocchi, M. A. and Dagan, R. isolates from

Moschioni, M., Donati, C., Muzzi, A., Masignani, V., Censini, S., Hanage, W. P., Bishop, C. J., Reis, J. N., Normark, B., Covacci, A., Rappuoli, R. and Barocchi, M. A. (2008). Streptococcus

Journal of Infectious Disease. 197, pp. 888-

Streptococcus pneumoniae in Mandell, Douglas and Bennett's Principles and Practice of

Page 16: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: O'Brien, K. L. and Santosham, M. (2004) Potential impact of conjugate pneumococcal vaccines on pediatric pneumococcal diseases. American Journal of Epidemiology Ogggioni, M. R., Memmi, G., Maggi, T., Chiavolini, D., Iannelli, F. and Pozzi, G. (2003) Pneumococcal zinc metalloproteinase ZmpC cleaves human matrix metalloproteinase 9 and is a virulence factor in experimental pneumonia. Molecular Microbiology. 49, pp. 795 Ogunniyi, A. D., Giammarinaro, P. and Paton, J. C. (2002) The genes encoding virulenceand the capsule of Streptococcus pneumoniaeMicrobiology. 148, pp. 2045-2053. Ogunniyi, A. D., LeMessurier, K. S., Graham, R. M., Watt, J. M., Briles, D. E., Stroeher, U. H. and Paton, J. C. (2007) Contributions of pneumolysin, pneumococcal surface protein A (PspA), and PspC to pathogenicity of Streptococcus pneumoniae D39 in a mouse model. Ogunniyi, A. D., Woodrow, M. C., Poolman, J. T. and Paton, J. C. (2001) Protection against pneumoniae elicited by immunization with pneumolysin and CbpA. Orihuela, C. J., Gao, G., Francis, K. P., Yu, J. and Tuomanen, E. I. (2004) Tissuepneumococcal virulence factors to pathogenesis. Ostergaard, C., Brandt, C., Konradsen, H. B. and Samuelsson, S. (2004) Differences in survival, brain damage, and cerebrospinal fluid cytokine kinetics due to meningitis caused by 3 different serotypes: Evaluation in humans and in 1220. Park, I. H., Pritchard, D. G., Cartee, R., Brandao, A., Brandileone, M. C. and Nahm, M. H. (2007) Discovery of a new capsular serotype (6C) within serogroup 6 of Microbiology. 45(4), pp. 1225-1233. Paterson, G. K., Blue, C. E. and Mitchell, T. J. (2006) Role of twoStreptococcus pneumoniae. Journal of Medical Microbiology Paton, J. C. (1996) The contributions of pneumolysin to the pathogenicity of in Microbiology. 4, pp. 103-106. Paton, J. C., Andrew, P.W., Boulnois, G. J. and Mitchell, T. J. (1993) Molecular analysis of the pathogenicity of Streptococcus pneumoniae: role of pneumococcal proteins. Paton, J. C., Rowan-Kelly, B. and Ferrante, A. (1984) Activation of human complement by the pneumococcal toxin pneumolysin.Infection and Immunity Pericone, C. D., Overweg, K., Hermans, P. W. and Weiser, J. N. (2000) Inhibitory and antibactericidal effects of hydrogen peroxide production by Streptococcus pneumoniaeInfection and Immunity. 68, pp. 3990-3997. Pericone, C. D., Park, S., Imlay, J. A. and Weiser, J. N. (2003) Factors contributing to hydrogen peroxide resistance in Streptococcus pneumoniae the Fenton reaction. Journal of Bacteriology. Pettigrew, M. M., Fennie, K. P., York, M. P., Daniels, J. and Ghaffar, F. (2006) Variation in the Presence of Neuraminidase Genes among Streptococcus pneumoniae Immunity.74, pp. 3360-3365. Pimenta, F. C., Miyaji, E. N., Areas, A. P., Oliveira, M. L., de Andrade, A. L., Ho, P. L., Hollingshead, S. K. and Leite, L. C. (2006) Intranasal immunization with the cholera toxin B subunitpneumococcal surface antig

is licensed under a Creative Commons Attribution 3.0 Unported License

42

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

O'Brien, K. L. and Santosham, M. (2004) Potential impact of conjugate pneumococcal vaccines on pediatric American Journal of Epidemiology. 159, pp. 634-644.

i, M. R., Memmi, G., Maggi, T., Chiavolini, D., Iannelli, F. and Pozzi, G. (2003) Pneumococcal zinc metalloproteinase ZmpC cleaves human matrix metalloproteinase 9 and is a virulence factor in experimental

. 49, pp. 795-805.

