Lung transplantation: infection, inflammation, and the microbiome

22
REVIEW Lung transplantation: infection, inflammation, and the microbiome Takeshi Nakajima & Vyachesav Palchevsky & David L. Perkins & John A. Belperio & Patricia W. Finn Received: 29 December 2010 / Accepted: 12 January 2011 /Published online: 27 January 2011 # Springer-Verlag 2011 Abstract Lung transplantation is the only therapeutic option for patients with end-stage pulmonary disorders. Despite the improvements in surgical techniques and immunosuppressive therapy, allograft function and long- term survival are limited by the development of chronic lung transplant rejection. In this review, we focus on bronchiolitis obliterans syndrome (BOS) which is the major manifestation of chronic lung allograft rejection. We specifically review the effect of infection, a risk factor for BOS, cytokines/chemokines in the pathogenesis of BOS, and the potential link between the allograft microbiome and immune responses that may mediate the development of BOS. Understanding the allograft microbiome and how it relates to the pathologic mechanisms of BOS may suggest targeted therapies to improve long-term survival post-lung transplantation. Keywords Lung transplantation . Bronchiolitis obliterans syndrome . Infection . Chemokines . Microbiome Problems of lung transplantation Lung transplantation is a therapeutic option for end-stage lung diseases. Lung transplant recipients (LTR) face higher mortality rates and more frequent loss of graft function as compared to other solid organ transplants, including the liver, kidney, and heart. The improvements in surgical techniques, immunosuppressive therapy, and postoperative management have increased the 1-year survival of LTR to approximately 80%, establishing transplantation as a treatment option for end-stage lung diseases [1]. However, the 5-year survival rate has remained approximately 50% and the 10-year graft survival rate is only 26% (the 2009 OPTN/SRTR Annual Report). This low survival rate is predominately due to chronic lung allograft rejection also known as bronchiolitis obliterans syndrome (BOS) [2]. In this review, we focus on BOS which is a major manifestation of chronic lung transplant rejection [3]. Although the precise mechanism(s) of BOS remains to be elucidated, allograft infection appears to stimulate aberrant allo-/autoimmune responses which may drive altered cytokine/chemokine expression, contributing to the development of BOS. Here, we review the effect of infection(s) on lung transplantation as well as cytokines/ chemokines which play a major role in directing alloantigen- primed T cells and other effector leukocytes into allografts, eventually causing rejection. In addition, we focus on the analysis of the microbiome as a means of identifying known and unknown microbes potentially associated with lung allograft rejection. We emphasize the potential link between the allograft microbiome and immune responses that may mediate the development of BOS. This article is published as part of the Special Issue on Transplantation and Tolerance. T. Nakajima : P. W. Finn(*) Division of Pulmonary and Critical Medicine, Department of Medicine, University of California, 9500 Gilman Drive 0643, La Jolla, CA 92093, USA e-mail: [email protected] V. Palchevsky : J. A. Belperio Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, CA, USA D. L. Perkins Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA Semin Immunopathol (2011) 33:135156 DOI 10.1007/s00281-011-0249-9

Transcript of Lung transplantation: infection, inflammation, and the microbiome

Page 1: Lung transplantation: infection, inflammation, and the microbiome

REVIEW

Lung transplantation: infection, inflammation,and the microbiome

Takeshi Nakajima & Vyachesav Palchevsky &

David L. Perkins & John A. Belperio & Patricia W. Finn

Received: 29 December 2010 /Accepted: 12 January 2011 /Published online: 27 January 2011# Springer-Verlag 2011

Abstract Lung transplantation is the only therapeuticoption for patients with end-stage pulmonary disorders.Despite the improvements in surgical techniques andimmunosuppressive therapy, allograft function and long-term survival are limited by the development of chroniclung transplant rejection. In this review, we focus onbronchiolitis obliterans syndrome (BOS) which is the majormanifestation of chronic lung allograft rejection. Wespecifically review the effect of infection, a risk factor forBOS, cytokines/chemokines in the pathogenesis of BOS,and the potential link between the allograft microbiome andimmune responses that may mediate the development ofBOS. Understanding the allograft microbiome and how itrelates to the pathologic mechanisms of BOS may suggesttargeted therapies to improve long-term survival post-lungtransplantation.

