The transient but not resident (TBNR) microbiome: A Yin Yang model for lung immune system
Article in Inhalation Toxicology · August 2015
DOI: 10.3109/08958378.2015.1070220
CITATIONS
12
READS
148 4 authors, including:
Some of the authors of this publication are also working on these related projects:
Improvement of Antioxidant Defense Systems of Wheat under Water StressView project
Ophiolite 2View project Pardis Saeedi
13PUBLICATIONS 90CITATIONS SEE PROFILE
Jafar Salimian
Baqiyatallah University of Medical Sciences 57PUBLICATIONS 451CITATIONS
SEE PROFILE
Abbas Ali Imani Fooladi 257PUBLICATIONS 2,168CITATIONS
SEE PROFILE
All content following this page was uploaded by Abbas Ali Imani Fooladi on 04 January 2016.
The user has requested enhancement of the downloaded file.
ISSN: 0895-8378 (print), 1091-7691 (electronic)
Inhal Toxicol, Early Online: 1–11
!2015 Taylor & Francis. DOI: 10.3109/08958378.2015.1070220
REVIEW ARTICLE
The transient but not resident (TBNR) microbiome: a Yin Yang model for lung immune system
Pardis Saeedi1, Jafar Salimian2, Ali Ahmadi1, and Abbas Ali Imani Fooladi1
1Applied Microbiology Research Center and2Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
Abstract
The concept of microbial content of the lung is still controversial. What make this more complicated are controversial results obtaining from different methodologies about lung microbiome and the definition of ‘‘lung sterility’’. Lungs may have very low bacteria but are not completely germ-free. Bacteria are constantly entering from the upper respiratory tract, but are then quickly being cleared. We can find bacterial DNA in the lungs, but it is much harder to ask about living bacteria. Here, we propose that if there is any trafficking of the microorganisms in the lung, it should be a ‘‘Transient But Not Resident (TBNR)’’ model. So, we speculate a
"Yin Yang model" for the lung immune system and TBNR. Despite beneficial roles of microbiome on the development of lung immune system, any disruption and alteration in the microbiota composition of upper and lower airways may trigger or lead to several diseases such as asthma, chronic obstructive pulmonary disease and mustard lung disease.
Keywords
Asthma, COPD, lung immune system, lung microbiome, lung sterility, mustard, TBNR microbiome
History
Received 27 January 2015 Revised 19 April 2015 Accepted 3 July 2015
Published online 24 August 2015
Introduction
The human body mucosal surfaces comprise a heterogeneous microbial composition which regulates the immune responses and immune homeostasis of the body. There is a close relationship between body health and body microbiota. In several diseases, immune functions of these mucosa may alter, which in turn, may cause various pathological compli- cations. Although many researchers recently believe that the lower airway is not sterile, it seems that the lung is not essentially harbor resident organisms. From upper airways, the organisms may constantly enter downward to the lung and then be eliminated by the lung defense systems. Here, we speculate that unlike other mucosal surfaces, the lung has a Transient But Not Resident (TBNR) microbiome pattern, which establish a dynamic relation with the immune system.
In this review, we try to define microbiome in general, and the lung microbiome and its origin, in particular. Also, we compare the germ-free and conventional animals in terms of their immune structural and functional differences due to the presence of lung microbiome. Then, we study the role of microbiome in the development and homeostasis of lung immune system, and interaction between lung immune responses and lung microbiome (Yin Yang model). Finally, we describe the factors altering the content of the lung
microbiome, which can lead to some diseases such as asthma and allergy, chronic obstructive pulmonary disease (COPD) and mustard lung disease.
The microbiome: definition
The ‘‘microbiome’’ is the total collection of viable and non- viable microorganisms and their products on the skin or mucosal surfaces of the human body [e.g. gastrointestinal (GI), urogenital and respiratory tracts] (Collado et al., 2012;
Grice & Segre, 2012; Ubeda & Pamer, 2012; Ursell et al., 2012). Two other related terms are ‘‘microbiota’’, that are all viable microbial community and taxa existing in humans (Grice & Segre, 2012; Petersen & Round, 2014); and
‘‘microflora’’, that are microorganisms that inhabit certain areas of the body (a subset of microbiota). There are about 1014symbiotic microbial cells, tenfold more than total human body cells (1013), which make up around 2% of total body mass and consist of commensal bacteria and fungi (Cho &
Blaser, 2012; Collado et al., 2012; Garzoni et al., 2013; Sze et al., 2014; Turnbaugh et al., 2007; Ursell et al., 2012; Yi &
Li, 2012). The concept of human microbiome, the collection of all human microorganisms and their products (Collado et al., 2012; Petersen & Round, 2014), was unraveled by evolving several methods (such as DNA sequencing, germ free-animal models, diversity profiling, the operational taxo- nomic units, and comparing core and transient microbiota) (Ursell et al., 2012). Besides being critical for organ development and health condition, the human microbiome and its components contribute in developing and even differentiating the host immune system. Microbiota compos- ition may have a variation in number and diversity among Address for correspondence: Abbas Ali Imani Fooladi, Applied
Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran. E-mail: [email protected] and imani- [email protected]
Jafar Salimian, Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran. E-mail: jafar.salimian@
gmail.com Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
different individuals, which in turn, may change during their lifespan. In spite of all crucial and beneficial roles, any disruption in the microbiota composition (i.e. dysbiosis or microbial imbalance) may trigger or lead to several diseases (Littman & Pamer, 2011; Ubeda & Pamer, 2012).
The lung microbiome
Still remain controversial issue?
The airways microbiome is the entire community of micro- organisms and their components that inhabit the respiratory tract (Garzoni et al., 2013; Gollwitzer & Marsland, 2014; Sze et al., 2014). The upper respiratory tract (URT) (nasopharynx, oropharynx and laryngopharynx) is colonized with a vast number of anaerobes and tenfold more aerobic bacteria. In the lower airways, the most common anaerobic bacteria found are Peptostreptococcus, Veillonella, Actinomyces and Fusobac- terium species, and the furthermost typical aerobic bacteria include Streptococcus, Haemophilus and Neisseria species (Murray et al., 2013); mostly influenced by the individual’s environment, gut microbiome and nutrition (Gollwitzer &
Marsland, 2014; Madan et al., 2012). The presence of normal flora in lower respiratory tract (LRT) (larynx, trachea, bronchioles and lower airways) is a controversial issue. At first, it is worth mentioning that there are no universal and validated criteria for defining the concept of ‘‘sterility’’ and especially ‘‘sterility in lungs’’. Whether the sterility is defined as lack of living microorganisms (undetectable in pure cultures), or the presence of rare transient bacteria and/or any microbial components such as nucleic acids and struc- tural/secretory products are the questions to be addressed. In fact, it all depends on what we mean by sterility. Lungs may have very low bacteria but are not completely germ-free.
Bacteria are constantly entering from the URT, but are then being cleared. We can find bacterial DNA in the lungs, but it is much harder to ask about living bacteria.
Taken together, we propose that if there is any trafficking of the microorganisms in the lung, it should be a transient model, but not resident microbiota. As a result, there is TBNR microbiome in the bronchial tree and lung. The key concept is the time of presence of organisms in that location; whether the bacteria have short-term visa for inhabiting there, or accord- ing to Zakharkina et al. (2013), there is a colonization and long validity visa for bacterial passengers to reside in the lung?