Ogunniyi, A. D., Giammarinaro, P. and Paton, J. C. (2002) The genes encoding virulence-associated proteins Streptococcus pneumoniae are upregulated and differentially expressed

D., LeMessurier, K. S., Graham, R. M., Watt, J. M., Briles, D. E., Stroeher, U. H. and Paton, J. C. (2007) Contributions of pneumolysin, pneumococcal surface protein A (PspA), and PspC to pathogenicity of

D39 in a mouse model. Infection and Immunity. 75, pp. 1843-1851.

Ogunniyi, A. D., Woodrow, M. C., Poolman, J. T. and Paton, J. C. (2001) Protection against elicited by immunization with pneumolysin and CbpA. Infection and Immunity. 69, pp. 5997

ela, C. J., Gao, G., Francis, K. P., Yu, J. and Tuomanen, E. I. (2004) Tissue-specific contributions of pneumococcal virulence factors to pathogenesis. Journal of Infectious Diseases.190, pp. 1661-1669.

Ostergaard, C., Brandt, C., Konradsen, H. B. and Samuelsson, S. (2004) Differences in survival, brain damage, and cerebrospinal fluid cytokine kinetics due to meningitis caused by 3 different Streptococcus pneumoniae serotypes: Evaluation in humans and in 2 experimental models. Journal of Infectious Diseases.

Park, I. H., Pritchard, D. G., Cartee, R., Brandao, A., Brandileone, M. C. and Nahm, M. H. (2007) Discovery of a new capsular serotype (6C) within serogroup 6 of Streptococcus pneumoniae. Journal of Clinical

Paterson, G. K., Blue, C. E. and Mitchell, T. J. (2006) Role of two-component systems in the virulence of Journal of Medical Microbiology. 55, pp. 355-363.

C. (1996) The contributions of pneumolysin to the pathogenicity of Streptococcus pneumoniae

Paton, J. C., Andrew, P.W., Boulnois, G. J. and Mitchell, T. J. (1993) Molecular analysis of the pathogenicity of : role of pneumococcal proteins. Annals Review in Microbiology. 47, pp. 89

Kelly, B. and Ferrante, A. (1984) Activation of human complement by the pneumococcal Infection and Immunity. 43, pp. 1085-1087.

Pericone, C. D., Overweg, K., Hermans, P. W. and Weiser, J. N. (2000) Inhibitory and antibactericidal effects of Streptococcus pneumoniae on other inhabitants of upper respiratory tract.

3997.

Pericone, C. D., Park, S., Imlay, J. A. and Weiser, J. N. (2003) Factors contributing to hydrogen peroxide Streptococcus pneumoniae include pyruvate oxidase (SpxB) and avoidance of the toxic effects of

Journal of Bacteriology.185, pp. 6815-6825.

Pettigrew, M. M., Fennie, K. P., York, M. P., Daniels, J. and Ghaffar, F. (2006) Variation in the Presence of Streptococcus pneumoniae Isolates with Identical Sequence Types.

Pimenta, F. C., Miyaji, E. N., Areas, A. P., Oliveira, M. L., de Andrade, A. L., Ho, P. L., Hollingshead, S. K. and Leite, L. C. (2006) Intranasal immunization with the cholera toxin B subunitpneumococcal surface antig

Creative Commons Attribution 3.0 Unported License

O'Brien, K. L. and Santosham, M. (2004) Potential impact of conjugate pneumococcal vaccines on pediatric

i, M. R., Memmi, G., Maggi, T., Chiavolini, D., Iannelli, F. and Pozzi, G. (2003) Pneumococcal zinc metalloproteinase ZmpC cleaves human matrix metalloproteinase 9 and is a virulence factor in experimental

associated proteins are upregulated and differentially expressed in vivo.