Keywords Lung transplantation . Bronchiolitis obliteranssyndrome . Infection . Chemokines .Microbiome

Problems of lung transplantation

Lung transplantation is a therapeutic option for end-stage lungdiseases. Lung transplant recipients (LTR) face highermortality rates and more frequent loss of graft function ascompared to other solid organ transplants, including the liver,kidney, and heart. The improvements in surgical techniques,immunosuppressive therapy, and postoperative managementhave increased the 1-year survival of LTR to approximately80%, establishing transplantation as a treatment option forend-stage lung diseases [1]. However, the 5-year survival ratehas remained approximately 50% and the 10-year graftsurvival rate is only 26% (the 2009 OPTN/SRTR AnnualReport). This low survival rate is predominately due tochronic lung allograft rejection also known as bronchiolitisobliterans syndrome (BOS) [2]. In this review, we focus onBOS which is a major manifestation of chronic lungtransplant rejection [3]. Although the precise mechanism(s)of BOS remains to be elucidated, allograft infection appearsto stimulate aberrant allo-/autoimmune responses which maydrive altered cytokine/chemokine expression, contributing tothe development of BOS. Here, we review the effect ofinfection(s) on lung transplantation as well as cytokines/chemokines which play a major role in directing alloantigen-primed T cells and other effector leukocytes into allografts,eventually causing rejection. In addition, we focus on theanalysis of the microbiome as a means of identifying knownand unknown microbes potentially associated with lungallograft rejection. We emphasize the potential link betweenthe allograft microbiome and immune responses that maymediate the development of BOS.

This article is published as part of the Special Issue on Transplantationand Tolerance.

T. Nakajima : P. W. Finn (*)Division of Pulmonary and Critical Medicine,Department of Medicine, University of California,9500 Gilman Drive 0643,La Jolla, CA 92093, USAe-mail: [email protected]

V. Palchevsky : J. A. BelperioDivision of Pulmonary, Critical Care Medicine, Allergy,and Clinical Immunology, Department of Internal Medicine,The David Geffen School of Medicine at UCLA,Los Angeles, CA, USA

D. L. PerkinsDivision of Nephrology, Department of Medicine,University of California, San Diego,La Jolla, CA, USA

Semin Immunopathol (2011) 33:135–156DOI 10.1007/s00281-011-0249-9

Page 2: Lung transplantation: infection, inflammation, and the microbiome

Bronchiolitis obliterans syndrome

Pulmonary complications after lung transplantation includehyperacute rejection, primary graft dysfunction, acuterejection, bronchial anastomotic obstruction, chronic rejec-tion, and infection. The main cause of the limited long-termsurvival is the development of chronic rejection [2]. BOS isthe clinical manifestation of chronic rejection which is afibroproliferative process that leads to an obstructiveairflow pattern and deterioration of allograft function [3].While the use of newer immunosuppressive strategiesimproves the treatment of acute rejection, there has beenno significant impact on BOS over the last 20 years. Theincidence of BOS approaches 50% within 5 years oftransplantation, and the median survival after the diagnosisof BOS is only 3 years [4]. Beyond the first year aftertransplantation, BOS accounts for over 25% of deaths. Theclinical features of BOS are progressive dyspnea and coughwith a progressive sustained decline in forced expiratoryvolume in 1 s (FEV1) that does not respond to broncho-dilators [5, 6]. The histological lesion of BOS is obliterativebronchiolitis, characterized by patchy fibroproliferation andobliteration of respiratory bronchioles, associated withperibronchiolar leukocytes, increased numbers of airwaymesenchymal cells, and increased deposition of extracellu-lar matrix [4].

