Sie sind auf Seite 1von 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/290625343

Pathogenesis of Mycoplasma pneumoniae: An update

Article  in  Indian Journal of Medical Microbiology · January 2016


DOI: 10.4103/0255-0857.174112

CITATIONS READS
13 53

3 authors, including:

Rama Chaudhry Amita Chandolia


All India Institute of Medical Sciences All India Institute of Medical Sciences
223 PUBLICATIONS   2,293 CITATIONS    9 PUBLICATIONS   111 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Microbiological and molecular diagnosis of prosthetic joint infections View project

Metagenomics in Health and Disease View project

All content following this page was uploaded by Amita Chandolia on 10 July 2018.

The user has requested enhancement of the downloaded file.


[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

Indian Journal of Medical Microbiology, (2016) 34(1): 7-16 1

Review Article

Pathogenesis of Mycoplasma pneumoniae: An update


*R Chaudhry, A Ghosh, A Chandolia

Abstract
Genus Mycoplasma, belonging to the class Mollicutes, encompasses unique lifeforms comprising of a small genome
of 8,00,000 base pairs and the inability to produce a cell wall under any circumstances. Mycoplasma pneumoniae is the
most common pathogenic species infecting humans. It is an atypical respiratory bacteria causing community acquired
pneumonia (CAP) in children and adults of all ages. Although atypical pneumonia caused by M. pneumoniae can be
managed in outpatient settings, complications affecting multiple organ systems can lead to hospitalization in vulnerable
population. M. pneumoniae infection has also been associated with chronic lung disease and bronchial asthma. With the
advent of molecular methods of diagnosis and genetic, immunological and ultrastructural assays that study infectious
disease pathogenesis at subcellular level, newer virulence factors of M. pneumoniae have been recognized by researchers.
Structure of the attachment organelle of the organism, that mediates the crucial initial step of cytadherence to respiratory
tract epithelium through complex interaction between different adhesins and accessory adhesion proteins, has been
decoded. Several subsequent virulence mechanisms like intracellular localization, direct cytotoxicity and activation of the
inflammatory cascade through toll-like receptors (TLRs) leading to inflammatory cytokine mediated tissue injury, have also
been demonstrated to play an essential role in pathogenesis. The most significant update in the knowledge of pathogenesis
has been the discovery of Community-Acquired Respiratory Distress Syndrome toxin (CARDS toxin) of M. pneumoniae
and its ability of adenosine diphosphate (ADP) ribosylation and inflammosome activation, thus initiating airway
inflammation. Advances have also been made in terms of the different pathways behind the genesis of extrapulmonary
complications. This article aims to comprehensively review the recent advances in the knowledge of pathogenesis of this
organism, that had remained elusive during the era of serological diagnosis. Elucidation of virulence mechanisms of M.
pneumoniae will help researchers to design effective vaccine candidates and newer therapeutic targets against this agent.

Key words: Epidemiology, Mycoplasma pneumoniae, pathogenesis

Introduction emerging pathogens.[4] Studies have demonstrated that


the atypical bacteria rank as the third or fourth leading
Community-acquired pneumonia (CAP) continues organisms causing CAP, being present in about 22–40%
to be a significant cause of mortality and morbidity of all CAP patients, often as co-pathogens with other
worldwide, particularly in the elderly population and bacteria.[5-7] In a multi-centric study involving 12 medical
under five children.[1,2] The mortality rate of CAP can centres across Asia, atypical respiratory bacteria were
rise to 10% in hospital ward admitted patients and can noted in more than 23% of CAP cases. M. pneumoniae was
exceed 30% in Intensive Care Unit patients requiring detected in approximately 12% of all CAP cases and was
mechanical ventilation and prolonged hospitalisation.[3] Of the most common atypical bacteria isolated.[8] In paediatric
all aetiological agents of CAP, atypical respiratory bacteria population, M. pneumoniae account for approximately
viz., Mycoplasma pneumoniae, Chlamydia pneumoniae 10–40% of all lower respiratory tract infections.[8,9]
and Legionella spp. are increasingly being recognised as
Infection with M. pneumoniae is mostly encountered
in outpatient setting. However, it is a significant cause
*Corresponding author (email: <drramach@gmail.com>) of hospitalisation also due to pneumonia, especially in
Department of Microbiology (RC, AG, AC), AIIMS, elderly population and immunocompromised patients.[10]
New Delhi, India
Manifestations of M. pneumoniae infection can range from
Received: 30-09-2014
Accepted: 08-09-2015
self-limiting upper respiratory illness to severe pneumonia.

This is an open access article distributed under the terms of the Creative
Access this article online
Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows
Quick Response Code: Website: others to remix, tweak, and build upon the work non-commercially, as long as the
www.ijmm.org author is credited and the new creations are licensed under the identical terms.
For reprints contact: reprints@medknow.com
PMID:
***
How to cite this article: Chaudhry R, Ghosh A, Chandolia A.
DOI: Pathogenesis of Mycoplasma pneumoniae: An update. Indian J Med
10.4103/0255-0857.174112 Microbiol 2016;34:7-16.

© 2016 Indian Journal of Medical Microbiology Published by Wolters Kluwer - Medknow