D., LeMessurier, K. S., Graham, R. M., Watt, J. M., Briles, D. E., Stroeher, U. H. and Paton, J. C. (2007) Contributions of pneumolysin, pneumococcal surface protein A (PspA), and PspC to pathogenicity of

Ogunniyi, A. D., Woodrow, M. C., Poolman, J. T. and Paton, J. C. (2001) Protection against Streptococcus . 69, pp. 5997-6003.

specific contributions of 1669.

Ostergaard, C., Brandt, C., Konradsen, H. B. and Samuelsson, S. (2004) Differences in survival, brain damage, Streptococcus pneumoniae

Journal of Infectious Diseases.190, pp. 1212-

Park, I. H., Pritchard, D. G., Cartee, R., Brandao, A., Brandileone, M. C. and Nahm, M. H. (2007) Discovery of Journal of Clinical

component systems in the virulence of

Streptococcus pneumoniae. Trends

Paton, J. C., Andrew, P.W., Boulnois, G. J. and Mitchell, T. J. (1993) Molecular analysis of the pathogenicity of . 47, pp. 89-115.

Kelly, B. and Ferrante, A. (1984) Activation of human complement by the pneumococcal

Pericone, C. D., Overweg, K., Hermans, P. W. and Weiser, J. N. (2000) Inhibitory and antibactericidal effects of on other inhabitants of upper respiratory tract.

Pericone, C. D., Park, S., Imlay, J. A. and Weiser, J. N. (2003) Factors contributing to hydrogen peroxide include pyruvate oxidase (SpxB) and avoidance of the toxic effects of

Pettigrew, M. M., Fennie, K. P., York, M. P., Daniels, J. and Ghaffar, F. (2006) Variation in the Presence of Isolates with Identical Sequence Types. Infection and

Pimenta, F. C., Miyaji, E. N., Areas, A. P., Oliveira, M. L., de Andrade, A. L., Ho, P. L., Hollingshead, S. K. and Leite, L. C. (2006) Intranasal immunization with the cholera toxin B subunitpneumococcal surface antigen

Page 17: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: A fusion protein induces protection against colonization with impact on the nasopharyngeal and oral microbiota of mice. pneumoniaedisease in India. Invasive Bacterial Infection Surveillance (IBIS) Group,Pracht, D., Elm, C., Gerber, J., Bergmann, S., Rhode, M., Seiler, M., Kim, K. S., Jenkinson, H. F., Nau, R. and Hammerschmidt, S. (2005) PavA of Streptococcus pneumoniae modulates adherence, invasion and meningeal inflammation. Immunity. 73, pp. 2680-2689. Poulsen, K., Reinholdt, J., Jespersgaard, C., Boye, K., Brown, T. A., Hauge, M. and Kilian, M. (1998) A comprehensive genetic study of streptococcal immunoglobulin A1 proteases: Evidence for recombination within and between species. Infection and Immunity. Ren, B., Szalai, A. J., Hollingshead, S. K. and Briles, D. E. (2004) Effects of PspA and antibodies to PspA oactivation and deposition of complement on the pneumococcal surface. Ren, B., Szalai, A. J., Thomas, O., Hollingshead, S. K. and Briles, D. E. (2003) Both family 1 and family 2 PspA proteins can inhibit complemenStreptococcus pneumoniae. Infection and Immunity. Ring, A., Weiser, J. N. and Tuomonen, E. I. (1998) Pneumococcal trafficking across the bloodJournal of Clinical Investigation. 102, pp. 347 Romero-Sterner, S., Pilishvili, T., Sampson, J. S., Johnson, S. E., Stinson, A., Carlone, G. M. and Ades, E. W. (2003) Inhibition of pneumococcal adherence to human nasopharyngeal epithelial cells by antiClincal Diagnosis and Laboratory Immunology Rosenow, C., Ryan, P., Weiser, J. N., Johnson, S., Fontan, P., Ortquvist, A. and Masure, H. R. (1997) Contribution of novel choline-binding proteins to adherence, colonization and immupneumoniae.Molecular Microbiology. 25, pp. 819 Rubins, J. B., Charboneau, D., Paton, J. C., Mitchell, T. J., Andrew, P. W. and Janoff, E. N. (1995) Dual function of pneumolysin in the early pathogenesis of murine pneumococInvestigation. 95, pp. 142-150. Rubins, J. B., Hanson, P. A., Charboneau, D., Berry, A. M., Paton, J. C. and Janoff, E. N. (1998) Pneumolysin in pneumococcal adherence and colonisation . Saha, S. K., Naheed, A., Arifeen El, S., Islam, M., AlBreiman, R. F., Sack, D. A. and Luby, S. P. (2009). Surveillance for invasive among hospitalized children in Bangladesh: antimicrobial susceptibility and serotype distribution. Infectious Disease.48 (Suppl 2), pp. S75 Sanchez-Beato, A. R., Lopez, R. and Garcia, J. L. (1998) Molecular characterisation of PcpA: a novel cholinebinding protein of Streptococcus pneumoniae Sandgren, A., Sjostrom, K., Olsson-Liljequist, B. O., Christensson, B., Samuelsson, A., Kronvall, G. and Normark, B. H. (2004) Effect of clonal and serotypeStreptococcus pneumoniae. Journal of Infectious Diseases. Shakhnovich, E. A., King, S. J. and Weisser, J. N. (2002) Neuraminidase expressed by pneumoniae desialylates the lipopolysaccharide of paradigm for interbacterial competition among pathogens of the human respiratory tract. Immunity. 70, pp. 7161-7164.