The pathogenesis of BOS is complex and involves bothalloimmune and non-alloimmune mechanisms that actalone or in combination [7, 8]. Alloimmune-independentfactors which contribute to BOS are primary graft dysfunc-tion, allograft infection, and gastroesophageal reflux.Alloimmune-dependent factors, such as acute rejectionand lymphocytic bronchitis (LB), are known to besignificant risk factors for the development of BOS [5].In the early 1990s, it was recognized that peribronchiolarmononuclear cell infiltration sometimes accompaniedacute rejection, especially when the transbronchial biopsysamples were adequate. As such, LB represents airway-directed rejection. Multiple studies have identified LB asan important risk factor for chronic rejection independentof acute rejection. According to the International Societyfor Heart and Lung Transplantation definition, low-gradeLB is characterized as mononuclear cell infiltration withinthe bronchiolar submucosa without epithelial cell damageor intraepithelial mononuclear cell infiltration [9]. High-grade LB is characterized by submucosal and intraepithe-lial infiltration by mononuclear cells and eosinophils aswell as epithelial cell necrosis and metaplasia. Glanville etal. [10] identified the severity of LB as the mostsignificant risk factor for chronic rejection and anindependent risk factor for death. Acute rejection, LB,and infection(s) have consistently been identified as strongrisk factors for BOS [10–14]. Antibody-mediated rejection

may also contribute to BOS; however, further investiga-tion is needed [15]. All these risk factors for BOS augmentthe host response to the allograft in a manner similar tothat which occurs with the pulmonary host defense tomicrobes, suggesting a link between infection and lungallograft rejection.

Pulmonary host defense mechanisms

Physical barriers

The pulmonary host defense system is composed ofmultiple components, including physical barriers andimmune cells, which include epithelial cells, macrophages,neutrophils, and lymphocytes. Respiratory infection isgenerally caused by the direct invasion of microbes (e.g.,virus, bacteria, fungus). In order for pathogens to invade theupper or lower airways, they have to fight multiple physicaland immunologic barriers. The hair in the lining of thenasal track acts as a physical barrier and can potentially trapinvading organisms. The wet mucus inside the nasal cavitycan engulf microbes. The small hair-like structures (cilia)that line the lung airways constantly move any foreigninvaders up toward the pharynx to be eventually swallowedinto the stomach.

Immune responses and cytokines/chemokines

In addition to the above physical barriers, the immunesystem is important for fighting off invasion of patho-gens. The immune system defense mechanisms can bedivided into an innate (non-antigen-dependent) immuneresponse and an adaptive (antigen-dependent) immuneresponse. The innate immune response can be triggeredwhen microbes are identified by pattern recognitionreceptors, which recognize components that are con-served among broad groups of microorganisms, or whendamaged, injured, or stressed cells send out alarmsignals. The innate leukocytes, including phagocytes(macrophages, neutrophils, and dendritic cells), mastcells, eosinophils, basophils, and natural killer cells,identify and eliminate pathogens, either by contact/mediators or by engulfing and then killing microorgan-isms. Adaptive immunity, which is promoted by innateimmunity, is composed of lymphocytes, including T andB cells. The adaptive immune response requires therecognition of specific “non-self” antigens during aprocess called antigen presentation. Antigen specificityallows for the generation of responses that are tailored tospecific pathogens or pathogen-infected cells.

A delicate balance between pro- and anti-inflammatorycytokines is critical to lung wound repair from injury.