[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

8 Indian Journal of Medical Microbiology vol. 34, No. 1

Children <5 years of age usually suffer from mild upper and mRNA expression systems. In this article, we try to
respiratory symptoms while older children and adolescents comprehensively review the pathogenesis of M. pneumoniae
develop bronchopneumonia, requiring hospitalisation.[11,12] highlighting the recent advances in terms of cytadherence,
M. pneumoniae has been detected in higher proportions in cytotoxic and inflammatory potential and the pathogenic
respiratory samples of adults and children with asthmatic role of CARDS toxin. We also discuss about the updates in
attacks or exacerbation, as compared to healthy controls.[13] molecular pathogenesis of extrapulmonary manifestations of
High rates of carriage of M. pneumoniae in the airways of M. pneumoniae infection.
chronic stable asthmatics also point towards the association
of M. pneumoniae with asthma.[14] In a study from our Cytadherence
centre too, there was statistically significant association The initial step of M. pneumoniae respiratory tract
between M. pneumoniae infection and children with severe infection involves cytadherence of the organism to the
persistent asthma and acute exacerbation of asthma in ciliated columnar epithelium of the respiratory tract, which
previously diagnosed asthmatic children.[15] However, it protects the organism from mucociliary clearance and local
is only in the recent studies, significant progress has been cytotoxic effects.[12] Cellular adherence to sialoglycoproteins
made in establishing the mechanism by which this organism and sulphated glycolipids is mediated by a specialised
can cause or exacerbate asthma, an issue that had remained organelle, the structure of which has been delineated by
debatable over the years.[16] electron microscopy. It consists of a central core composed
Mycoplasmas, which belong to the bacterial of dense filaments and a tip-like structure composed of
class  Mollicutes, are cell-wall deficient smallest adhesins and accessory proteins.[24,25] The major proteins,
self-replicating organisms capable of cell free that have been experimentally proven directly involved
survival.[17] The genome of M. pneumoniae is small in receptor binding, are the 170 kilodalton P1-adhesin
(approximately 816 kilo base-pairs) which accounts for and the P30-adhesin. High molecular weight protein-1
its limited biosynthetic capabilities and slow replication (HMW-1), HMW-2, HMW-3 and proteins A, B and C
rate.[18] Thus, a major obstacle in understanding the act as accessory proteins, interacting with P1 and P30 and
pathogenic role of this organism has been the lack of assisting in cytadherence.[26,27] A schematic representation of
knowledge of its biological properties due to difficulties M. pneumoniae adherence organelle is provided in Figure 1.
associated with cultivation of this organism. P1-adhesin
Early researches that have counted the role of The P1-adhesin is a transmembrane protein,
M. pneumoniae in the pathogenesis of different clinical concentrated primarily at the tip of the attachment organelle
entities depended only on serological assays which were of M. pneumoniae. Mutant strains lacking P1-adhesin fail
less specific and could not provide a definite association to adhere to animal cells and are avirulent.[28,29] In vitro
with disease pathogenesis.[12] Since, IgM antibodies studies have demonstrated that P1-adhesin is also involved
may not be present early in the course of infection, in gliding motility of the organism that may help in cell
diagnosis based only on serology may not be accurate. to cell transfer. Repeated gliding motility allows the
In fact, seroprevalence of M. pneumoniae in adults with organism to bind and release itself from the attaching
pneumoniae has been found to be highly variable ranging surface which eventually increases the infective surface
from 1.9% to over 30%.[19] Moreover, simultaneous
detection of co-pathogens, which included respiratory
viruses and other bacteria, often considered M. pneumoniae
as an innocent bystander organism.[20,21] Advances in
molecular diagnosis such as nucleic-acid amplification tests
(NAATs), sequencing and proteomic studies have helped us
to gain knowledge about the pathogenesis of this organism
that had remained elusive.[22,23] In the past few years,
potential pathogenic factors associated with this organism
have been studied in details using molecular methods
by researchers who had looked into different aspects of
pathogenesis such as complex adherence mechanism
of M. pneumoniae, the community-acquired respiratory
distress syndrome toxin (CARDS toxin) that activate
the inflammasomes and the genesis of extrapulmonary
complications. The inflammatory pathways leading to Figure 1: Mycoplasma pneumoniae cytadherence organelle with
tissue injury following M. pneumoniae infection have adherence proteins (Reproduced from Frontiers in Bioscience 2007;
been elucidated using highly sensitive cytokine assays 12: 690-699 by R Chaudhry et al.)

www.ijmm.org
[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

January-March 2016 Chaudhry, et al.: M. pneumoniae pathogenesis update 9

area. Anti-P1 monoclonal antibody had inhibitory effects Free radical mediated cellular damage is one of the
on gliding motility and adherence to tracheal rings pathways by which M. pneumoniae affects respiratory
by M. pneumoniae.[30] epithelium. Oxidative stress as an important step in
pathogenesis of M. pneumoniae results from the hydrogen
Studies by Kenri et al. and Su et al. have shown that there peroxide and superoxide radicals generated by the
are variable regions on the P1-adhesin protein attributed organisms as well as by the host immune system.[40]
to the repetitive sequences of this cytadhesin gene.[31,32] Superoxide radicals also inhibit host cell catalase, thereby
According to Jacobs, the adherence mediating domains of augmenting the effect of free radical damage.[41] Sun et al.
P1-adhesin are highly conserved while the immunodominant
in 2008 demonstrated that M. pneumoniae infection of
epitopes are variable. Thus, antibodies which are
A549 human lung carcinoma cell line led to generation of
predominantly against the immunodominant epitopes, fail to
reactive oxygen species (ROS). Generation of ROS induced
block the antigenically distinct cytadherent protein, allowing
changes in proteomic profile of the cell line in terms of
the organism to evade antibody response.[33] In a recent study
exposure of host oxidative stress response enzymes such as
from our centre, we have shown that immunodominant
glucose-6-phosphate dehydrogenase, nicotinamide adenine
regions are actually distributed throughout the length of P1-
dinucleotide (NAD) dehydrogenase and also caused DNA
protein. Only the amino and carboxy-terminals of P1-protein
double-stranded breaks.[42] Similar role of ROS in mediating
are surface exposed and antibodies directed to these regions
lung injury by M. pneumoniae has been demonstrated in
blocked adherence of M. pneumoniae to Hep-2 cell line.
mice. Mycoplasma infection in mice caused impairment of
Antibodies to the middle part failed to block cytadhesion.[34]
alveolar ion transport leading to decreased alveolar fluid
P30-adhesin clearance due to ROS-mediated damage to epithelial sodium
channels.[43]
The other important protein involved in adherence of
M. pneumoniae is the P30-adhesin. It is a transmembrane Pulmonary injury due to M. pneumoniae infection
protein present in cluster at the attachment organelle tip. has also been attributed to the host inflammatory
Additional function of P30 includes gliding motility and co- response. M. pneumoniae infection is characterised by
ordination of cell division with biogenesis of the attachment peribronchiolar alveolar infiltration with neutrophils,
organelle.[35,36] lymphocytes and plasma cells.[44] In vitro studies in human
lung epithelial carcinoma cells have shown increased levels
Accessory adhesion proteins of proinflammatory cytokines-like interleukin-8 (IL-8)
Interactions of major adhesin proteins with the accessory and tumour necrosis factor α (TNF-α) in culture media
proteins such as HMW-1, HMW-2, HMW-3 and protein-A, and cellular expression of IL-1β mRNA. These cytokines
protein-B and protein-C help in establishing the complete act as potent chemoattractants for inflammatory cells.
attachment organelle structure. Mutations in genes encoding Increased levels of these proinflammatory cytokines result
the accessory proteins lead to cytadherence defects.[36,37] from the activation of pattern recognition receptors (PRRs)
Two other proteins viz., P65 and P116 have recently namely toll-like receptor-1 (TLR-1), TLR-2 and TLR-6 by
been shown to be present at the distal end of the terminal M. pneumoniae membrane proteins.[45] Lipid-associated
organelle, which probably act by interacting with P1 and membrane protein or a dipalmitoylated lipoprotein of
P30 adhesins thus assisting in attachment.[38,39] However, M. pneumoniae activates TLR-1, TLR-2 and TLR-6 leading
further role of these accessory proteins in pathogenesis of to activation of monocytic nuclear factor-κβ (NF-κβ), a
M. pneumoniae needs to be studied for better understanding key regulator of proinflammatory cytokine release.[45,46] In
of interactions between host cells and the organism. mice models it was demonstrated that a previous challenge
The adhesion proteins may serve as potential targets for with M. pneumoniae extracts actually upregulated TLR-2
therapeutic and vaccine strategies against M. pneumoniae. expression in alveolar macrophages which resulted in a
magnified inflammatory response to subsequent challenges
To summarise, the adhesins of M. pneumoniae through by the same extract.[45]
their complex interactions allow close association of the
organism with host cells which is followed by cytotoxicity Thus, a major pathway of M. pneumoniae pathogenesis
and inflammation. Intracellular localisation may occur in is induction of inflammation via the TLR-mediated
M. pneumoniae before it induces cytotoxicity. However, cytokine release and generation of free radicals. In
the extent of intracellular localisation is not known in vivo. addition an exaggerated innate immune response in
The fusion of Mycoplasma cell membrane with the host M. pneumoniae infection can be due to a positive
cell enabling intracellular location may help in establishing feedback effect of previous M. pneumoniae colonisation
latent or chronic states, avoiding immune response and or asymptomatic infection. This recent update in the
impairing drug therapy. concept of immunopathogenesis of M. pneumoniae will
help researchers to target potential molecules such as
Cytotoxicity and Inflammation the membrane lipoproteins and other TLR ligands for
www.ijmm.org
[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