is licensed under a Creative Commons Attribution 3.0 Unported License

43

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

A fusion protein induces protection against colonization with Streptococcus pneumoniae and has negligible impact on the nasopharyngeal and oral microbiota of mice. Infection and Immunity. 74, pp. 4939

disease in India. Invasive Bacterial Infection Surveillance (IBIS) Group,Pracht, D., Elm, C., Gerber, J., Bergmann, S., Rhode, M., Seiler, M., Kim, K. S., Jenkinson, H. F., Nau, R. and Hammerschmidt, S. (2005)

modulates adherence, invasion and meningeal inflammation.

Poulsen, K., Reinholdt, J., Jespersgaard, C., Boye, K., Brown, T. A., Hauge, M. and Kilian, M. (1998) A tudy of streptococcal immunoglobulin A1 proteases: Evidence for recombination within

Infection and Immunity.66, pp. 181-190.

Ren, B., Szalai, A. J., Hollingshead, S. K. and Briles, D. E. (2004) Effects of PspA and antibodies to PspA oactivation and deposition of complement on the pneumococcal surface. Infection and Immunity. 72, pp. 114

Ren, B., Szalai, A. J., Thomas, O., Hollingshead, S. K. and Briles, D. E. (2003) Both family 1 and family 2 PspA proteins can inhibit complement deposition and confer virulence to a capsular serotype 3 strain of

Infection and Immunity.71, pp. 75-85.

Ring, A., Weiser, J. N. and Tuomonen, E. I. (1998) Pneumococcal trafficking across the blood-brain barrier.. 102, pp. 347-360.

Sterner, S., Pilishvili, T., Sampson, J. S., Johnson, S. E., Stinson, A., Carlone, G. M. and Ades, E. W. (2003) Inhibition of pneumococcal adherence to human nasopharyngeal epithelial cells by anti-Clincal Diagnosis and Laboratory Immunology. 10, pp. 246-251.

Rosenow, C., Ryan, P., Weiser, J. N., Johnson, S., Fontan, P., Ortquvist, A. and Masure, H. R. (1997) binding proteins to adherence, colonization and immunogenicity of

. 25, pp. 819-829.

Rubins, J. B., Charboneau, D., Paton, J. C., Mitchell, T. J., Andrew, P. W. and Janoff, E. N. (1995) Dual function of pneumolysin in the early pathogenesis of murine pneumococcal pneumonia. Journal of Clinical

Rubins, J. B., Hanson, P. A., Charboneau, D., Berry, A. M., Paton, J. C. and Janoff, E. N. (1998) Pneumolysin in pneumococcal adherence and colonisation . Microbial Pathogenesis. 25, pp. 337-342.

Saha, S. K., Naheed, A., Arifeen El, S., Islam, M., Al-Emran, H., Amin, R., Fatima, K., Brooks, W. A., Breiman, R. F., Sack, D. A. and Luby, S. P. (2009). Surveillance for invasive Streptococcus pneumoniae

ngladesh: antimicrobial susceptibility and serotype distribution. .48 (Suppl 2), pp. S75-81.