Early studies applying traditional methods indicated that the lower airway in healthy persons is sterile; but modern analytical techniques, such as 16S rRNA sequencing, disclose a new concept supporting the microbial presence in the human lower airways. As pioneers, Laurenzi et al. (1961), Pecora (1963), Austrian (1968) and also Murray et al. (2013), based on traditional culture-based methods, have revealed that
‘‘the lungs are sterile organs’’ (Austrian, 1968; Laurenzi et al., 1961; Murray et al., 2013; Pecora, 1963). On the other hand, other studies by Monso et al. (1999), Harris et al.
(2007), Bousbia et al. (2010), Hilty et al. (2010), Charlson et al. (2011), Erb-Downward et al. (2011) and Huang et al.
(2011), Sze et al. (2012), Duff et al. (2013), Garzoni et al.
(2013), Rogers et al. (2013) and Segal et al. (2013) and Sze et al. (2014) supported the contradictory concept that lungs
are not sterile using culture-independent methods based on genomics and metagenomics (Bousbia et al., 2010; Charlson et al., 2011; Duff et al., 2013; Erb-Downward et al., 2011;
Garzoni et al., 2013; Harris et al., 2007; Hilty et al., 2010, Huang et al., 2011; Monso et al., 1999; Rogers et al., 2013;
Segal et al., 2013; Sze et al., 2012, 2014). Thus, as listed in Table 1, different studies have resulted in distinct outcomes for and against the presence of lung microbiome. Herein, some points are considerable. First, the culture-dependent methods have lower detection limits than culture-independent methods (e.g. 16S rRNA sequence analysis and quanitative PCR). Second, the specimen type is critical. For example, nasopharyngial and oropharyngial swabs are not as valuable as bronchoalveolar lavage fluid (BALF) and brushing sam- ples, mainly due to their cross contamination. Third, many lower airway bacteria are fastidious and need selective, enriched culture media and an experienced technician. Fourth, VBNC (viable but not culturable) bacteria may exist as microflora which cannot recover by traditional culturing.
Fifth, the culture-independent methods have been established on detection of microbial nucleic acids, instead of detecting living bacteria in the lower airways. Moreover, it is note worthy that the volume of sample and statistical analyses are essential to a valid interpretation. Furthermore, a few bacteria present in LRT may be detectable by molecular but not cultural methods, due to the fact that these organisms should not be considered as definite LRT microbiota. It seems that they could originate from microaspiration from the URT (Berger & Wunderink, 2013; Charlson et al., 2011, 2012a;
Pragman et al., 2012; Segal et al., 2013; Sze et al., 2014;
Zakharkina et al., 2013) or may come down through bronchoscopic contamination (Charlson et al., 2011, 2012b;
Lozupone et al., 2013; Pragman et al., 2012; Zakharkina et al., 2013). Conflicting conclusions about the lung microbiome among different studies is most likely due to the methodological differences, sample collection techniques, specimen volume and type, population variances, topography and alterations in DNA sequencing approaches, and the data analyses (Morris et al., 2013).
To sum up, we conclude that the LRT is not sterile since the lungs are constantly exposed to environmental microbes and those from the upper airways, so may not be considered to be completely free of bacteria.
Origin of lung microbiome
The hypothesis supporting the non-sterility of LRT poses a new question about the primary source of the lower airways microbiome (Garzoni et al., 2013; Gollwitzer & Marsland, 2014). There are some possible explanations including the impact of the environment, gut microbiome, composition of the lower airways microbiota (Madan et al., 2012), and even URT cross contamination via sampling (Gollwitzer &
Marsland, 2014).
It is obvious that the origin of major population of lower airway microbiome is from the upper airways. Moreover, there is small proportion of bacteria that are capable of tolerating the unique condition of the lungs (Shaikh-Lesko, 2014). Nevertheless, there are few reports using modern molecular techniques indicating the presence of distinctive Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
and persistent bacteria in the lower airways (Gollwitzer &
Marsland, 2014; Sze et al., 2012). Table 2 illustrates that the lower airways harbor similar and exclusive microbiota. Those that are similar to what is found in the upper airways descending to the lungs, but those that are not similar may be environmental microorganisms able to survive by the absence of sufficient microbial load in the lower airways competing with them.
Evidence for presence of lung microbiome:
comparison between germ free and conventional animals
The role of microbiome in immune system development is supported by comparing the conventional and germ-free animals. Germ-free animals anatomically and physiologically have underdeveloped immune system. For example, they have fewer and smaller lymph nodes, fewer plasma cells and lymphocytes, lower Ig levels (especially IgA), lower TLR (Toll Like Receptor) expression and more susceptibility to pathogens (Round & Mazmanian, 2009). In mucosal sites (airway, GI and urogenital tracts), the lack of microbial colonization can cause partial development of mucosa associated lymphoid tissue, and subsequently decreasing B and T lymphocytes and sIgA, decreased production of
particular antimicrobial proteins (e.g. defensin) and mucus accumulation due to mucociliary defects (Round &
Mazmanian, 2009). In these animals, upon re-exposure to foreign microbes, mucosal immunity started to develop (Gaboriau-Routhiau et al., 2009; Mazmanian et al., 2005;
Salzman, 2011). Table 3 summarizes some studies comparing immune system components between germ-free and conven- tional animals.
Lung immune system and microbiome Airways immune system structure
Human airways are exposed to a large number of pollutants, infectious pathogens and allergens. So, in order to establish a normal and effective immune response, some specific local tissues are needed in airways. Nasal associated lymphoid tissue (NALT) and bronchus-associated lymphoid tissue (BALT) are widely diffused lymphoid tissues present throughout the airways. In addition, highly organized and specialized mucosal lymphoid tissues (palatine, tubal and lingual tonsils, and adenoids, so called the Waldeyer’s ring) are present in the NALT and the BALT is widely diffused lymphoid tissues lining the airways. Furthermore, other non- lymphoid areas such as the bronchial-alveolar parenchyma can promote immune reactions and thus establish different Table 1. Studies with different methodologies that is indicative of the presence or absence of lungs microbiota.
Methodology Sampling Result References
1. Culture-based method (The LRT is sterilea)
2485 Sputum/Nasophrynx swabs
Identification of upper microbiome
Austrian (1968) Culture of BALF/swab/44
patients
Identification of lung pathogens
Pecora (1963) Pharynx swab/Sputum/
BALF/52 patients
Identification of lung pathogens
Laurenzi et al. (1961) 2. Culture-independent
methods (the LRT is not sterile)
2.1. 16S rRNA sequence analysis
Naso/Oropharyngial swabs/
BALF/6 healthy control cases
There was no unique lung microbiome
Charlson et al. (2011, 2012b)
BALF/10 healthy control cases
The composition of the lung microbiota was determined
Pragman et al. (2012)
Swabs/brushings/BALF/8 healthy control cases
The bronchial tree contains a characteristic microbiota
Hilty et al. (2010) Swabs/BALF/9 healthy
control cases
Healthy lungs harbor microbiome
Garzoni et al. (2013) BALF/7 healthy smokers
and 3 none smokers
Similar aerobic and anaer- obic genera have been detected, using culture/
molecular methods
Erb-Downward et al. (2011)
BALF/oral wash/64 healthy cases
Many specific bacteria in the lungs
Morris et al. (2013) 2.2. 16S rRNA sequence
analysis and qPCR
BALF/210 patients in ICU Pathogen strain in 6 cases Bousbia et al. (2010) BALF/29 asymptomatics
cases
Similar microbiome to UTR
Segal et al. (2013) BALF/77 HIV-negative
cases
Pathogen strain in one case Lozupone et al. (2013) 2.3. 16S rRNA sequence
analysis and qPCR and TRFLP
Surgical lung tissue sam- ples/8 healthy non- smoker cases
Specific bacteria in the lungs
Sze et al. (2012)
BALF/14 control cases Molecular identification of bacteria
Harris et al. (2007) BALF/9 healthy control
cases
A diverse microbiome in healthy cases
Zakharkina et al. (2013)
LRT, lower respiratory tract; UTR, upper respiratory tract; BALF, bronchoalveolar lavage fluid; TRFLP, terminal-restriction fragment length polymorphism.
aSterile:lack of viable microorganism.
Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
types of immune tissues in the host (Williams, 2011).
The lung innate immune system contains the mucus, mucocilliary apparatus, serum products, epithelia, dendritic cells and other types of leukocytes (Derscheid & Ackermann, 2013) (Figure 1).
Role of microbiome in development and homeostasis of lung immune system
The human immune system is divided into innate and adaptive immune responses. In the innate immune system, pattern recognition receptors [PRRs, e.g. TLR, NLR Table 3. Comparison of germ-free (GF) and conventional (CV) animals’ immune system components.
Cases GF versus CV animals References
Airways immune system anatomy
Lymph nodes Fewer and smaller Round & Mazmanian (2009) and Yi & Li (2012)
Airways immune system function
Airway hyper responsiveness (AHR) Increased Herbst et al. (2011)
Serum immunoglobin levels Lower Round & Mazmanian (2009)
Production of secretory IgA Reduced Herbst et al. (2011), Round & Mazmanian
(2009), Vieira et al. (1998), Yi & Li (2012)
Production of IgE Increased Herbst et al. (2011) and Hazebrouck et al. (2009)
Th1 responses Less Herbst et al. (2011), Gollwitzer & Marsland
(2014), Gollwitzer et al. (2014)
Th2 responses More Herbst et al. (2011), Gollwitzer & Marsland
(2014) and Gollwitzer et al. (2014)
Treg cells Lower and not effective Hazebrouck et al. (2009), Littman & Pamer
(2011), Ostman et al. (2006) and Round &
Mazmanian (2009) Immune cells levels
Levels of leukocytes Lower Round & Mazmanian (2009)
Alveolar macrophages and dendritic cells Decreased Herbst et al. (2011)
Antigen-presenting cells more efficient Gollwitzer & Marsland (2014)
NK cells in mucosa Much more Ostman et al. (2006)
NK cell priming and antiviral immunity severely compromised Thaiss et al. (2014) Basophils (producers of IL4¼allergic response) Increased Herbst et al. (2011)
Th17 cells Absent Littman & Pamer (2011)
CD4 + T cells Fewer Littman & Pamer (2011) and Round &
Mazmanian (2009)
CD8 + T cells Reduced cytotoxicity Round & Mazmanian (2009)
Cell products
Expression of MHC class II molecules Reduced Round & Mazmanian (2009)
Expression and localization of PRRs (TLRs) Reduced and defective Round & Mazmanian (2009) and Sharma et al.
(2012)
Levels of IL25 Reduced Round & Mazmanian (2009)
IL17 Decreased Littman & Pamer (2011) and Prabhudas et al.
(2011)
Induce expression of various IFN Failed Thaiss et al. (2014)
IL6, TNFa, IL12, IL1 and IL18 Failed Thaiss et al. (2014) and Vieira et al. (1998)
Expression of AMPs Defective Round & Mazmanian (2009)
IL12 and early IFNgproduction The same Vieira et al. (1998)
Cells producing IL10 The same Herbst et al. (2011)
Production of IL10 and IL13 by splenocytes Reduced Ostman et al. (2006)
Table 2. The profile of microbiota existed in lower airways which can be similar to upper airways or unique.
Lower airways harbor Bacteria References
Similar microbiota to upper Moraxellaceae, Comamonadaceae, Aerococcaceae, Rhizobiaceae, Lactobacillaceae, Nocardiaceae, Capnocytophagaceae, Microbacteriaceae, Sphingomonadaceae, Flavobacteriaceae, Peptostreptococcaceae, Fusobacteriaceae, Staphylococcaceae, Porphyromonadaceae, Pasteurellaceae, Neisseriaceae, Acidaminococcaceae, Streptococcaceae, Prevotellaceae
Garzoni et al. (2013)
Pseudomonas, Streptococcus, PrevotellaandFusobacterium, Haemophilus, Veillonella,andPorphyromonas
Erb-Downward et al. (2011) Corynebacterium, Prevotella, Staphylococcus, Streptococcus, Veilonella,
HaemophilusandNeisseria
Hilty et al. (2010)
Unique bacteria Tropherymawhipplei Berger & Wunderink (2013)
and Charlson et al. (2011) Comamonadaceae, Diaphorobacter unclassified, Brevundimonasdiminuta Sze et al. (2012)
Prevotella, Porphyromonas, leptotrichia, Neisseria elcogata, Acinetobacterjohnsoni, Haemophilus influanzae
Hilty et al. (2010)
Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
(NOD-like receptors)] are scanning microbial components.
After binding of ligands to their related TLRs, signaling pathways through adaptor proteins [e.g. MyD88 or TRIF (TIR-domain-containing adapter-inducing interferon-b)]
induced various gene expressions in innate immune cells (e.g. dendritic cells) (Chang, 2012; Sharma et al., 2012).
These genes include inflammatory or regulatory cytokines, phagocytosis mediators, co-stimulatory and other molecules involved in antigen presentation. T cells recognize antigens in MHC cleft and become activated via cytokine and co- stimulatory molecules and differentiate into helper cells such as Th1 (cell mediated immunity), Th2 (humoral immunity), Th17 (inflammatory condition) or Treg (regulatory T cell) (Han et al., 2012; Ivanov et al., 2009).
Development of human immune system startsin uteroand continues postnatal. Before birth, the default pattern of immune system is dictated by Th2 responses. Following birth, Th2 polarity in the fetal immune system will switch to a non-allergic Th1 phenotype (Azad & Kozyrskyj, 2012; Holt et al., 2005; Leme et al., 2006). In fact, after birth, inhalation of air microorganisms can cause immune system development in the neonatal lung. In a normal state, inhalation of air microorganisms promote TLR expression (especially TLR9 in the lung) and direct immune system response toward cell- mediated immunity (Th1 cells) and production of IFNg and TNFa in the lung. It seems that air microorganisms or their products bind to PRRs and can stimulate dendritic cells and shift naı¨ve T cells to Th1 cells (Imani Fooladi et al., 2011). In fact, the fetus produces high levels of anti-inflammatory cytokines like IL10 and TGFb(Sharma et al., 2012), but has very low levels of pro-inflammatory cytokines such as IL1b, IL6, TNFaand IFNa(Strunk et al., 2004). From infancy to adolescence, the production of lung Th1 cytokines such as IFNg, IL12 and IFNa is increased (Figure 2). This phenom- enon has a crucial role to prevent lung immune response shift
into the Th2 pattern. Principally, it is an inverse relationship between IFNg level and serum IgE. Immune system devel- opment into Th1 protects against neonatal asthma and allergic disease. Non-existence of microbiome in germ free mice can cause immune response development toward Th2 type and leads to allergic reaction. In a mouse model, mucosal administration of component or whole bacterium of innocu- ous Escherichia coli such as lipopeptide (stimulator of Figure 1. The schematic structure of lung innate immune system. The lung innate immune system contains epithelial cells and the mucus, mucocilliary apparatus, serum products, epithelial cell, dendritic cells and other types of leukocytes. TLR, toll like receptor; AMP, antimicrobial peptides; DC, dendretic cell; MQ, macrophage; E, eosinophil; B, B cell; T, T cell.