136 Semin Immunopathol (2011) 33:135–156

Page 3: Lung transplantation: infection, inflammation, and the microbiome

Changes in this balance can influence lung remodeling. Thespecific mechanisms that lead to lung dysfunction mayinvolve the interactions between type 1, 2, 9, and 17 cells/immune responses. Naive cells [i.e., CD4+ T helper cells(Th), CD8+ T cytotoxic cells (Tc), mononuclear phagocytes(M), and natural killer cells (NK)] can all differentiate intomultiple distinct cell subsets, [e.g., type 1 immune cells(Th1, Tc1, M1, and NK1) and type 2 immune cells (Th2, Tc2,M2, and NK2)]; which have distinct functions and cytokineexpression profiles [16–20]. Type 1 immune response ismainly associated with the cell-mediated immune responseand is identified by the production of IL-12 and IFN-γwhich drives a cytotoxic T lymphocyte (CTL) and delayed-type hypersensitivity response. Type 2 immune response isidentified by the production of IL-4, IL-5, and IL-13 andpromotes mucosal, allergic, and humoral immunity [17–20]. Type 9 immune response is induced by IL-4 and TGF-β which augments IL-9 expression and is involved inmucus production and tissue inflammation [16]. Type 17immune response is driven by several cytokines includingTGF-β, IL-6, IL-21, and IL-23, leading to the production ofIL-6, IL-8, IL-17A, IL17F, IL-22, and IL-26, all importantin extracellular pathogen containment and a neutrophilicinflammation [16].

The nature of the antigen and the pattern of cytokinesreleased into the microenvironment are considered to be themost important factors dictating whether the immuneresponse is directed toward a type 1, 2, 9, or 17 response.In addition, during specific disease states, type 1, 2, 9, and17 immune cells can cross-regulate each other through theirrespective cytokine profiles [16, 21, 22]. These immuneresponses are important in altering lung remodeling as wellas containing and eliminating microbes.

The predominant function of chemokines is to induce themigration and activation of the leukocytes [23]. Leukocytechemotactic and activating effects of chemokines are vitalto the development of effective host responses to an antigenand the subsequent shift from innate to polarized adaptiveimmunity with the generation of CD4+ T cell subsets (e.g.,Th1, 2, 9, and 17 cells; Fig. 1).

T regulatory cells

T regulatory cells (Tregs) are a specialized subpopulation ofT cells that act to suppress the activation of the immunesystem and thereby maintain immune system homeostasisand tolerance to self-antigens. Interest in Tregs has beenheightened by evidence from experimental mouse modelsdemonstrating that the immunosuppressive potential ofthese cells can be harnessed by microbes to evadecontainment as well as their potential therapeutic use totreat autoimmune diseases and facilitate tolerance [24](Fig. 1).

Infectious susceptibility of the allograft

Despite multiple defense processes, invading microbesadapt various mechanisms to resist destruction, allowingthem to exist in symbiosis with the airway/parenchymatissue without causing disease (colonization), or producedisease by breaking through the defense barriers andinvading into the airway/parenchyma. Compared with otherorgan transplantations, LTR are more susceptible toinfections due to the direct communication of the lungwith the external environment contributing to an imbalancebetween the host’s immune system and the virulence ofmicroorganism(s).

Predisposing infectious risk factors for the allograft areits constant exposure to the external environment, surgeryvia anastomotic problems (e.g., stenosis), early airwayischemia and reperfusion injury with delayed regenerationof airway function, abnormal lymphatic drainage, anti-rejection immunosuppressive medications, spillage ofmicrobes from naïve airways and sinuses, and the devel-opment of architecture distortion of the lung allograft frominfection and rejection episodes [25].

Bronchial anastomotic problems

Anastomotic airway complications have been mainlyattributed to ischemia of the donor bronchus during theimmediate post-transplant period and occur in about onefifth of patients following lung transplantation [26, 27]. Thebronchial anastomosis is susceptible to infections in partdue to its relative devascularization after transplant,disruption of lymphatic drainage, and altered alveolarphagocytic function. Furthermore, abnormal ciliary actionin the donor epithelium, bronchial stenosis, and lack ofcough reflex due to denervation may result in microbialcolonization and subsequent complications including de-hiscence, stenosis, malacia, fistulas, granulation tissue,abscess formation, and mediastinitis.