Beato, A. R., Lopez, R. and Garcia, J. L. (1998) Molecular characterisation of PcpA: a novel cholinecoccus pneumoniae. FEMS Microbiology Letters. 164, pp. 207-214.

Liljequist, B. O., Christensson, B., Samuelsson, A., Kronvall, G. and Normark, B. H. (2004) Effect of clonal and serotype-specific properties on the invasiv

Journal of Infectious Diseases.189, pp. 785-796.

Shakhnovich, E. A., King, S. J. and Weisser, J. N. (2002) Neuraminidase expressed by desialylates the lipopolysaccharide of Neisseria meningitidis and Haemophilus influenzae

paradigm for interbacterial competition among pathogens of the human respiratory tract.

Creative Commons Attribution 3.0 Unported License

and has negligible . 74, pp. 4939-4944.

disease in India. Invasive Bacterial Infection Surveillance (IBIS) Group,Pracht, D., Elm, C., Gerber, J., Bergmann, S., Rhode, M., Seiler, M., Kim, K. S., Jenkinson, H. F., Nau, R. and Hammerschmidt, S. (2005)

modulates adherence, invasion and meningeal inflammation. Infection and

Poulsen, K., Reinholdt, J., Jespersgaard, C., Boye, K., Brown, T. A., Hauge, M. and Kilian, M. (1998) A tudy of streptococcal immunoglobulin A1 proteases: Evidence for recombination within

Ren, B., Szalai, A. J., Hollingshead, S. K. and Briles, D. E. (2004) Effects of PspA and antibodies to PspA on . 72, pp. 114-122.

Ren, B., Szalai, A. J., Thomas, O., Hollingshead, S. K. and Briles, D. E. (2003) Both family 1 and family 2 t deposition and confer virulence to a capsular serotype 3 strain of

brain barrier.

Sterner, S., Pilishvili, T., Sampson, J. S., Johnson, S. E., Stinson, A., Carlone, G. M. and Ades, E. W. PsaP antibodies.

Rosenow, C., Ryan, P., Weiser, J. N., Johnson, S., Fontan, P., Ortquvist, A. and Masure, H. R. (1997) nogenicity of Streptococcus

Rubins, J. B., Charboneau, D., Paton, J. C., Mitchell, T. J., Andrew, P. W. and Janoff, E. N. (1995) Dual Journal of Clinical

Rubins, J. B., Hanson, P. A., Charboneau, D., Berry, A. M., Paton, J. C. and Janoff, E. N. (1998) Pneumolysin

Emran, H., Amin, R., Fatima, K., Brooks, W. A., Streptococcus pneumoniae disease

ngladesh: antimicrobial susceptibility and serotype distribution. Clinical

Beato, A. R., Lopez, R. and Garcia, J. L. (1998) Molecular characterisation of PcpA: a novel choline-

Liljequist, B. O., Christensson, B., Samuelsson, A., Kronvall, G. and specific properties on the invasive capacity of

Shakhnovich, E. A., King, S. J. and Weisser, J. N. (2002) Neuraminidase expressed by Streptococcus Haemophilus influenzae: a

paradigm for interbacterial competition among pathogens of the human respiratory tract. Infection and