10 Indian Journal of Medical Microbiology vol. 34, No. 1

modulating high levels of airway inflammation and prevent broth culture of inherently slow growing mycoplasmas.[53]
lung injury due to M. pneumoniae. Treatment of Chinese hamster ovary cells or HEp-2 cells
with the recombinant toxin showed its ability to transfer
Community-acquired Respiratory Distress Syndrome ADP-ribosyl group from nicotinamide adenine diphosphate
Toxin (NAD+) to amino acids of cellular protein. The ribosylating
Community-acquired respiratory distress syndrome toxin: property of CARDS toxin resulted in alteration of the
Background protein targets including enzymes of host cell metabolic
pathways which lead to cellular toxicity. Cytopathic effects
Early studies in the 1970s and 1980s had tried to describe in the form of vacuolisation, cellular rounding and cellular
the pathology of M. pneumoniae infection using organ culture distortion were also exhibited.[53] In organ culture of
and cell culture systems.[47,48] Infection of hamster tracheal baboon tracheal rings, recombinant CARDS toxin caused
culture was characterised by ciliostasis and cytoplasmic loss of ciliary function of respiratory epithelium, intensive
vacuolisation followed by aggregation of intracellular cytoplasmic vacuolisation, karyopyknosis and disruption
vacuoles leading to cytoplasmic distortion and cell of epithelial integrity.[54] These toxin-mediated progressive
damage.[48] Based on the cytotoxic effects of M. pneumoniae cytotoxic changes in tracheal rings were partly explanatory
infection, researchers hypothesised that a Mycoplasma for the mechanism of pathogenesis of airway disease of
membrane associated toxic factor could be responsible for M. pneumoniae which had remained elusive over the years.
pathogenesis.[49] Toxic effects of free radical generation
could only partially explain such effects of M. pneumoniae CARDS toxin: Role in pathogenesis
virulence.[40,50] In 1975, Hu et al. demonstrated the inhibition
Effect of the recombinant toxin on the respiratory
of RNA and protein synthesis along with decreased uptake
tract has also been studied in mice and primate models.
of metabolic substrates in M. pneumoniae infected host cells
Intranasal inoculation in BALB/c mice induced airway
which could not be explained by cytadherence process of the
mucous production with extensive peribronchiolar and
organism and effects of generation of ROS. Hu et al. thus,
perivascular inflammation. Along with transient neutrophilia
suggested that the primary detrimental effect on the host cell
followed by significant increase in eosinophils at day 7 of
was at the transcriptional or translational level. The fact that
exposure in the broncho-alveolar lavage fluid (BALF).[55]
cytopathic effects of M. pneumoniae infection could only
Hardy et al. demonstrated that the early cytokine response
be reversed by early addition of erythromycin to the organ
in mice was predominantly proinflammatory which
culture (within 24 h of infection) pointed to the conclusion
included IL-1, IL-6, IL-12 and TNF-α. In baboons toxin
that the organism required to synthesise a certain protein or
inoculation led to increase in chemokines such as regulated
toxin to mediate host cell injury.[48]
on activation, normal T cell expressed and secreted, IL-8,
Subsequently, Kannan et al. in their attempt to interferon-γ (IFN-γ) and granulocyte-colony stimulating
characterise the virulence factors of M. pneumoniae factor in addition to proinflammatory cytokines, closely
observed calcium dependent, trypsin sensitive binding resembling human M. pneumoniae infection.[56] Significant
M. pneumoniae to human surfactant protein-A. Surfactant increases in expression of TH2 cytokines viz., IL-4 and
protein-A binding of M. pneumoniae was found to be a IL-13 and chemokines, chemokine (C-C motif) ligand-7
crucial factor in the colonisation of the respiratory tract by (CCL-7) and CCL-32, were also demonstrated using
the organism.[51] This protein of M. pneumoniae, required for quantitative reverse transcriptase-polymerase chain
binding, was identified by affinity chromatography followed reaction (PCR) of M. pneumoniae infected mice BALF
by purification and sequencing. The 68-kDa protein, which by Medina et al.[57] Recombinant CARDS toxin exposure
was different from adhesin, cytadherence-associated resulted in exaggerated TH2 inflammatory response in
proteins or fibronectin binding proteins, was initially the respiratory tract of mice, previously sensitised with
termed as MPN 372. It shared amino acid homology at ovalbumin. Invasive pulmonary measurements in infected
the catalytic site with that of the S1 subunit of Bordetella mice recorded increased airway resistance and decreased
pertussis toxin. The MPN 372 protein was found to catalyse lung compliance due to increased airway reactivity.[57]
adenosine diphosphate ribosylation (ADP-ribosylation), Thus, in animal models CARDS toxin exposure induced a
similar to Bordetella pertussis toxin S1 subunit, and possess proinflammatory response in the respiratory tract.
vacuolating function in infected cells.[51] Using mammalian
cell lines and baboon tracheal organ culture, Kannan and Chemoattractant activities of CCL-17 and CCL-22
Baseman confirmed the virulent properties of the protein leading to lymphocyte and eosinophil recruitment and
and gave it the name CARDS toxin.[52] increased expression of IL-4 and IL-13, eosinophil mediated
pulmonary inflammation and increased airway reactivity
Recombinant CARDS toxin was synthesised by are determining factors in the pathogenesis of asthma
expression in Escherichia coli to overcome the problem of in humans. A  similar cytokine and cellular profile of the
isolating the toxin in minute or insufficient quantities in the respiratory tract secretions are induced by CARDS toxin in