Cystic fibrosis

Although the number of lung transplants has increasedsubstantially in the past two decades, the leading indica-tions for lung transplantation remain relatively unchanged:chronic obstructive pulmonary disease (COPD), idiopathicpulmonary fibrosis, cystic fibrosis (CF), alpha1-antitrypsindeficiency with emphysema, and pulmonary arterial hyper-tension [1]. Patients with CF have excellent survival rateswhich are similar to those with COPD after lung transplan-tation. However, they are still prone to common complica-tions including graft dysfunction and a variety of infections.Patients with CF are known to have their upper airways andsinuses colonized by Pseudomonas species, and subsequent

Semin Immunopathol (2011) 33:135–156 137

Page 4: Lung transplantation: infection, inflammation, and the microbiome

downstream infection in the allograft lung is common. Inpatients receiving single lung transplants, the remainingstructurally altered native lung may harbor infection andseed the allograft [28]. The high propensity of CF patientsto re-colonization led to speculations regarding paranasalsinuses acting as a reservoir for bacterial pathogens, leadingto allograft reinfection. Patients with CF frequently havemulti-organ involvement and comorbid conditions, includ-ing malnutrition, chronic infections of the upper respiratorytract, and colonization with resistant pathogens. Coloniza-tion with multidrug-resistant Pseudomonas aeruginosa,Staphylococcus aureus, Stenotrophomonas maltophilia,and Aspergillus fumigatus are not contraindications fortransplantation because there is no evidence of effect onpost-transplant survival [29, 30]. The exception is Burkhol-deria cepacia genomovar III, which is associated with anunacceptably high risk of post-transplant mortality [31, 32].Patients with multidrug-resistant pathogens should undergofrequent evaluation for changes in their colonizing micro-flora and resistance patterns. Analysis of the microfloraguides early prophylaxis and treatment in the immediatepostoperative period. Aspergillus species are isolated beforetransplantation in 10–25% of patients with CF. Aspergilluscolonization pre-transplant can be in the form of intra-cavitary aspergilloma in patients with CF. There is lessinformation available on the significance of colonization orinfection with fungi in CF. However, subpleural cavitaryaspergillomas potentially cause empyema and decreasedpost-transplant survival. Aggressive pre- and post-transplant therapy can lead to successful transplantation[33]. Nontuberculous mycobacteria are isolated in 10–15%of CF patients prior to lung transplantation [34]. Mycobac-

terium abscessus is inherently resistant to many antibioticsand has a propensity to infect soft tissues. Individualizedapproaches are need for patients with M. abscessus whorequire lung transplantation.

Rejection

Graft rejection has historically been classified into hyper-acute rejection, acute rejection, and chronic rejection.Hyperacute rejection is a complement-mediated responsein recipients with preexisting antibodies to the donor andoccurs within minutes of reperfusion. Hyperacute rejectionis only encountered when a solid organ graft is inadver-tently transplanted across an ABO blood group barrier orwhen the recipient has been sensitized to alloantigen fromprevious exposure to blood products, past pregnancies, or afailed transplant. The clinical approach to this problem isbased on prevention through appropriate donor–recipientmatching. Acute rejection involves T cells infiltrating theallograft, predominantly in the perivascular and peribron-chiolar regions. The risk of acute rejection is highest in thefirst 3 months after transplantation. However, acute rejec-tion can also occur months to years after transplantation. Asingle episode of acute rejection is not a cause for concernif recognized and treated promptly and rarely leads to organfailure. But recurrent episodes are associated with chronicrejection. The hallmark of chronic rejection is the develop-ment of irreversible scarring, compromising the lumen ofthe small airways and the vessels. The small allograftairways develop a dense scar occluding their lumen as aresult of persistent immunologic injury. While the smallallograft airways obliterate, the large allograft airways

Fig. 1 Differentiation of CD4+ T cell subsets. Upon stimulation, naïveCD4+ T cells differentiate into five main subsets: Th1, Th2, Th9, Th17,and T regulatory cells (Tregs). IFN-γ and IL-12 induce the formation ofIFN-γ producing Th1 cells. IL-4 induces Th2 cells, producing IL-4, IL-5, and IL-13. TGF-β induces Tregs, an immune-modulating subset of