Page 18: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: Shaper, M., Hollingshead, S. K., Benjamin, W. H. J. and Briles, D. E. (2004) PspA protects pneumoniae from killing by apolactoferrin, and antibody to PspA enhances killing of pneumococci by apolactoferrin. Infection and Immunity. 72, pp. 5031 Shouval, D. S., Greenberg, D., Givon-Lavi, N., Porat, N. and Dagan, R. (2006) Siteindividual Streptococcus pneumoniae conjunctivitis. Paediatric Infectious Disease Journal. Soewignjo, S., Gessner, B. D. and Sutanto, A. (2001) prevalence, serotype distribution, and resistance patterns among children on Lombok Island, Indonesia. Infectious Disease. 32, pp. 1039-1043. Spellerberg, B., Cundell, D. R., Sandros, J., Pearce, B. J., Idanpaan(1996) Pyruvate oxidase, as a determinant of virulence in 19, pp. 803-813. Swiatto, E. and Ware, D. (2003) Novel vaccine strategies with protein antigens of pneumoniae.FEMS Immunology and Medical Microbiology Syrjänen, R. K., Kilpi, T. M., Kaijalainen, T. H., Herva, E. E. and of Streptococcus pneumoniae in Finnish children younger than 2 years old. pp. 451-459. Syrogiannopoulos, G. A., Grivea, I. N., Davies, T.A., Katopodis, G. D., Appelbaum, P. C. and Beratis, N. G. (2000) Antimicrobial use and colonisation with erythromycinduring the first 2 years of life. Clinical Infectious Disease Tafuri, S., Martinelli, D., Grimaldi, A., Lopatriello, A., Giorgio, V. and Prato, R. (2011) 23pneumococcal vaccine failure in a patient who developed pneumonia: a case report.417-420. Talkington, D. F., Brown, B. G., Tharpe, J. A., Koenig, A. and Russell, H. (1996) Protection of mice against fatal pneumococcal challenge by immunization with pneumococcal surface adhesion A (PsaA).and Pathology. 2, pp. 17-22. Talkington, D. F., Crimmins, D. L., Voellinger, D. C., Yother, J. and Briles, D. E. (1991) A 43pneumococcal surface protein, PspA: isolation, protective abilities, and structure analysis of the amino acidterminal sequence. Infection and Immunity Tong, H. H., Blue, L. E., James, M. A. and DeMaria, T. F. (2000) Evaluation of the virulence of a pneumoniae neuraminidase-deficient mutant in nasopharyngeal colonisation and development of otitis media in Chinchilla model. Infection and Immunity. 68, pp. 921 Tseng, H. J., McEwan, A. G., Paton, J. C. and Jennings, M. P. (2002) Virulence of PsaA mutants are hypersensitive to oxidative stress. Tu, A. H., Fulgham, R. L., McCrory, M. A., Briles, D. E. and SzalaiA inhibits complement activation by Streptococcus pneumoniaeUnited States. Journal of Infectious Disease

is licensed under a Creative Commons Attribution 3.0 Unported License

44

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

Shaper, M., Hollingshead, S. K., Benjamin, W. H. J. and Briles, D. E. (2004) PspA protects from killing by apolactoferrin, and antibody to PspA enhances killing of pneumococci by

. 72, pp. 5031-5040.

Lavi, N., Porat, N. and Dagan, R. (2006) Site-specific disease potential of Streptococcus pneumoniae serotypes in pediatric invasive disease, acute otitis media and acute

fectious Disease Journal.25, pp. 602-607.

Soewignjo, S., Gessner, B. D. and Sutanto, A. (2001) Streptococcus pneumoniae nasopharyngeal carriage prevalence, serotype distribution, and resistance patterns among children on Lombok Island, Indonesia.

Spellerberg, B., Cundell, D. R., Sandros, J., Pearce, B. J., Idanpaan-Heikkila, I., Rosenow, C. and Masure, H. R. (1996) Pyruvate oxidase, as a determinant of virulence in Streptococcus pneumoniae. Molecular Microbi

Swiatto, E. and Ware, D. (2003) Novel vaccine strategies with protein antigens of FEMS Immunology and Medical Microbiology. 38, pp. 1-7.

Syrjänen, R. K., Kilpi, T. M., Kaijalainen, T. H., Herva, E. E. and Takala, A. K. (2001) Nasopharyngeal carriage in Finnish children younger than 2 years old. Journal of Infectious Disease

Syrogiannopoulos, G. A., Grivea, I. N., Davies, T.A., Katopodis, G. D., Appelbaum, P. C. and Beratis, N. G. (2000) Antimicrobial use and colonisation with erythromycin-resistant Streptococcus pneumoniae

Clinical Infectious Disease. 31, pp. 887-893.

Tafuri, S., Martinelli, D., Grimaldi, A., Lopatriello, A., Giorgio, V. and Prato, R. (2011) 23pneumococcal vaccine failure in a patient who developed pneumonia: a case report.New Microbiologica

Talkington, D. F., Brown, B. G., Tharpe, J. A., Koenig, A. and Russell, H. (1996) Protection of mice against fatal pneumococcal challenge by immunization with pneumococcal surface adhesion A (PsaA).

D. F., Crimmins, D. L., Voellinger, D. C., Yother, J. and Briles, D. E. (1991) A 43pneumococcal surface protein, PspA: isolation, protective abilities, and structure analysis of the amino acid

Infection and Immunity. 59, pp. 1285-1289.