www.ijmm.org
[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

January-March 2016 Chaudhry, et al.: M. pneumoniae pathogenesis update 11

mice as well as primates such as baboons. These findings has been shown to be internalised into mammalian cells via
point towards a role of this toxin in asthmatic exacerbation clathrin-mediated endocytosis.[64] Hence, following cellular
and further confirm the association between M. pneumonia entry CARDS toxin mediated activation of inflammasomes
infection and asthma. in alveolar macrophages can a potent mechanism of release
of IL-1β, a pro-inflammatory cytokine, into the airways. In
Community-acquired respiratory distress syndrome toxin fact, higher levels of IL-1β has been detected in respiratory
and inflammasome samples of symptomatic asthmatic patients compared to
A recent discovery about M. pneumoniae pathogenesis has healthy controls.[65] Thus, CARDS toxin of M. pneumoniae
been the elucidation of the pathway, by which CARDS toxin by its ability to activate inflammasome can act as a trigger in
induces inflammation through regulation of inflammasome initiating airway inflammation recapitulating the events the
activity Figure 2. A component of the innate immune response occurs in asthma However, the hypothesis of M. pneumoniae
system in mammals, the inflammasome comprises of a CARDS toxin being a causative agent or a triggering factor
multiprotein complex present in the cell cytoplasm and acts of asthma still remains to be tested in human models.
by activation of the enzyme caspase-1 which in turn cleaves Pathogenesis of Extrapulmonary Manifestations
pro-IL-1β to active IL-1β.[58] The IL-1β is an inflammatory
cytokine, which through activation of nuclear factor Respiratory infections with M. pneumoniae is often
kappa-light-chain-enhancer of activated B cells (NF-κB) complicated in as many as 25% of cases by the involvement
dependent pathways amplifies inflammatory responses of various extrapulmonary systems such as nervous system,
against infectious agents.[59] However, over-activaion of cardiovascular system, bones and joints, kidney, skin
inflammasomes after infection can cause tissue injury due to and mucosa. However, extrapulmonary manifestations
‘hyperinflammation’.[58,60] Allergen induced inflammasome can occur even in the absence of pneumonia.[66,67] A
activation also leading to lung injury and lung remodelling, retrospective study by Pönkä had shown that neurological
which has been implicated in the pathogenesis asthma and complications, in absence of any respiratory symptoms,
chronic obstructive pulmonary disease.[61] can occur in up to 18% of M. pneumoniae infected cases.[68]
Pathogenic mechanisms of extrapulmonary complications
Nucleotide-binding oligomerization domain leucine- have been partly elucidated and further studies are for
rich repeats containing receptors (NLRs) are intracellular complete understanding of these processes. Complications
PRRs, present in a wide variety of cells viz., lymphocytes, of central and peripheral nervous systems rank as the most
macrophages and dendritic cells. The NLRs act as sensors common of all extrapulmonary complications and can
for pathogen associated molecular patterns (PAMPs). The occur in approximately 1–7% of serologically confirmed
most well-characterised NLR that is, NLRP-3, which is M. pneumoniae hospitalised for respiratory illnesses.[69,70]
expressed in myeloid cells, complexes with inflammasome Initial studies on central nervous system (CNS)
after being upregulated in response to PAMPs. This NLRP- complications were primarily based on serological diagnosis
3-inflammososme complex can activate caspase-1 which in of M. pneumoniae infection and neural tissue damage was
turn leads to IL-1β release.[60] The CARDS toxin co-localizes thought to be due to autoimmune mechanisms or due to
with NLRP-3-inflammasome within mice bone-marrow other pathogens in presence of a false positive Mycoplasma
derived macrophages in vitro and by its ADP-ribosylating serology.[66,71] However, detection of Mycoplasma nucleic-
property of NLRP-3 activates the inflammasome followed acid by NAATs in neurological specimen has changed
by release of IL-1β.[62,63] The M. pneumoniae CARDS toxin the perspective. M. pneumoniae associated neurological
complications are now hypothesised to be caused by direct
inflammatory injury due to cytokines induced by the
organism, generation of antibodies cross-reactive with host
tissue components and vascular occlusion due to vasculitis
and/or thrombosis.[72] Interplay between these mechanisms
lead to generation of neurological symptoms.

The most common form neurological complication


is encephalitis, more common in children <10 years of
age than in adults.[72] Up to 60% of paediatric encephalitis
patients end up with neurological sequelae.[73] Other
neurological manifestations include aseptic meningitis,
meningoencephalitis, acute transverse myelitis, acute
disseminating encephalomyelitis (ADEM), Guillain–Barré
syndrome (GBS) and stroke.[74] As proposed by Narita,
Figure 2: Schematic representation of the pathway for M. pneumoniae encephalitis due to M. pneumoniae can be classified into
CARDS toxin mediated inflammation early-onset (onset of neurological symptoms within 7 days
www.ijmm.org
[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

12 Indian Journal of Medical Microbiology vol. 34, No. 1

of onset of fever) and late-onset (onset of neurological polyradiculopathies and optic neuritis. Both GM1
symptoms at 8 days or after onset of fever) varieties.[75] ganglioside and galactocerebroside epitopes have been
Bitnun et al. and Socan et al. have also supported such a detected on M. pneumoniae cell surface that cross-reacts
classification in their studies.[76,77] It has been hypothesised with host myelin glycolipid moieties expressed in neuronal
that early-onset encephalitis is the result of cytokine tissues. Cross-reacting antibodies are either intrathecally
mediated direct neural tissue injury while late-onset produced or cross the blood-brain barrier that become
encephalitis is due to autoimmune mechanisms.[75] permeable during cytokine-mediated inflammation by direct
effect of the organism.[84,85]
M. pneumoniae was isolated from the autopsied brain
specimen of a 30-year-old woman with encephalitis, as early Autoantibodies to lipid-associated neural antigens in
as 1980. Isolation of this organism from lung, kidney and patients with Mycoplasma induced CNS disease were
trachea of this patient was the evidence of dissemination first described by Biberfield in 1971.[86] Subsequently,
of this organism.[66,78] Subsequently, M. pneumoniae anti-galactocerebroside and anti-GM1 ganglioside
nucleic-acid was detected by PCR and nucleic-acid antibodies were demonstrated in CSF of patients with post
hybridisation from brain tissues of patients with early-onset M. pneumoniae CNS dysfunction. In these cases CNS
encephalitis following pneumonia.[79] In a case series of manifestations were preceded by respiratory symptoms
patients with features of encephalitis, meningoencephalitis by 8 days or more which coincides with the generation
and ADEM, antibodies against M. pneumoniae were of these antibodies.[87,88] Approximately 5–15% GBS
detected in 74% of cases in the cerebrospinal fluid (CSF) cases are associated with a preceding M. pneumoniae
PCR.[80] Even M. pneumoniae antigen has been detected by infection and (GBS) patients with evidence of recent
immunohistochemistry in macrophages from perivascular M. pneumoniae infection have been shown to be more
infiltrates of cerebral hemispheres, medulla oblongata and likely positive for anti-galactocerebroside and anti-GM1
spinal cord in patients positive for M. pneumoniae DNA ganglioside antibodies than other GBS patients.[89,90,91] The
in tracheobronchial secretions.[81] Since, M. pneumoniae antigen-antibody complexes activates the complement
positive serology or culture or PCR of respiratory tract cascade and formation of membrane-attack complex
samples cannot differentiate between colonisation and leading to demyelination. Since most of the GBS cases have
infection, demonstration of the organism by PCR or culture been diagnosed by serological testing for M. pneumoniae
or antigen detection from CSF samples or detection of following respiratory illness, establishing a definite causal
intrathecal antibodies have been the key factor behind the relationship is difficult in absence of a positive culture or a
hypothesis of dissemination and direct tissue injury by PCR result.
the organism. In fact, other Mycoplasma species have the
ability to infect brain tissue of animals such as Mycoplasma Anti-galactocerebroside C and antiganglioside
galliseptica in birds, Mycoplasma pulmonis in rodents and antibodies such as anti-GQ1B have also been detected in
Mycoplasma hyopneumoniae in pigs.[12] serum and CSF of patients with acute meningoencephalitis
and Bickerstaff’s brain-stem encephalitis following
Dissemination to distant tissues is the earliest step in M. pneumoniae infection.[92] However, association of
the pathogenesis of neurological complications due to M. pneumoniae infection and pathogenesis of these
M. pneumoniae. It has been shown that M. pneumoniae disorders remains unclear and further studies are required.
gets passively transferred through gaps between injured Role of these antibodies in CNS complications has
respiratory epithelial cells and probably reaches by been questioned in a study by Biberfeld et al. in which
circulating in blood in a cell-free form.[72,82] Tissue injury is 80% of M. pneumoniae infected individuals without
mediated by local production of cytokine by the interaction CNS manifestations had these circulating antibodies.[93]
of M. pneumoniae glycolipid antigen and TLR-1, TLR-2 However, further evidence of molecular mimicry of
and TLR-6 of inflammatory cells leading to activation of M. pneumoniae has been provided by in vitro studies, where
cytokine cascade. The two major cytokine that have been anti-P1-adhesion antibodies cross-reacted with intracellular
found to be elevated intrathecally in early-onset encephalitis enzymes viz., glyceralde-3-phosphate dehydrogenase and
has been IL-6 and IL-8, of which IL-8 is a potent neutrophil 2-phospho D-glycerate hydrolase in eukaryotic cell lines.[86]
chemoattractant. Neural tissue injury is probably due to
inflammatory cell-mediated damage. In contrast to bacterial The third mechanism for CNS manifestations has been
meningoencephalitis due to other organisms, elevated levels proposed to be vascular occlusion. Vascular occlusion
of IFN-γ and TNF-α were not observed. Another cytokine, is caused by local vascular injury due to cytokines and
IL-18 has been found to be elevated in CSF in patients with chemokines due to M. pneumoniae infection.[72,75] This form
late-onset encephalitis due to M. pneumoniae.[75,83] of ischaemic injury to CNS occurs in absence of thrombotic
mechanism and is the main pathophysiology for bilateral
Autoimmunity is the proposed mechanism of striatal necrosis following M. pneumoniae infection.
pathogenesis for M. pneumoniae associated late-onset However, considering stroke in children and adults in whom
encephalitis along with acute transverse myelitis, GBS, M. pneumoniae DNA has been detected in CSF, the cause
www.ijmm.org
[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