CD4+ T cells. Interestingly, in the presence of TGF-β, IL-4 induces thedifferentiation of IL-9 producing Th9 cells, and IL-6 induces Th17 cells.IL-21 and IL-23 promote the development of Th17 cells. Each subset isgenerally (but not exclusively) associated with the disease processes andpathogens listed

138 Semin Immunopathol (2011) 33:135–156

Page 5: Lung transplantation: infection, inflammation, and the microbiome

develop cylindrical bronchiectasis. Such changes result inaltered airflow, decreased mucus clearances, and loss ofciliated respiratory epithelial cells. These airways are easilycolonized by gram negative rods, particularly Pseudomonas.

Immunosuppression (immunosuppressive drug)

Most lung transplant recipients are maintained on life-long pharmacologic immunosuppression, and there is asmall subset of patients who become tolerant to theirgraft. The majority of immunosuppressive regimens inclinical use exert their effect by blocking the pathwaysinvolved in the clonal expansion of alloreactive T cells orby reducing lymphocyte subpopulations. Insufficientimmunosuppression results in graft loss due to rejection,while excess immunosuppression results in increasedmorbidity and mortality from infections. The discoveryof the calcineurin inhibitor cyclosporine A (CSA)ushered in the modern era of successful solid organtransplantation. However, CSA and other current drugsused as immunosuppressants are toxic, relatively nonspe-cific regarding donor/recipient interactions, and causeproblems that contribute to morbidity and mortality aftertransplant. The complications of immunosuppressantsoccur both because of specific organ toxicity (especiallyrenal) and the increased risk of infections [35]. Immuno-suppression for lung transplantation can be consideredunder three clinical contexts: maintenance immunosup-pression, induction therapy, and anti-rejection treatment.In maintenance immunosuppression, triple-drug therapywith a calcineurin inhibitor (cyclosporine or tacrolimus),an antimetabolite (azathioprine or mycophenylate), and acorticosteroid has been the standard. Mammalian target ofrapamycin (mTOR) inhibitors that are in clinical useinclude sirolimus and everolimus. This new class of drugsacts by interfering with T cell proliferation by blocking akinase, causing cell cycle arrest [36, 37]. Early use ofmTOR inhibitors in lung recipients has been limited due toits antifibrotic effects on bronchial wound healing [38].However, the new agents have found utility in replacingthe traditional antimetabolites in triple-drug regimensbecause of their antifibrotic effects on progression ofBOS [39]. Induction agents consist of several classes ofantibodies that exhibit protective effects when adminis-tered in the peri-transplant period. Polyclonal and mono-clonal antibodies (anti-lymphocyte globulin, anti-thymocyte globulin, CD3 antibody, and IL-2 receptorantibody) have been approved for clinical use as inductionagents. Treatment of acute rejection is intensification ofimmunosuppression which is generally a “steroid pulse”consisting of 3 days of high-dose intravenous cortico-steroids, followed by a slow steroid taper back to previouslevels. Refractory acute rejection is treated with the use of

induction agents. Most clinicians attempt to both intensifyand modify the immunosuppression regimen for patientswith chronic rejection. While much clinical success hasbeen made in the treatment of acute rejection, littleprogress has been made in the treatment of chronicrejection. Although immunosuppressive therapies renderthe patients at high risk for opportunistic infection, theirbenefit for BOS is relatively-scarce. An ideal approach topreventing rejection would be to introduce a state oftolerance to the graft rather than generalized suppressionof acquired immunity. Most immunosuppressive agentsinhibit T cell activation and therefore may simultaneouslyinhibit the generation of Tregs which play an importantrole in transplant tolerance. Studies show that calcineurininhibitors diminish or abrogate Tregs activity, whereasmTOR inhibitors do not [40, 41].