Figure 2. Development of lung immune system before and after birth.
Before birth, default of lung immune system is Th2 type. After birth, neonatal airways were colonized by air microorganisms (TBNR) that trigger TLR expression. This TBNR microbiome induces lung immune system and shift naı¨ve T cells toward Th1 Type. In pathologic condition, alteration in microbiota can change immune system response into Th2 or Th17 types (Renz et al., 2012).
Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
TLR2), peptidoglycan (TLR2, TLR4), LPS (TLR4) and DNA (TLR9) can induce Th1 immune response and protect mice against asthma and allergy (Marsland et al., 2013; Thaiss et al., 2014). In most cases, the absence of Th2 response means that immune response has switched to Th1 type. Also, nasal administration of innocuous Gram-positive and Gram- negative bacteria induces Th1 type immune response (Gollwitzer et al., 2014). Several studies have used probiotics (oral administration) as a safe and noninvasive strategy to ameliorate some diseases. Through balancing the resident microbiota, it regulates immune responses in mucosal surfaces, moderates pathophysiological symptoms and diminishes the prevalence of allergy and cancers (Guandalini et al., 2015; Imani Fooladi et al., 2013; Khani et al., 2012).
Apart from airways microbiome, the non-airway micro- biome also has an important role in the lung immune- regulation (Th1/Th2 balancing). GI microbiome induces regulatory T cells that help keep the balance of Th1/Th2. In addition, recognition of microbiota compositions by PRRs enhances systemic innate immunity (Azad & Kozyrskyj, 2012; Kaplan et al., 2011). For maintaining the homeostasis of intestinal cells, PRRs sense microbial compounds which induce Treg and Th17 cells (Ichinohe et al., 2011). The most significant functional role of the human microbiota is mainly in the GI tract and can be involved in the maturation of the host immune system. Despite beneficial roles, such as critical production of microelements and assistance in metabolism, disruptions in the microbiota may cause some diseases (Garzoni et al., 2013). The GI microbiome has a profound effect on shaping and regulating the whole host immune system as well as pulmonary immune responses by: (1) activating the gut dendritic cells and regulating the systemic balance of Th17 cells and Treg cells (Atarashi et al., 2011;
Han et al., 2012; Ivanov et al., 2008, 2009; Ostman et al., 2006; Worbs et al., 2006); (2) shaping the systemic normal tolerance (Abraham & Cho, 2009; Han et al., 2012); (3) mucosal maturation and immune response establishment by evolving gut-associated lymphoid tissues including Peyer’s patches, isolated lymphoid follicles and lymph nodes, (Chervonsky, 2009; Hooper & Gordon, 2001) which are underdeveloped in early infants and germ-free animals (Ichinohe et al., 2011; Round & Mazmanian, 2009).
The role of airway microbiome in diseases
As mentioned above, microbiome has an important role in physiological homeostasis of the body. It most likely acts through colonization competition, maintenance of epithelial integrity and immune system regulation (Erb-Downward et al., 2011). Although the precise effect of airway micro- biome in diseases is a complicated issue (Han et al., 2012;
Huang et al., 2013; Huang & Lynch, 2011), many researchers have demonstrated that the community of airway microbiome may change between healthy and diseased conditions (Huang
& Lynch, 2011; Zakharkina et al., 2013). There are some factors that seem to contribute to alterations (stated below) in airway microbiota that, in turn, may lead to various subse- quent infectious and non-infectious diseases. Here we sum- marize these items in more detail.
Factors altering the content of lung microbiota
There are some factors which disrupt the lower airway microbiota and may lead to several chronic lung diseases (Erb-Downward et al., 2011; Garzoni et al., 2013; Han et al., 2012; Hilty et al., 2010; Pragman et al., 2012; Sze et al., 2012). These factors may be classified as: (I) Anatomical injuries (either inherited or acquired) including genetic malfunctions, mucociliary dysfunction, traumas, surgeries and burns; (II) Pathological effects including microbial infections, prolonged antibiotic therapy and exposure to hazardous chemicals; (III) Physiological changes such as hormonal alterations (e.g. puberty, menopause, nutrition and stress) and finally, (IV) Immune system defects including defects in B and T cells and phagocytes, antibodies and complement system. Bacterial colonization in the LRT may be a consequence of immune system impairment. These patients are not completely able to phagocytose the inhaled particles and bacteria and also deficient in efferocytosis of apoptotic airway epithelial cells. Some researchers have declared that, phagocytosis requires adequate function of microtubules which has failed in COPD (Barnes, 2014;
Donnelly & Barnes, 2012; Hodge et al., 2003; Mu¨llerova et al., 2012; Taylor et al., 2010). Persistent inflammation and exacerbation in COPD patients is due to failure of phagocytes in clearance of bacterial pathogens and necrotic material elimination from the lung (Barnes, 2014; Donnelly & Barnes, 2012).
Emerging alterations in the lung microbiota
With respect to the type and degree of factors mentioned previously, the possible shifts in microbiome may include: (I) Reduction in microbiome complexity (as loss of diversity in bacterial composition in COPD (Erb-Downward et al., 2011;
Sze et al., 2014) and Crohn’s disease (Cho & Blaser, 2012);
(II) Specific microbiota abundance (e.g. due to antibiotic or antiretroviral therapy) (Lozupone et al., 2013) and (III) Increased microbial pathogenicity (due to acquiring toxin, epithelial adhesive molecules, and antibiotic resistant genes) through different genetic exchanges between diverse species and genera of the microbiota resulting in new genetic traits such as antibiotic resistance and virulence in human microbiome (Cho & Blaser, 2012; Liu et al., 2012; Smillie et al., 2011).
Diseases affected by microbiome disruption
Studies have made clear that disruption in the content of microbiome may lead to clinical outcomes. Now, the question is how microbiome can cause these clinical outcomes? Many efforts have been made to determine the causative relation between abundance and diversity of microbiome and patho- logical conditions (Cho & Blaser, 2012). Reduction of microbiome diversity and complexity is accompanied by the overrepresentation of particular microbial species which results in inflammation (Boyton et al., 2013; Han et al., 2012). Loss of keystone microbial species may trigger some cascade events (targeting the imbalance of immune regulation and increase of immune responses) that may lead to more tissue injuries (Cho & Blaser, 2012) (Figure 3). So, we Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
conclude that there is a dynamic balance between the lung microbiome and immune system, as a ‘‘Yin and Yang phenomenon’’. Here are some diseases in which the role of microbiota and shifts of their composition are decisive.