January-March 2016 Chaudhry, et al.: M. pneumoniae pathogenesis update 13

still remains to be elucidated. organism.

M. pneumoniae infection has also been associated Conclusion


with dermatological complications in 1–5% cases,
manifested by erythema multiforme and Mycoplasma Although majority of patients with M. pneumoniae
induced rash and mucositis (MIRM), which includes mild infection can be treated in outpatient or ambulatory setting,
erythematous maculo-papular rash, vesicular eruptions, severe infections in susceptible population may require
toxic epidermal necrolysis and rarely Steven–Johnson hospitalisation. Moreover, extrapulmonary manifestations,
syndrome.[94] Systematic corticosteroid therapy and/or particularly the neurological complications, can lead to poor
intravenous immunoglobin may be required in some cases. outcome in infected patients.
Cause of MIRM has been attributed to the molecular Our knowledge regarding the pathogenesis of this
mimicry between Mycoplasma P1-adhesin molecule and organism has improved ever since the introduction of
keratinocyte antigen leading to generation of cross-reacting highly sensitive molecular assays. Newer cytadherence
antibodies, immune-complex formation and complement proteins have been discovered, immunological pathways
activation. Erythema multiforme following M. pneumoniae leading to tissue injury have been elucidated and researchers
infection is hypothesised to be a type-IV hypersensitivity have also characterised the novel CARDS toxin molecule.
reaction or a cytotoxic damage through a Fas Ligand There have been significant advances in the knowledge
and granulysin mediated perforin-granzyme pathway.[95] of immunopathogenesis in terms of the role of TLRs
Haematologic manifestations of M. pneumoniae infection and inflammasomes, which can be used as potential
include haemolytic anaemia due to autoimmune cold therapeutic targets in modulation of the disease process
agglutinins and thrombotic thrombocytopenic purpura eg. inhibitors or antibodies against relevant peptides.
due to cross-reactive antibodies inactivating plasma von However, most of the studies have been performed in cell
Willebrand factor-cleaving protease.[12,96] For other systemic lines or in animal models. Hence, the authors feel that
complications, additional studies are required to elucidate for designing a vaccine which will produce high levels of
the pathological pathways. protective immunity in susceptible population, larger studies
Drug Resistance: An Emerging Problem regarding the pathogenesis at subcellular levels need to be
undertaken in humans. Such studies will further improve
Effective treatment includes macrolides, the our understanding about the disease process, particularly
antimicrobial of choice and tetracycyclines and in terms of association of the organism with chronic lung
fluoroquinolones. However, high rates of macrolide diseases and debilitating extrapulmonary complications.
resistance have been reported from Asian countries A holistic approach of studying the molecular virulence
such as China and Japan. More than 90% of Chinese properties of the organism will not only help in designing
and approximately 87% of Japanese isolates have been an effective vaccine but can also lead to development of
shown to be macrolide resistant.[97,98] Macrolide resistance newer therapeutic modalities and better diagnostic assays
strains are also emerging from other parts of the world enabling us in comprehensive management of infections due
including USA and European countries such as Germany to M. pneumoniae.
and France.[99,100] Macrolide resistance in M. pnuemoniae
has been attributed to nucleotide mutations in the 23S Financial support and sponsorship
rRNA genes which have led to increased minimal Nil.
inhibitory concentrations to erythromycin, azithromycin
and clarithromycin. Although resistance to quinolones or Conflicts of interest
tetracyclines among clinical isolates of M. pneumoniae
has not been reported, considering the side effects of There are no conflicts of interest.
tetracycline therapy and fluoroquinolones not being
References
recommended for use in children, an effective vaccine
against M. pneumoniae is desirable. Developments of a 1. Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K,
successful vaccine will not only enable us to prevent the Aboyans V, et al. Global and regional mortality from
serious complications of M. pneumoniae infection but also 235 causes of death for 20 age groups in 1990 and 2010:
act as a preventive strategy against outbreaks, particularly A systematic analysis for the Global Burden of Disease Study
in closed community settings. Inactivated vaccines have 2010. Lancet 2012;380:2095-128.
2. Nair H, Simões EA, Rudan I, Gessner BD,
been tested in clinical trials and the summarised efficacy
Azziz-Baumgartner E, Zhang JS, et al. Global and regional
was only 40% approximately against M. pneumoniae burden of hospital admissions for severe acute lower
associated pneumonia.[101] Hence, further efforts for respiratory infections in young children in 2010: A systematic
developing a more efficacious, yet safe, vaccine against M. analysis. Lancet 2013;381:1380-90.
pneumoniae should continue based on the recent advances 3. Nair GB, Niederman MS. Community-acquired pneumonia:
in knowledge regarding the virulence properties of this An unfinished battle. Med Clin North Am 2011;95:1143-61.
www.ijmm.org
[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