Tong, H. H., Blue, L. E., James, M. A. and DeMaria, T. F. (2000) Evaluation of the virulence of a deficient mutant in nasopharyngeal colonisation and development of otitis media in

ion and Immunity. 68, pp. 921-924.

Tseng, H. J., McEwan, A. G., Paton, J. C. and Jennings, M. P. (2002) Virulence of Streptococcus pneumoniaePsaA mutants are hypersensitive to oxidative stress. Infection and Immunity. 70, pp. 1635-1639.

Tu, A. H., Fulgham, R. L., McCrory, M. A., Briles, D. E. and Szalai, A. J. (1999) Pneumococcal surface protein Streptococcus pneumoniae. Infection and Immunity. 67, pp. 4720

Journal of Infectious Disease. 188(5), pp. 643-652.

Creative Commons Attribution 3.0 Unported License

Shaper, M., Hollingshead, S. K., Benjamin, W. H. J. and Briles, D. E. (2004) PspA protects Streptococcus from killing by apolactoferrin, and antibody to PspA enhances killing of pneumococci by

specific disease potential of serotypes in pediatric invasive disease, acute otitis media and acute

nasopharyngeal carriage prevalence, serotype distribution, and resistance patterns among children on Lombok Island, Indonesia. Clinical

Heikkila, I., Rosenow, C. and Masure, H. R. Molecular Microbiology.

Swiatto, E. and Ware, D. (2003) Novel vaccine strategies with protein antigens of Streptococcus

Takala, A. K. (2001) Nasopharyngeal carriage Journal of Infectious Disease. 184,

Syrogiannopoulos, G. A., Grivea, I. N., Davies, T.A., Katopodis, G. D., Appelbaum, P. C. and Beratis, N. G. Streptococcus pneumoniae in Greece

Tafuri, S., Martinelli, D., Grimaldi, A., Lopatriello, A., Giorgio, V. and Prato, R. (2011) 23-valent New Microbiologica. 34, pp.

Talkington, D. F., Brown, B. G., Tharpe, J. A., Koenig, A. and Russell, H. (1996) Protection of mice against fatal pneumococcal challenge by immunization with pneumococcal surface adhesion A (PsaA).Microbiology

D. F., Crimmins, D. L., Voellinger, D. C., Yother, J. and Briles, D. E. (1991) A 43-kilodalton pneumococcal surface protein, PspA: isolation, protective abilities, and structure analysis of the amino acid-

Tong, H. H., Blue, L. E., James, M. A. and DeMaria, T. F. (2000) Evaluation of the virulence of a Streptococcus deficient mutant in nasopharyngeal colonisation and development of otitis media in

Streptococcus pneumoniae:

, A. J. (1999) Pneumococcal surface protein . 67, pp. 4720-4724.

Page 19: STREPTOCOCCUS PNEUMONIAE: VIRULENCE FACTORS AND THEIR ROLE IN PATHOGENESIS.

All rights reserved This work by Wilolud Journals is licensed under a

Ukpai A. Eze et al.,: Waltman, W. D., McDaniel, L. S., Gray, B. M. and Briles, D. E. (1990) Variation in the molecular weight of PspA (pneumococcal surface protein A) among 69. Whatmore, A. M., Efstratiou, A., Pickerill, A. P., Broughton, K., Woodard, G., Sturgeon, D., George, R. and Dowson, C.G. (2000). Genetic relationships between clinical isolates of Streptococcus oralis, and Streptococcus mitisto S. mitis harboring S. pneumonia virulence factor1382. Whitney, C. G., Farley, M. M., Hadler, J., Harrison, L. H., LexaR., Cetron, M., Zell, E. R., Jorgensen, J. H. and Schuchat, A. (2000) Increasing prevalence of multidrugresistant Streptococcus pneumoniae in the United States. 1924. Yother, J. and Briles, D. E. (1992) Structural properties and evolutionary relationships of PspA, a surface protein of Streptococcus pneumoniae, as revealed by sequence analysis. 609. Yuste, J., Botto, M., Paton, J. C., Holden, D. W. and Brown, J. S. (2005). Additive inhibition of complement deposition by pneumolysin and PspA facilitates 175(3), pp. 1813-1819. Vidyashankara, I., Lei, H., Mangala, RSangeeta B. J. and Russell M. C. (2012) Preformulation Characterization of an Aluminum SaltTrivalent Recombinant Protein-Based Vaccine Candidate Against Pharmaceutical Sciences. 101, pp. 3078 Zhang, J. R., Mostov, K. E., Lamm, M. E., Nanno, M., Schimida, S., Ohwaki, M. and Tuomanen, E. I. (2000) The polymeric immunoglobulin translocates pneumococci across human nasopharyngeal epithelia102, pp. 827-837.