Asthma and allergy
Some studies propose that the lower airways of patients with stable asthma may be colonized by distinctive microbiota and the microbiota community and diversity lead to bronchial hyper responsiveness (Huang & Lynch, 2011). There is some evidence that overprescribing or inappropriate use of anti- microbial agents in childhood may result in asthma and allergy development (Risnes et al., 2010), which is probably due to disruption of the GI microbiome, replacement of pathogens and perturbation of mechanisms of mucosal tolerance (Dickson et al., 2013; Huffnagle, 2010). A clear outgrowth in the proportion of Firmicutes (Streptococcus), Proteobacteria (particularly Haemophilus, Moraxella and Neisseria species), Pseudomonodaceae, Comamonadaceae, Oxalobacteriaceae, Shingomonadaceae in the LRT have been shown in asthmatic groups as compared with healthy controls (Dickson et al., 2013; Hilty et al., 2010; Marsland et al., 2013) which can be both, cause and effect of the disease. By contrast, there may be a reduction of Bacteroidetes and Prevotellaspecies which is a direct inhibitory effect of other bacteria (Beck et al., 2012; Hilty et al., 2010; Huang & Lynch, 2011; Huang et al., 2011). In both cases (increase or decrese of specific bacteria) dysbiosis or imbalance of microbiome leads to these diseases.
Chronic obstructive pulmonary disease
There is a correlation between COPD, the airflow obstruction subsequent to emphysema or chronic bronchitis, and the lower airway microbiome (Beck et al., 2012). Once microbiome imbalance happens, the bacterial phyla ofProteobacteria(in particularHaemophilusspp.), Firmicutes, Comamonadaceae, Shingomonadaceae, Oxalobacteriaceae, Pseudomonodaceae are overrepresented in the lungs of allergic and COPD patients, whereas Bacteroidetes and Prevotella are reduced (Beck et al., 2012; Dickson et al., 2013; Hilty et al., 2010;
Huang & Lynch, 2011; Huang et al., 2011; Marsland et al., 2013).
COPD patients are subjected to several other conditions resulting in acute worsening of superimposed chronic disease.
Despite the destructive impacts on the lungs, COPD should be considered as a complex systemic disease (Karadag et al., 2008). COPD exacerbation, a very common distressing event affecting some patients with moderate-to-severe COPD, impacts impressively on health state and alleviation of symptoms (Bourbeau, 2009). Apart from the clinical diversity of COPD, the nature and course of inflammation commonly affects the airways, which exacerbates with the progress of disease. There is evidence of the impact of microbiota alteration on triggering inflammatory events. The lungs inflammatory response initiates by infiltration of inflamma- tory and immune cells leading to tissue destruction, alveolar airspace enlargement and disease progression (Stefanska &
Walsh, 2009; Sze et al., 2014). There is also evidence for the existence of auto-immunity contributing to COPD Figure 3. Illustration of the lung microbiome disruption resulting in vicious cycle.
Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
progression. The presentation of endogenous antigens com- mences tissue destruction and gives rise to a classical autoimmune response (Stefanska & Walsh, 2009).
Mustard lung disease
Sulfur mustard (or mustard gas, SM [2,20-dichlorodiethyl sulfide]) is a potent chemical vesicant (blistering) with extensive morbidity (Ghasemi et al., 2009; Kehe & Szinicz, 2005; Malaviya et al., 2010; Roshan et al., 2013; Shams et al., 2009). It is a toxic DNA alkylating agent and identified as a human carcinogen (Jafari & Ghanei, 2010; Lari et al., 2012;
Roshan et al., 2013; Shams et al., 2009; Steinritz et al., 2007).
It was used during the World War I and in the Iraq-Iran war during 1980–1988 (Balali-Mood & Hefazi, 2006). Over 100 000 people were subjected to severe injuries and approximately 45 000 patients continue suffering from long term exposure (Roshan et al., 2013). SM has both short- and long-term toxic effects in the eyes, skin, nervous system, respiratory and hematopoietic systems (Balali-Mood &
Hefazi, 2006; Ghasemi et al., 2013, Ghazanfari et al., 2009, 2013; Lari et al., 2014; Shohrati et al., 2007; Yaraee et al., 2009). The respiratory epithelial cells are very susceptible to the effects of SM exposure and undergo sloughing which enhance airways secretions and nasal discharge, then airway obstruction, and even bronchospasm. After acute mustard exposure, airway damage leads to pulmonary insufficiency and infection (Razavi et al., 2013). Hematological results indicated a major drop in WBC and an increase in lympho- cytes (Ayyobi et al., 2014; Ghazanfari et al., 2013; Mirzamani et al., 2012), as well as neutrophilia (Beheshti et al., 2006;
Ebrahimi et al., 2010).
According to some Iranian researchers’ findings in indi- viduals chronically exposed to SM, there is elevation in expression levels of TGF-b1, TGF-b2, C-fos, NF-kB and inflammatory cytokines (Pourfarzam et al., 2009), but reduc- tion in heme oxygenase in airways epithelial cells (Imanifooladi et al., 2010; Nourani et al., 2010, 2011), and efferocytosis (Pirzad et al., 2011; Zarin et al., 2010). The serum level of some chemokines such as SDF-1a (Stromal- Derived Factor 1a or CXCL12) is reduced (Ayyobi et al., 2014). Mitigating vesicant effect and toxicity of SM requires operative strategies. Inhaled corticosteroids, long-acting b2- agonists and macrolide antibiotics are effective in treatment of these patients (Ghanei et al., 2007). However, steroid treatment can disrupt the microbial balance.
SM can directly affect or eliminate some lower airway microbiome by producing free radicals and sloughing (Brimfield et al., 2012; Graham et al., 2005; Sadraei et al., 2011). On the other hand, mucuciliary clearance deficiency helps microbial pathogen colonization to lower airways of mustard-exposed lungs. Inspecting microbial communities, in 25% of BAL samples, bacteria was observed including S. aureus, coagulase-negative Staphylococcus species and E. coli (Moghadam et al., 2003). Ghanei et al. reported the prevalence of bacteria in BAL culture of 79 lung mustard patients for the first time. It consists of 55% SNGA (non- group A streptococci), 13% Pseudomonas aeroginosa, 9%
enterobacter, 7%S. aureus, 6% SGA (group A streptococci) and 5%Klebsiellaand the other microorganisms totalling 5%.
AlsoH. influenza,S. pneumoniae andMoraxella catarrhalis are responsible for 85–95% of exacerbations. Besides, S.
viridansis found as pneumonia agent in COPDs.H. influenza is an intracellular pathogen, which accelerates the risk of chlamydia pnemoniaeinfections (Panahi et al., 2004).
In an experiment, Imani et al. utilized siRNA to inhibit opportunistic bacteria such as pseudomonas in veterans’
airways. They found that this treatment method was not efficient because of high growth speed of bacteria (Fooladi et al., 2013).
Conclusion
Despite the recent studies implying the presence of lung microbiome, the topic remains controversial. The final decision about the presence of lung microbiome may depend on the obtaining of completely sterile specimens, isolation of pure and live organisms (through culture based techniques) and precise characterization of isolated strains. It seems that some key questions should be addressed. Are the organisms resident? Or they are transient in the location (short validity visa). Can we apply the concept of TBNR micro- biome or not? Is there any colonization? On the other hand, we cannot ignore the results of studies reporting about 2200 different genes in the bronchial tree in addition to the role of lower airway microbiome in the development and homeosta- sis of lung immune system between normal and germ-free animals. There is no doubt about the role of lung microbiome on the homeostasis of lung immune system. In fact, what we usually call ‘‘Yin-Yang phenomenon’’, is a dynamic balance between TBNR microbiome of lower airways and immune responses.