14 Indian Journal of Medical Microbiology vol. 34, No. 1

4. File TM Jr, Tan JS, Plouffe JF. The role of atypical pathogens: 22. Nilsson AC, Björkman P, Persson K. Polymerase chain
Mycoplasma pneumoniae, Chlamydia pneumoniae, and reaction is superior to serology for the diagnosis of acute
Legionella pneumophila in respiratory infection. Infect Dis Mycoplasma pneumoniae infection and reveals a high rate of
Clin North Am 1998;12:569-92. persistent infection. BMC Microbiol 2008;8:93.
5. Woodhead M. Community-acquired pneumonia guidelines 23. Thurman KA, Walter ND, Schwartz SB, Mitchell SL,
– An international comparison: A view from Europe. Chest Dillon MT, Baughman AL, et al. Comparison of laboratory
1998;113:183S-7S. diagnostic procedures for detection of Mycoplasma
6. Ruiz-González A, Falguera M, Nogués A, Rubio-Caballero M. pneumoniae in community outbreaks. Clin Infect Dis
Is Streptococcus pneumoniae the leading cause of pneumonia 2009;48:1244-9.
of unknown etiology? A microbiologic study of lung aspirates 24. Krivan HC, Olson LD, Barile MF, Ginsburg V, Roberts
in consecutive patients with community-acquired pneumonia. DD. Adhesion of Mycoplasma pneumoniae to sulfated
Am J Med 1999;106:385-90. glycolipids and inhibition by dextran sulfate. J Biol Chem
7. Vergis EN, Yu VL. Macrolides are ideal for empiric therapy 1989;264:9283-8.
of community-acquired pneumonia in the immunocompetent 25. Roberts DD, Olson LD, Barile MF, Ginsburg V, Krivan HC.
host. Semin Respir Infect 1997;12:322-8. Sialic acid-dependent adhesion of Mycoplasma pneumoniae to
8. Ngeow YF, Suwanjutha S, Chantarojanasriri T, Wang F, purified glycoproteins. J Biol Chem 1989;264:9289-93.
Saniel M, Alejandria M, et al. An Asian study on the prevalence 26. Krause DC. Mycoplasma pneumoniae cytadherence:
of atypical respiratory pathogens in community - Acquired Unravelling the tie that binds. Mol Microbiol 1996;20:247-53.
pneumonia. Int J Infect Dis 2005;9:144-53. 27. Krause DC, Leith DK, Wilson RM, Baseman JB.
9. Meyer Sauteur PM, Jacobs BC, Spuesens EB, Jacobs E, Identification of Mycoplasma pneumoniae proteins
Nadal D, Vink C, et al. Antibody responses to Mycoplasma associated with hemadsorption and virulence. Infect Immun
pneumoniae: Role in pathogenesis and diagnosis of 1982;35:809-17.
encephalitis? PLoS Pathog 2014;10:e1003983. 28. Kahane I, Tucker S, Leith DK, Morrison-Plummer J,
10. Marston BJ, Plouffe JF, File TM Jr, Hackman BA, Baseman JB. Detection of the major adhesin P1 in triton
Salstrom SJ, Lipman HB, et al. Incidence of community- shells of virulent Mycoplasma pneumoniae. Infect Immun
acquired pneumonia requiring hospitalization. Results of 1985;50:944-6.
a population-based active surveillance Study in Ohio. The 29. Kahane I. In vitro studies on the mechanism of adherence
Community-Based Pneumonia Incidence Study Group. Arch and pathogenicity of mycoplasmas. Isr J Med Sci
Intern Med 1997;157:1709-18. 1984;20:874-7.
11. Foy HM. Infections caused by Mycoplasma pneumoniae and 30. Seto S, Kenri T, Tomiyama T, Miyata M. Involvement of P1
possible carrier state in different populations of patients. Clin adhesin in gliding motility of Mycoplasma pneumoniae as
Infect Dis 1993;17:S37-46. revealed by the inhibitory effects of antibody under optimized
12. Waites KB, Talkington DF. Mycoplasma pneumoniae gliding conditions. J Bacteriol 2005;187:1875-7.
and its role as a human pathogen. Clin Microbiol Rev 31. Kenri T, Taniguchi R, Sasaki Y, Okazaki N, Narita M,
2004;17:697-728. Izumikawa K, et al. Identification of a new variable sequence
13. Rollins DR, Good JT Jr, Martin RJ. The role of atypical in the P1 cytadhesin gene of Mycoplasma pneumoniae:
infections and macrolide therapy in patients with asthma. Evidence for the generation of antigenic variation by DNA
J Allergy Clin Immunol Pract 2014;2:511-7. recombination between repetitive sequences. Infect Immun
14. Martin RJ, Kraft M, Chu HW, Berns EA, Cassell GH. A link 1999;67:4557-62.
between chronic asthma and chronic infection. J Allergy Clin 32. Su CJ, Chavoya A, Dallo SF, Baseman JB. Sequence
Immunol 2001;107:595-601. divergency of the cytadhesin gene of Mycoplasma
15. Varshney AK, Chaudhry R, Saharan S, Kabra SK, pneumoniae. Infect Immun 1990;58:2669-74.
Dhawan B, Dar L, et al. Association of Mycoplasma 33. Jacobs E. Mycoplasma pneumoniae disease manifestations
pneumoniae and asthma among Indian children. FEMS and epidemiology. In: Razin S, Herrman R, editors. Molecular
Immunol Med Microbiol 2009;56:25-31. Biology and Pathogenicity of Mycoplasmas. New York, N.Y:
16. Guilbert TW, Denlinger LC. Role of infection in the Kluwer Academic/Plenum Publishers; 2002. p. 519-30.
development and exacerbation of asthma. Expert Rev Respir 34. Chourasia BK, Chaudhry R, Malhotra P. Delineation
Med 2010;4:71-83. of immunodominant and cytadherence segment(s) of
17. Wilson MH, Collier AM. Ultrastructural study of Mycoplasma Mycoplasma pneumoniae P1 gene. BMC Microbiol
pneumoniae in organ culture. J Bacteriol 1976;125:332-9. 2014;14:108.
18. Himmelreich R, Hilbert H, Plagens H, Pirkl E, Li BC, Herrmann 35. Balish MF, Krause DC. Cytadherence and the cytoskeleton.
R. Complete sequence analysis of the genome of the bacterium In: Razin S, Hermann R, editors. Molecular Biology and
Mycoplasma pneumoniae. Nucleic Acids Res 1996;24:4420-49. Pathogenicity of Mycoplasmas. New York, NY: Kluwer
19. Hammerschlag MR. Mycoplasma pneumoniae infections. Academic/Plenum Publishers; 2002. p. 491-518.
Curr Opin Infect Dis 2001;14:181-6. 36. Chaudhry R, Varshney AK, Malhotra P. Adhesion proteins of
20. Bitnun A, Richardson SE. Mycoplasma pneumoniae: Innocent Mycoplasma pneumoniae. Front Biosci 2007;12:690-9.
bystander or a true cause of central nervous system disease? 37. Waldo RH 3rd, Krause DC. Synthesis, stability, and function
Curr Infect Dis Rep 2010;12:282-90. of cytadhesin P1 and accessory protein B/C complex of
21. Atkinson TP, Balish MF, Waites KB. Epidemiology, clinical Mycoplasma pneumoniae. J Bacteriol 2006;188:569-75.
manifestations, pathogenesis and laboratory detection of 38. Hasselbring BM, Sheppard ES, Krause DC. P65 truncation
Mycoplasma pneumoniae infections. FEMS Microbiol Rev impacts P30 dynamics during Mycoplasma pneumoniae
2008;32:956-73. gliding. J Bacteriol 2012;194:3000-7.