is licensed under a Creative Commons Attribution 3.0 Unported License

45

.,:Continental J. Medical Research 7 (1): 27 -45, 2013

Waltman, W. D., McDaniel, L. S., Gray, B. M. and Briles, D. E. (1990) Variation in the molecular weight of PspA (pneumococcal surface protein A) among Streptococcus pneumoniae. Microbial Pathogenesis

Whatmore, A. M., Efstratiou, A., Pickerill, A. P., Broughton, K., Woodard, G., Sturgeon, D., George, R. and Dowson, C.G. (2000). Genetic relationships between clinical isolates of Streptococcus pneumoniae

occus mitis: characterization of "Atypical" pneumococci and organisms allied virulence factor-encoding genes. Infection and Immunity. 68(3), pp. 1374

Whitney, C. G., Farley, M. M., Hadler, J., Harrison, L. H., Lexau, C., Reingold, A., Lefkowitz, L., Cieslak, P. R., Cetron, M., Zell, E. R., Jorgensen, J. H. and Schuchat, A. (2000) Increasing prevalence of multidrug

in the United States. New England Journal of Medicine. 343(26), pp.

Yother, J. and Briles, D. E. (1992) Structural properties and evolutionary relationships of PspA, a surface , as revealed by sequence analysis. Journal of Bacteriology

Paton, J. C., Holden, D. W. and Brown, J. S. (2005). Additive inhibition of complement deposition by pneumolysin and PspA facilitates Streptococcus pneumonia septicemia. Journal of Immunology

Vidyashankara, I., Lei, H., Mangala, R. L., Reza, E., Christoph, R., Andreas, M., Jeff, M., David, B. V., Sangeeta B. J. and Russell M. C. (2012) Preformulation Characterization of an Aluminum Salt

Based Vaccine Candidate Against Streptococcus pneumonia. 101, pp. 3078–3090.

Zhang, J. R., Mostov, K. E., Lamm, M. E., Nanno, M., Schimida, S., Ohwaki, M. and Tuomanen, E. I. (2000) The polymeric immunoglobulin translocates pneumococci across human nasopharyngeal epithelia

Creative Commons Attribution 3.0 Unported License

Waltman, W. D., McDaniel, L. S., Gray, B. M. and Briles, D. E. (1990) Variation in the molecular weight of Microbial Pathogenesis. 8, pp. 61-

Whatmore, A. M., Efstratiou, A., Pickerill, A. P., Broughton, K., Woodard, G., Sturgeon, D., George, R. and Streptococcus pneumoniae,

: characterization of "Atypical" pneumococci and organisms allied . 68(3), pp. 1374-

u, C., Reingold, A., Lefkowitz, L., Cieslak, P. R., Cetron, M., Zell, E. R., Jorgensen, J. H. and Schuchat, A. (2000) Increasing prevalence of multidrug-

343(26), pp. 1917-

Yother, J. and Briles, D. E. (1992) Structural properties and evolutionary relationships of PspA, a surface Journal of Bacteriology. 174, pp. 601-

Paton, J. C., Holden, D. W. and Brown, J. S. (2005). Additive inhibition of complement Journal of Immunology.

. L., Reza, E., Christoph, R., Andreas, M., Jeff, M., David, B. V., Sangeeta B. J. and Russell M. C. (2012) Preformulation Characterization of an Aluminum Salt-Adjuvanted

Streptococcus pneumoniae. Journal of

Zhang, J. R., Mostov, K. E., Lamm, M. E., Nanno, M., Schimida, S., Ohwaki, M. and Tuomanen, E. I. (2000) The polymeric immunoglobulin translocates pneumococci across human nasopharyngeal epithelial cells. Cell.