In conclusion, if considering microbiota as transient bacteria rather than resident (TBNR), prescribing steroids and antibiotics to ameliorate the lung inflammation can weaken immune system and favor the growth of opportunistic microorganisms. These therapeutic strategies cannot specif- ically be helpful in the treatment of mustard-exposed lungs.
Declaration of interest
The authors report no conflicts of interest. The authors alone are responsible for the content and writing of this article.
References
Abraham C, Cho JH. (2009). Inflammatory bowel disease. N Engl J Med 361:2066–78.
Atarashi K, Tanoue T, Shima T, et al. (2011). Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331:
337–41.
Austrian R. (1968). The bacterial flora of the respiratory tract: some knowns and unknowns. Yale J Biol Med 40:400–13.
Ayyobi F, Ghazanfari T, Askari N, et al. (2014). Chemokine (SDF-1a CXCL12) level in chemical veterans with pulmonary problems. Tebe Janbazan Military J 6:55–63.
Azad MB, Kozyrskyj AL. (2012). Perinatal programming of asthma: the role of gut microbiota. Clin Dev Immunol 2012:932072.
Balali-Mood M, Hefazi M. (2006). Comparison of early and late toxic effects of sulfur mustard in Iranian veterans. Basic Clin Pharmacol Toxicol 99:273–82.
Barnes PJ. (2014). Cellular and molecular mechanisms of chronic obstructive pulmonary disease. Clin Chest Med 35:71–86.
Beck JM, Young VB, Huffnagle GB. (2012). The microbiome of the lung. Transl Res 160:258–66.
Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
Beheshti J, Mark EJ, Akbaei HMH, et al. (2006). Mustard lung secrets:
long term clinicopathological study following mustard gas exposure.
Pathol Res Pract 202:739–44.
Berger G, Wunderink RG. (2013). Lung microbiota: genuine or artifact?
Isr Med Assoc J 15:731–3.
Bourbeau J. (2009). Activities of life: the COPD patient. J Chron Obstruct Pulmon Dis 6:192–200.
Bousbia S, Papazian L, Auffray JP, et al. (2010). Tropheryma whipplei in patients with pneumonia. Emerg Infect Dis 16:258–63.
Boyton RJ, Reynolds CJ, Quigley KJ, Altmann DM. (2013). Immune mechanisms and the impact of the disrupted lung microbiome in chronic bacterial lung infection and bronchiectasis. Clin Exp Immunol 171:117–23.
Brimfield AA, Soni SD, Trimmer KA, et al. (2012). Metabolic activation of sulfur mustard leads to oxygen free radical formation. Free Radic Biol Med 52:811–17.
Chang C. (2012). Neonatal autoimmune diseases: a critical review.
J Autoimmun 38:J223–38.
Charlson ES, Bittinger K, Chen J, et al. (2012a). Assessing bacterial populations in the lung by replicate analysis of samples from the upper and lower respiratory tracts. PLoS One 7:e42786.
Charlson ES, Bittinger K, Haas AR, et al. (2011). Topographical continuity of bacterial populations in the healthy human respiratory tract. Am J Respir Crit Care Med 184:957–63.
Charlson ES, Diamond JM, Bittinger K, et al. (2012b). Lung-enriched organisms and aberrant bacterial and fungal respiratory microbiota after lung transplant. Am J Respir Crit Care Med 186:536–45.
Chervonsky A. (2009). Innate receptors and microbes in induction of autoimmunity. Curr Opin Immunol 21:641–7.
Cho I, Blaser MJ. (2012). The human microbiome: at the interface of health and disease. Nat Rev Genet 13:260–70.
Collado MC, Bauerl C, Perez-Martinez G. (2012). Defining microbiota for developing new probiotics. Microb Ecol Health Dis 23:18579. doi:
10.3402/mehd.v23i0.18579.
Derscheid RJ, Ackermann MR. (2013). The innate immune system of the perinatal lung and responses to respiratory syncytial virus infection.
Veterinary Pathol Online 50:827–41.
Dickson RP, Erb-Downward JR, Huffnagle GB. (2013). The role of the bacterial microbiome in lung disease. Expert Rev Respir Med 7:
245–57.
Donnelly LE, Barnes PJ. (2012). Defective phagocytosis in airways diseasephagocytic dysfunction and airways disease. Chest J 141:
1055–62.
Duff RM, Simmonds NJ, Davies JC, et al. (2013). A molecular comparison of microbial communities in bronchiectasis and cystic fibrosis. Eur Respir J 41:991–3.
Ebrahimi M, Roudkenar MH, Fooladi I, et al. (2010). Discrepancy between mRNA and protein expression of neutrophil gelatinase- associated lipocalin in bronchial epithelium induced by sulfur mustard. J Biomed Biotechnol 2010:823131.
Erb-Downward JR, Thompson DL, Han MK, et al. (2011). Analysis of the lung microbiome in the ‘‘healthy’’ smoker and in COPD. PLoS One 6:e16384.
Fooladi AAI, Aghelimansour A, Nourani MR. (2013). Evaluation of the pathogenesis ofPseudomonas aeruginosa’s flagellum before and after flagellar gene knockdown by small interfering RNAs (siRNA) 6:273–8.
Gaboriau-Routhiau V, Rakotobe S, Le´cuyer E, et al. (2009). The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31:677–89.
Garzoni C, Brugger SD, Qi W, et al. (2013). Microbial communities in the respiratory tract of patients with interstitial lung disease. Thorax 68:1150–6.
Ghanei M, Shohrati M, Harandi AA, et al. (2007). Inhaled corticoster- oids and long-actingb2-agonists in treatment of patients with chronic bronchiolitis following exposure to sulfur mustard. Inhal Toxicol 19:
889–94.
Ghasemi H, Ghazanfari T, Yaraee R, et al. (2009). Systemic and ocular complications of sulfur mustard: a panoramic review. Toxin Rev 28:
14–23.
Ghasemi H, Mostafaie A, Yaraee R, et al. (2013). Association of serum immunoglobulins levels and eye injuries in sulfur mustard exposed:
Sardasht-Iran Cohort Study. Int Immunopharmacol 17:944–51.
Ghazanfari T, Mostafaie A, Yaraee R, et al. (2013). Are serum levels of immunoglobulin classes and IgG subclasses involved in delayed
pulmonary complications induced by sulfur mustard? Sardasht-Iran Cohort Study. Int Immunopharmacol 17:936–43.
Ghazanfari T, Sharifnia Z, Yaraee R, et al. (2009). Serum soluble Fas ligand and nitric oxide in long-term pulmonary complications induced by sulfur mustard: Sardasht-Iran Cohort Study. Int Immunopharmacol 9:1489–93.
Gollwitzer ES, Marsland BJ. (2014). Microbiota abnormalities in inflammatory airway diseases – potential for therapy. Pharmacol Ther 141:32–9.
Gollwitzer ES, Saglani S, Trompette A, et al. (2014). Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med 20:
642–7.
Graham JS, Chilcott RP, Rice P, et al. (2005). Wound healing of cutaneous sulfur mustard injuries: strategies for the development of improved therapies. J Burns Wounds 4:e1.