www.ijmm.org
[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

January-March 2016 Chaudhry, et al.: M. pneumoniae pathogenesis update 15

39. Drasbek M, Nielsen PK, Persson K, Birkelund S, Christiansen Care Med 2010;182:797-804.
G. Immune response to Mycoplasma pneumoniae P1 and 55. Kannan TR, Coalson JJ, Cagle M, Musatovova O, Hardy RD,
P116 in patients with atypical pneumonia analyzed by ELISA. Baseman JB. Synthesis and distribution of CARDS toxin
BMC Microbiol 2004;4:7. during Mycoplasma pneumoniae infection in a murine model.
40. Tryon VV, Baseman JB. Pathogenic determinants and J Infect Dis 2011;204:1596-604.
mechanisms. In: Maniloff J, editor. Mycoplasmas: Molecular 56. Hardy RD, Coalson JJ, Peters J, Chaparro A,
Biology and Pathogenesis. Washington, D.C: American Techasaensiri C, Cantwell AM, et al. Analysis of pulmonary
Society for Microbiology; 1992. p. 457-71. inflammation and function in the mouse and baboon after
41. Almagor M, Kahane I, Yatziv S. Role of superoxide anion exposure to Mycoplasma pneumoniae CARDS toxin. PLoS
in host cell injury induced by Mycoplasma pneumoniae One 2009;4:e7562.
infection. A study in normal and trisomy 21 cells. J Clin Invest 57. Medina JL, Coalson JJ, Brooks EG, Le Saux CJ, Winter VT,
1984;73:842-7. Chaparro A, et al. Mycoplasma pneumoniae CARDS toxin
42. Sun G, Xu X, Wang Y, Shen X, Chen Z, Yang J. Mycoplasma exacerbates ovalbumin-induced asthma-like inflammation in
pneumoniae infection induces reactive oxygen species and BALB/c mice. PLoS One 2014;9:e102613.
DNA damage in A549 human lung carcinoma cells. Infect 58. Latz E, Xiao TS, Stutz A. Activation and regulation of the
Immun 2008;76:4405-13. inflammasomes. Nat Rev Immunol 2013;13:397-411.
43. Hickman-Davis JM, McNicholas-Bevensee C, Davis IC, 59. Gabay C, Lamacchia C, Palmer G. IL-1 pathways in
Ma HP, Davis GC, Bosworth CA, et al. Reactive species inflammation and human diseases. Nat Rev Rheumatol
mediate inhibition of alveolar type II sodium transport 2010;6:232-41.
during Mycoplasma infection. Am J Respir Crit Care Med 60. Baroja-Mazo A, Martín-Sánchez F, Gomez AI,
2006;173:334-44. Martínez CM, Amores-Iniesta J, Compan V, et al. The NLRP3
44. Hardy RD, Jafri HS, Olsen K, Hatfield J, Iglehart J, inflammasome is released as a particulate danger signal
Rogers BB, et al. Mycoplasma pneumoniae induces chronic that amplifies the inflammatory response. Nat Immunol
respiratory infection, airway hyperreactivity, and pulmonary 2014;15:738-48.
inflammation: A murine model of infection-associated chronic 61. Besnard AG, Togbe D, Couillin I, Tan Z, Zheng SG,
reactive airway disease. Infect Immun 2002;70:649-54.
Erard F, et al. Inflammasome-IL-1-Th17 response in allergic
45. Saraya T, Nakata K, Nakagaki K, Motoi N, Iihara K,
lung inflammation. J Mol Cell Biol 2012;4:3-10.
Fujioka Y, et al. Identification of a mechanism for lung
62. Segovia JA Jr, Bose S, Somarajan SR, Chang TH, Kannan
inflammation caused by Mycoplasma pneumoniae using a
TR, Baseman JB. Mycoplasma pneumoniae CARDS toxin
novel mouse model. Results Immunol 2011;1:76-87.
regulates NLRP3 inflammosome activation. J  Allergy Clin
46. Shimizu T, Kida Y, Kuwano K. A dipalmitoylated lipoprotein
Immunol 2015;135:S153.
from Mycoplasma pneumoniae activates NF-kappa B through
63. Bose S, Segovia JA, Somarajan SR, Chang TH, Kannan TR,
TLR1, TLR2, and TLR6. J Immunol 2005;175:4641-6.
Baseman JB. ADP-ribosylation of NLRP3 by Mycoplasma
47. Clyde WA Jr. Models of Mycoplasma pneumoniae infection.
pneumoniae CARDS toxin regulates inflammasome activity.
J Infect Dis 1973;129:S69-71.
MBio 2014;5:e02186-14.
48. Hu PC, Collier AM, Baseman JB. Alterations in the
metabolism of hamster tracheas in organ culture after 64. Krishnan M, Kannan TR, Baseman JB. Mycoplasma
infection by virulent Mycoplasma pneumoniae. Infect Immun pneumoniae CARDS toxin is internalized via clathrin-
1975;11:704-10. mediated endocytosis. PLoS One 2013;8:e62706.
49. Gabridge MG, Johnson CK, Cameron AM. Cytotoxicity 65. Barnes PJ. The cytokine network in asthma and
of Mycoplasma pneumoniae membranes. Infect Immun chronic obstructive pulmonary disease. J Clin Invest
1974;10:1127-34. 2008;118:3546-56.
50. Cohen G, Somerson NL. Mycoplasma pneumoniae: Hydrogen 66. Talkington DF, Waites KB, Schwartz SB, Besser RE.
peroxide secretion and its possible role in virulence. Ann N Y Emerging from obscurity: Understanding pulmonary and
Acad Sci 1967;143:85-7. extrapulmonary syndromes, pathogenesis, and epidemiology
51. Kannan TR, Provenzano D, Wright JR, Baseman JB. of human Mycoplasma pneumoniae infections. In: Scheld
Identification and characterization of human surfactant protein WM, Craig WA, Hughes JM, editors. Emerging Infections.
A binding protein of Mycoplasma pneumoniae. Infect Immun Vol. 5. Washington, D.C: American Society for Microbiology;
2005;73:2828-34. 2001. p. 57-84.
52. Kannan TR, Baseman JB. ADP-ribosylating and vacuolating 67. Murray HW, Masur H, Senterfit LB, Roberts RB. The protean
cytotoxin of Mycoplasma pneumoniae represents unique manifestations of Mycoplasma pneumoniae infection in
virulence determinant among bacterial pathogens. Proc Natl adults. Am J Med 1975;58:229-42.
Acad Sci U S A 2006;103:6724-9. 68. Pönkä A. The occurrence and clinical picture of serologically
53. Kannan TR, Musatovova O, Balasubramanian S, Cagle M, verified Mycoplasma pneumoniae infections with emphasis on
Jordan JL, Krunkosky TM, et al. Mycoplasma pneumoniae central nervous system, cardiac and joint manifestations. Ann
Community acquired respiratory distress syndrome toxin Clin Res 1979;11:1-60.
expression reveals growth phase and infection-dependent 69. Koskiniemi M. CNS manifestations associated with
regulation. Mol Microbiol 2010;76:1127-41. Mycoplasma pneumoniae infections: Summary of cases
54. Techasaensiri C, Tagliabue C, Cagle M, Iranpour P, Katz K, at the University of Helsinki and review. Clin Infect Dis
Kannan TR, et al. Variation in colonization, ADP-ribosylating 1993;17:S52-7.
and vacuolating cytotoxin, and pulmonary disease severity 70. Mok JY, Inglis JM, Simpson H. Mycoplasma pneumoniae
among Mycoplasma pneumoniae strains. Am J Respir Crit infection. A retrospective review of 103 hospitalised children.

www.ijmm.org
[Downloaded free from http://www.ijmm.org on Monday, July 3, 2017, IP: 14.139.245.2]

16 Indian Journal of Medical Microbiology vol. 34, No. 1

Acta Paediatr Scand 1979;68:833-9. with antiganglioside antibodies. Pediatr Neurol