Infection and BOS

Bacterial

Airway infection itself is also a reported risk factor forprogression to BOS. Bacterial bronchopulmonary infectionsare the most frequent infectious complication after lungtransplantation and comprise approximately half of allinfectious complications. In recent studies, bacterialmicrobes were isolated in up to 80% of transplant recipients[25, 42, 43]. Although antibiotic prophylaxis and a promptadjustment of the antibiotic regimen based on the isolatedmicroorganism has reduced the frequency of bacterialpneumonia, the risk of pneumonia remains marked in thelater period after lung transplantation. Late onset ofbacterial pneumonia has an association with BOS. Further-more, episodes of pulmonary infection in LTR with BOShave been associated with a faster progression through BOSstages and to death [44].

P. aeruginosa is one of the most commonly isolatedpathogen in bacterial pneumonia after lung transplanta-tion as it can be found in up to 39.4% of cases [42, 45,46]. Recipients with CF have a higher susceptibility topulmonary P. aeruginosa colonization/infection comparedwith recipients without CF [47]. Later in the time courseafter lung transplantation, the frequency of P. aeruginosacolonization in airways increases and can be associatedwith BOS, while colonization with other Gram-negativebacilli is not associated with the subsequent developmentof BOS [48]. Allograft colonization by P. aeruginosaincreases bronchoalveolar lavage fluid (BALF) IL-8 andneutrophilia and is associated with BOS. [49]. Vos et al.[50] observe in their single-center study that post-transplant P. aeruginosa colonization is an independentrisk factor for BOS and responsible for a worse survival

Semin Immunopathol (2011) 33:135–156 139

Page 6: Lung transplantation: infection, inflammation, and the microbiome

with BOS. Although there is insufficient evidence of theimpact of pre-transplant P. aeruginosa colonization onpost-transplant mortality, patients with a history ofrecurrent sinusitis should be evaluated for drainage pre-transplantation.

B. cepacia complex (BCC) infection has historically beenassociated with poor outcomes after lung transplantation [51,52]. Many transplant centers consider infection with Bur-kholderia species a contraindication to lung transplantationin CF. Among BCC, Burkholderia cenocepacia (genomovarIII) and Burkholderia multivorans (genomovar II) accountfor the majority of infections. Boussaud et al. [53] recentlyreport that B. cenocepacia is associated with an increasedearly post-transplant morbidity and mortality. Alexander etal. [54] also show that B. cenocepacia infection status issignificantly associated with decreased survival at 1 yearcompared with those infected with BCC other than cenoce-pacia. Other studies show that patients infected with B.multivorans have a similar outcome to uninfected patients[55]. The role that B. cenocepacia plays in the pathogenesisof BOS remains uncertain.

Mycobacterium tuberculosis is an infrequent but impor-tant complication, afflicting 1–6% of patients [56, 57]. M.tuberculosis may be due to reactivation, occult disease inthe remaining native lung after single lung transplantation,or transmission by the transplanted lung. Identification oflatent tuberculosis infection prior to transplantation isparticularly important. Therapy is challenging due tocomplex drug interactions with immunosuppressive agents.Current guidelines recommend tuberculin skin test screen-ing in all transplant candidates followed by radiographictesting to rule out active disease.

Fungal

Fungal infection occurs in 15–35% of LTR, and over80% are caused by Candida and Aspergillus with anoverall mortality rate of nearly 60% [58–63]. WhileCandida mainly infests the upper tracheobronchial treewith only an occasional chance of dissemination, Asper-gillus has the potential to involve the deeper parenchyma.Colonization with Aspergillus species occurs in 22–85%of LTR. Invasive aspergillosis occurs in 13–26% of thecolonized LTR and is uniformly fatal. Aspergillusfumigates is the most commonly isolated species andcauses the majority of invasive diseases which may arisein the native lung after single-lung transplantation.Weight et al. [64] suggests a potential role of Aspergilluscolonization in stimulating airway inflammation leadingto the development of BOS. Significant risk factors forinvasive aspergillosis are previous cytomegalovius(CMV) disease, colonization with Aspergillus, and BOS[65–67].