Grice EA, Segre JA. (2012). The human microbiome: our second genome. Annu Rev Genom Hum Genet 13:151–70.
Guandalini S, Cernat E, Moscoso D. (2015). Prebiotics and probiotics in irritable bowel syndrome and inflammatory bowel disease in children.
Benef Microbes 6:209–17.
Han MK, Huang YJ, Lipuma JJ, et al. (2012). Significance of the microbiome in obstructive lung disease. Thorax 67:456–63.
Harris JK, De Groote MA, Sagel SD, et al. (2007). Molecular identification of bacteria in bronchoalveolar lavage fluid from children with cystic fibrosis. Proc Natl Acad Sci USA 104:
20529–33.
Hazebrouck S, Przybylski-Nicaise L, Ah-Leung S, et al. (2009). Allergic sensitization to bovine beta-lactoglobulin: comparison between germ- free and conventional BALB/c mice. Int Arch Allergy Immunol 148:
65–72.
Herbst T, Sichelstiel A, Schar C, et al. (2011). Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am J Respir Crit Care Med 184:198–205.
Hilty M, Burke C, Pedro H, et al. (2010). Disordered microbial communities in asthmatic airways. PLoS One 5:e8578.
Hodge S, Hodge G, Scicchitano R, et al. (2003). Alveolar macrophages from subjects with chronic obstructive pulmonary disease are deficient in their ability to phagocytose apoptotic airway epithelial cells. Immunol Cell Biol 81:289–96.
Holt PG, Upham JW, Sly PD. (2005). Contemporaneous maturation of immunologic and respiratory functions during early childhood:
implications for development of asthma prevention strategies.
J Allergy Clin Immunol 116:16–24.
Hooper LV, Gordon JI. (2001). Commensal host-bacterial relationships in the gut. Science 292:1115–18.
Huang YJ, Charlson ES, Collman RG, et al. (2013). The role of the lung microbiome in health and disease. A National Heart, Lung, and Blood Institute workshop report. Am J Respir Crit Care Med 187:
1382–7.
Huang YJ, Lynch SV. (2011). The emerging relationship between the airway microbiota and chronic respiratory disease: clinical implica- tions. Expert Rev Respir Med 5:809–21.
Huang YJ, Nelson CE, Brodie EL, et al. (2011). Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally con- trolled asthma. J Allergy Clin Immunol 127:372–81.e1–3.
Huffnagle GB. (2010). The microbiota and allergies/asthma. PLoS Pathog 6:e1000549.
Ichinohe T, Pang IK, Kumamoto Y, et al. (2011). Microbiota regulates immune defense against respiratory tract influenza A virus infection.
Proc Natl Acad Sci USA 108:5354–9.
Imani Fooladi AA, Mahmoodzadeh Hosseini H, Nourani MR, et al.
(2013). Probiotic as a novel treatment strategy against liver disease.
Hepat Mon 13:e7521.
Imani Fooladi AA, Mousavi SF, Seghatoleslami S, et al. (2011). Toll-like receptors: role of inflammation and commensal bacteria. Inflamm Allergy Drug Targets 10:198–207.
Imanifooladi AA, Yazdani S, Nourani MR. (2010). The role of nuclear factor-kB in inflammatory lung disease. Inflamm Allergy Drug Targets 9:197–205.
Ivanov II, Atarashi K, Manel N, et al. (2009). Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139:
485–98.
Ivanov II, Frutos RDL, Manel N, et al. (2008). Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4:337–49.
Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
Jafari M, Ghanei M. (2010). Evaluation of plasma, erythrocytes, and brochoalveolar lavage fluid antioxidant defense system in sulfur mustard-injured patients. Clin Toxicol 48:184–92.
Kaplan JL, Shi HN, Walker WA. (2011). The role of microbes in developmental immunologic programming. Pediatr Res 69:465–72.
Karadag F, Karul AB, Cildag O, et al. (2008). Biomarkers of systemic inflammation in stable and exacerbation phases of COPD. Lung 186:
403–9.
Kehe K, Szinicz L. (2005). Medical aspects of sulphur mustard poisoning. Toxicology 214:198–209.
Khani S, Hosseini HM, Taheri M, et al. (2012). Probiotics as an alternative strategy for prevention and treatment of human diseases: a review. Inflamm Allergy Drug Targets 11:79–89.
Lari SM, Attaran D, Towhidi M. (2012). COPD due to sulfur mustard (mustard lung). Chronic Obstruct Pulmon Dis Curr Concepts Pract Croatia InTech 2:231–8.
Lari SM, Ghobadi H, Attaran D, et al. (2014). COPD assessment test (CAT): simple tool for evaluating quality of life of chemical warfare patients with chronic obstructive pulmonary disease. Clin Respir J 8:
116–23.
Laurenzi GA, Potter RT, Kass EH. (1961). Bacteriologic flora of the lower respiratory tract. N Engl J Med 265:1273–8.
Leme AS, Hubeau C, Xiang Y, et al. (2006). Role of breast milk in a mouse model of maternal transmission of asthma susceptibility.
J Immunol 176:762–9.
Littman DR, Pamer EG. (2011). Role of the commensal microbiota in normal and pathogenic host immune responses. Cell Host Microbe 10:
311–23.
Liu L, Chen X, Skogerbø G, et al. (2012). The human microbiome: a hot spot of microbial horizontal gene transfer. Genomics 100:265–70.
Lozupone C, Cota-Gomez A, Palmer BE, et al. (2013). Widespread colonization of the lung by Tropheryma whipplei in HIV infection.
Am J Respir Crit Care Med 187:1110–17.
Madan J, Koestler D, Stanton B, et al. (2012). Serial analysis of the gut and respiratory microbiome in cystic fibrosis in infancy: interaction between intestinal and respiratory tracts and impact of nutritional exposures. MBio 3:e00251–12.
Malaviya R, Sunil VR, Cervelli J, et al. (2010). Inflammatory effects of inhaled sulfur mustard in rat lung. Toxicol Appl Pharmacol 248:
89–99.
Marsland BJ, Yadava K, Nicod LP. (2013). The airway microbiome and disease. Chest 144:632–7.
Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. (2005). An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122:107–18.
Mirzamani M, Zare S, Imanifooladi AA, Nourani MR. (2012). TGF-b gene expression in fibroblast cells of chemical veterans. Tebe Janbazan J 4:6–15.
Moghadam KG, Keshmiri M, Omidi A-A, Tohidi M. (2003). Pulmonary complications of chemical warfare 15 years after exposure. Tanaffos 2:45–50.
Monso E, Rosell A, Bonet G, et al. (1999). Risk factors for lower airway bacterial colonization in chronic bronchitis. Eur Respir J 13:
338–42.
Morris A, Beck JM, Schloss PD, et al. (2013). Comparison of the respiratory microbiome in healthy nonsmokers and smokers. Am J Respir Crit Care Med 187:1067–75.
Mu¨llerova H, Chigbo C, Hagan GW, et al. (2012). The natural history of community-acquired pneumonia in COPD patients: a population database analysis. Respir Med 106:1124–33.
Murray PR, Rosenthal KS, Pfaller MA. (2013). Medical microbiology.
7th ed. Philadelphia: Elsevier/Saunders.