71. Kleemola M, Käyhty H. Increase in titers of antibodies to 1998;18:160-4.
Mycoplasma pneumoniae in patients with purulent meningitis. 89. Sinha S, Prasad KN, Jain D, Pandey CM, Jha S,
J Infect Dis 1982;146:284-8. Pradhan S. Preceding infections and anti-ganglioside
72. Narita M. Pathogenesis of neurologic manifestations antibodies in patients with Guillain-Barré syndrome: A single
of Mycoplasma pneumoniae infection. Pediatr Neurol centre prospective case-control study. Clin Microbiol Infect
2009;41:159-66. 2007;13:334-7.
73. Bitnun A, Ford-Jones E, Blaser S, Richardson S. 90. Kusunoki S, Shiina M, Kanazawa I. Anti-Gal-C antibodies
Mycoplasma pneumoniae ecephalitis. Semin Pediatr Infect in GBS subsequent to Mycoplasma infection: Evidence of
Dis 2003;14:96-107.
molecular mimicry. Neurology 2001;57:736-8.
74. Behan PO, Feldman RG, Segerra JM, Draper IT. Neurological
91. Sharma MB, Chaudhry R, Tabassum I, Ahmed NH, Sahu JK,
aspects of mycoplasmal infection. Acta Neurol Scand
Dhawan B, et al. The presence of Mycoplasma pneumoniae
1986;74:314-22.
75. Narita M. Pathogenesis of extrapulmonary manifestations of infection and GM1 ganglioside antibodies in Guillain-Barré
Mycoplasma pneumoniae infection with special reference to syndrome. J Inf Dev Ctries 2011;5:459-464.
pneumonia. J Infect Chemother 2010;16:162-9. 92. Kikuchi M, Tagawa Y, Iwamoto H, Hoshino H, Yuki N.
76. Bitnun A, Ford-Jones EL, Petric M, MacGregor D, Heurter H, Bickerstaff’s brainstem encephalitis associated with IgG
Nelson S, et al. Acute childhood encephalitis and Mycoplasma anti-GQ1b antibody sub- sequent to Mycoplasma pneumoniae
pneumoniae. Clin Infect Dis 2001;32:1674-84. infection: Favorable response to immunoadsorption therapy.
77. Socan M, Ravnik I, Bencina D, Dovc P, Zakotnik B, Jazbec J. J Child Neurol 1997;12:403-5.
Neurological symptoms in patients whose cerebrospinal fluid 93. Biberfeld G, Johnsson T, Jonsson J. Studies on Mycoplasma
is culture-  and/or polymerase chain reaction-positive for pneumoniae infection in Sweden. Acta Pathol Microbiol
Mycoplasma pneumoniae. Clin Infect Dis 2001;32:E31-5. Scand 1965;63:469-75.
78. Talkington DF. Mycoplasma and Ureaplasma in central 94. Canavan TN, Mathes EF, Frieden I, Shinkai K. Mycoplasma
nervous system disorders. In: Scheld M, Whitley R, pneumoniae-induced rash and mucositis as a syndrome
Marra, C, editors. Infections of the Central Nervous System. distinct from Stevens-Johnson syndrome and erythema
Philadelphia, USA: In Press, Lippincott, Williams and multiforme: A systematic review. J  Am Acad Dermatol
Wilkins; 2004. 2015;72:239-45.
79. Narita M, Itakura O, Matsuzono Y, Togashi T. Analysis 95. Schalock PC, Dinulos JG, Pace N, Schwarzenberger K,
of mycoplasmal central nervous system involvement by
Wenger JK. Erythema multiforme due to Mycoplasma
polymerase chain reaction. Pediatr Infect Dis J 1995;14:236-7.
pneumoniae infection in two children. Pediatr Dermatol
80. Bencina D, Dovc P, Mueller-Premru M, Avsic-Zupanc T,
2006;23:546-55.
Socan M, Beovic B, et al. Intrathecal synthesis of specific
antibodies in patients with invasion of the central nervous 96. Bar Meir E, Amital H, Levy Y, Kneller A, Bar-Dayan
system by Mycoplasma pneumoniae. Eur J Clin Microbiol Y, Shoenfeld Y. Mycoplasma pneumonia - Induced
Infect Dis 2000;19:521-30. thrombotic thrombocytopenic purpura. Acta Haematol
81. Stamm B, Moschopulos M, Hungerbuehler H, Guarner 2000;103:112-5.
J, Genrich GL, Zaki SR. Neuroinvasion by Mycoplasma 97. Bradley JS, Byington CL, Shah SS, Alverson B, Carter ER,
pneumoniae in acute disseminated encephalomyelitis. Emerg Harrison C, et al. The management of community-acquired
Infect Dis 2008;14:641-3. pneumonia in infants and children older than 3 months of
82. Ogata S, Kitamoto O. Clinical complications of age: Clinical practice guidelines by the pediatric infectious
Mycoplasma 
pneumoniae disease - Central nervous system. diseases society and the infectious diseases society of
Nihon Kyobu 
Shikkan Gakkai Zasshi 1982;20:1084-9. America. Clin Infect Dis 2011;53:617-30.
83. Bruch LA, Jefferson RJ, Pike MG, Gould SJ, Squier W. 98. Okada T, Morozumi M, Tajima T, Hasegawa M, Sakata H,
Mycoplasma pneumoniae infection, meningoencephalitis, and Ohnari S, et al. Rapid effectiveness of minocycline or
hemophagocytosis. Pediatr Neurol 2001;25:67-70. doxycycline against macrolide-resistant Mycoplasma
84. Christie LJ, Honarmand S, Yagi S, Ruiz S, Glaser CA. pneumoniae infection in a 2011 outbreak among Japanese
Anti-galactocerebroside testing in Mycoplasma pneumoniae- children. Clin Infect Dis 2012;55:1642-9.
associated encephalitis. J Neuroimmunol 2007;189:129-31. 99. Eshaghi A, Memari N, Tang P, Olsha R, Farrell DJ,
85. Jacobs E, Bartl A, Oberle K, Schiltz E. Molecular mimicry by
Low DE, et al. Macrolide-resistant Mycoplasma pneumoniae
Mycoplasma pneumoniae to evade the induction of adherence
in humans, Ontario, Canada, 2010-2011. Emerg Infect Dis
inhibiting antibodies. J Med Microbiol 1995;43:422-9.
2013;19:1525-7.
86. Biberfeld G. Antibodies to brain and other tissues in cases
of Mycoplasma pneumoniae infection. Clin Exp Immunol 100. Peuchant O, Ménard A, Renaudin H, Morozumi M,
1971;8:319-33. Ubukata K, Bébéar CM, et al. Increased macrolide resistance
87. Kumada S, Kusaka H, Okaniwa M, Kobayashi O, Kusunoki S. of Mycoplasma pneumoniae in France directly detected
Encephalomyelitis subsequent to Mycoplasma infection with in clinical specimens by real-time PCR and melting curve
elevated serum anti-Gal C antibody. Pediatr Neurol analysis. J Antimicrob Chemother 2009;64:52-8.
1997;16:241-4. 101. Linchevski I, Klement E, Nir-Paz R. Mycoplasma pneumoniae
88. Komatsu H, Kuroki S, Shimizu Y, Takada H, Takeuchi Y. vaccine protective efficacy and adverse reactions: Systematic
Mycoplasma pneumoniae meningoencephalitis and cerebellitis review and meta-analysis. Vaccine 2009;27:2437-46.

www.ijmm.org

View publication stats

Das könnte Ihnen auch gefallen