Nourani M, Imanifooladi A, Yazdani S. (2011). Genome analysis of C- fos molecule after increasing free radicals in mustard exposed of airways of COPD paitents. Tebe Janbazan J 3:46–52.
Nourani MR, Yazdani S, Roudkenar MH, et al. (2010). HO1 mRnA and protein do not change in parallel in bronchial biopsies of patients after long term exposure to sulfur mustard. Gene Regul Syst Biol 4:
83–90.
Ostman S, Rask C, Wold AE, et al. (2006). Impaired regulatory T cell function in germ-free mice. Eur J Immunol 36:2336–46.
Panahi Y, Ghaneei M, Aslani J, et al. (2004). Paper: Evaluation of microbial resistance to antibiotics in patients with chronic pulmonary lesions due to chemical agents and non-chemical agents. J Mil Med 6:181–6.
Pecora DV. (1963). A comparison of transtracheal aspiration with other methods of determining the bacterial flora of the lower respiratory tract. N Engl J Med 269:664–6.
Petersen C, Round JL. (2014). Defining dysbiosis and its influence on host immunity and disease. Cell Microbiol 16:
1024–33.
Pirzad G, Jafari M, Tavana S, et al. (2011). The role of fas-fasl signaling pathway in induction of apoptosis in patients with sulfur mustard-induced chronic bronchiolitis. J Toxicol 2010:
373612.
Pourfarzam S, Ghazanfari T, Yaraee R, et al. (2009). Serum levels of IL-8 and IL-6 in the long term pulmonary complications induced by sulfur mustard: Sardasht-Iran Cohort Study. Int Immunopharmacol 9:
1482–8.
Prabhudas M, Adkins B, Gans H, et al. (2011). Challenges in infant immunity: implications for responses to infection and vaccines. Nat Immunol 12:189–94.
Pragman AA, Kim HB, Reilly CS, et al. (2012). The lung microbiome in moderate and severe chronic obstructive pulmonary disease. PLoS One 7:e47305.
Razavi SM, Salamati P, Harandi AA, Ghanei M. (2013). Prevention and treatment of respiratory consequences induced by sulfur mustard in Iranian casualties. Int J Prev Med 4:383–9.
Renz H, Brandtzaeg P, Hornef M. (2012). The impact of perinatal immune development on mucosal homeostasis and chronic inflam- mation. Nat Rev Immunol 12:9–23.
Risnes KR, Belanger K, Murk W, Bracken MB. (2010). Antibiotic exposure by 6 months and asthma and allergy at 6 years: findings in a cohort of 1,401 US children. Am J Epidemiol 173:310–18.
Rogers GB, Van Der Gast CJ, Cuthbertson L, et al. (2013). Clinical measures of disease in adult non-CF bronchiectasis correlate with airway microbiota composition. Thorax 68:731–7.
Roshan R, Rahnama P, Ghazanfari Z, et al. (2013). Long-term effects of sulfur mustard on civilians’ mental health 20 years after exposure (The Sardasht-Iran Cohort Study). Health Qual Life Outcomes 11:69.
Round JL, Mazmanian SK. (2009). The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 9:
313–23.
Sadraei S, Bahadoran H, Dashtnavard H, et al. (2011). Effect of sulfur mustard on the epithelial cell necrosis of urinary duct of kidney in rat.
MilMed J 12:203–9.
Salzman NH. (2011). Microbiota–immune system interaction: an uneasy alliance. Curr Opin Microbiol 14:99–105.
Segal LN, Alekseyenko AV, Clemente JC, et al. (2013). Enrichment of lung microbiome with supraglottic taxa is associated with increased pulmonary inflammation. Microbiome 1:19.
Shaikh-Lesko R. (2014). Breathing life into lung microbiome research.
The Scientist Magazine 9:1.
Shams J, Ghazanfari T, Yaraee R, et al. (2009). Long-term hematological consequences of sulfur mustard on civilians of Sardasht 20 years after exposure. Toxin Rev 28:39–43.
Sharma AA, Jen R, Butler A, Lavoie PM. (2012). The developing human preterm neonatal immune system: a case for more research in this area. Clin Immunol 145:61–8.
Shohrati M, Davoudi M, Ghanei M, et al. (2007). Cutaneous and ocular late complications of sulfur mustard in Iranian veterans. Cutaneous Ocular Toxicol 26:73–81.
Smillie CS, Smith MB, Friedman J, et al. (2011). Ecology drives a global network of gene exchange connecting the human microbiome. Nature 480:241–4.
Stefanska AM, Walsh PT. (2009). Chronic obstructive pulmonary disease: evidence for an autoimmune component. Cell Mol Immunol 6:81–6.
Steinritz D, Emmler J, Hintz M, et al. (2007). Apoptosis in sulfur mustard treated A549 cell cultures. Life Sci 80:2199–201.
Strunk T, Temming P, Gembruch U, et al. (2004). Differential maturation of the innate immune response in human fetuses. Pediatr Res 56:219–26.
Sze MA, Dimitriu PA, Hayashi S, et al. (2012). The lung tissue microbiome in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 185:1073–80.
Sze MA, Hogg JC, Sin DD. (2014). Bacterial microbiome of lungs in COPD. Int J Chron Obstruct Pulmon Dis 9:229–38.
Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
Taylor A, Finney-Hayward T, Quint J, et al. (2010). Defective macrophage phagocytosis of bacteria in COPD. Eur Respir J 35:
1039–47.
Thaiss CA, Levy M, Suez J, Elinav E. (2014). The interplay between the innate immune system and the microbiota. Curr Opin Immunol 26:
41–8.
Turnbaugh PJ, Ley RE, Hamady M, et al. (2007). The human microbiome project. Nature 449:804–10.
Ubeda C, Pamer EG. (2012). Antibiotics, microbiota, and immune defense. Trends Immunol 33:459–66.
Ursell LK, Metcalf JL, Parfrey LW, Knight R. (2012). Defining the human microbiome. Nutr Rev 70:S38–44.
Vieira LQ, Oliveira MR, Neumann E, et al. (1998). Parasitic infections in germfree animals. Braz J Med Biol Res 31:
105–10.
Williams AE. (2011). Immunology: mucosal and body surface defences.
Oxford: Wiley-Blackwell.
Worbs T, Bode U, Yan S, et al. (2006). Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J Exp Med 203:519–27.
Yaraee R, Ghazanfari T, Ebtekar M, et al. (2009). Alterations in serum levels of inflammatory cytokines (TNF, IL-1alpha, IL-1beta and IL- 1Ra) 20years after sulfur mustard exposure: Sardasht-Iran Cohort Study. Int Immunopharmacol 9:1466–70.
Yi P, Li L. (2012). The germfree murine animal: an important animal model for research on the relationship between gut microbiota and the host. Vet Microbiol 157:1–7.
Zakharkina T, Heinzel E, Koczulla RA, et al. (2013). Analysis of the airway microbiota of healthy individuals and patients with chronic obstructive pulmonary disease by T-RFLP and clone sequencing.
PLoS One 8:e68302.
Zarin AA, Behmanesh M, Tavallaei M, et al. (2010). Overexpression of transforming growth factor (TGF)-b1 and TGF-b3 genes in lung of toxic-inhaled patients. Exp Lung Res 36:284–91.
Inhalation Toxicology Downloaded from informahealthcare.com by 188.34.160.243 on 08/24/15 For personal use only.
View publication stats View publication stats