Viral

Khalifah et al. [68] demonstrate that respiratory viralinfections are distinct risk factors for BOS. Bharat et al.[69] demonstrate that respiratory viruses induce suppres-sive Treg apoptosis which leads to the development ofautoimmunity and contributes to the immunopathogenesisof BOS both in humans and murine models of chronicrejection.

CMV is one of the most important and common viralinfections after lung transplantation [70]. CMV is foundin a latent stage in 60% of all adults. Routine screening ofdonor and recipient for CMV IgG allows stratification ofthose at highest risk of CMV reactivation and pneumoni-tis. Transplantation of CMV-seropositive donor lungstransmits a significant viral load to recipients. Recipientswith evidence of previous/latent CMV infection (seropos-itive recipients) are at risk, while seronegative recipientstransplanted with a seropositive pulmonary graft are athighest risk of developing CMV infection and disease afterlung transplantation. CMV infection and disease are themost relevant risk factors for the development of BOS [6].Interestingly, pulmonary CMV increases several chemo-kines associated with BOS and mortality post-lungtransplantation [71]. Strategies for prophylaxis, as wellas early diagnosis and treatment of CMV disease, may becrucial for further improvement of the results of lungtransplantation.

Adenovirus infection poses a serious problem in thepediatric LTR [72]. At least half of the patients die inrespiratory failure because of diffuse alveolar damageinduced by the virus. Occasional cases of adults withadenovirus infections are documented. In most of them, theclinical course is fatal and obliterative bronchiolitis devel-ops in the survivors [73, 74].

Epstein–Barr virus (EBV) is an oncogenic virus and isstrongly associated with post-transplant lymphoprolifer-ative disease (PTLD) [75, 76]. PTLD is believed to occurin almost half of the patients who are EBV-seronegativeand receive an EBV-seropositive graft. PTLD is thought tobe a lymphoproliferation of EBV-infected B cells arisingin the setting of over-immunosuppression. The relation-ship between active EBV infection and transplant dys-function or BOS is not clear.

Immune responses to an allograft

The CD4+ T cell plays a pivotal role in orchestrating theimmune response to an allograft. There are two pathwaysby which alloreactive T cells recognize foreign antigenand become activated: direct allorecognition and indirectallorecognition [77] (Fig. 2). The direct pathway involves

140 Semin Immunopathol (2011) 33:135–156

Page 7: Lung transplantation: infection, inflammation, and the microbiome

donor antigen-presenting cells (APCs) that are carried as“passenger leukocytes” into the recipient. Direct allor-ecognition occurs when the recipient’s CD4+ T cellrecognizes intact donor major histocompatibility complex(MHC) molecules on APCs of donor origin. AlloreactiveT cells infiltrate the allograft and release cytokines/chemokines, which activate other cells (e.g., macrophages,B cells, and plasma cells) and elicit the release of growthfactors, cytotoxic mediators, and alloantibodies, all cul-minating in allograft injury [78]. Passenger leukocytes areno longer present in the allograft within weeks/monthspost-transplantation, ending the direct pathway of allor-ecognition. Thus, this recognition is more relevant to theprocess of acute rejection. Indirect allorecognition occurswhen recipient APCs processes alloantigens in theconventional manner, and the resultant peptide fragmentsare presented to recipient CD4+ T cells in the context ofself-MHC molecules. Because all of the parenchymal cellsof an allograft can serve as substrate for antigen process-ing by recipient APCs, indirect allorecognition is an everpresent mechanism in the long-term alloresponse of a hostto a graft and is important in the pathogenesis of chronicrejection [79, 80] (Fig. 2).

The role of type 1 cytokines during the pathogenesisof lung allograft dysfunction

IL-1 and TNF-α are proximal pro-inflammatory type 1cytokines that increase many pro-inflammatory molecules,including chemokines and adhesion molecules [81–86]. IL-1 and TNF-α have both been implicated in ischemia–reperfusion injury (IRI) and acute and chronic rejection inhuman studies as well as animal models of lung and airwayallograft dysfunction [87–90]. In animal models, theinhibition of either cytokine can attenuate allograft dys-function [87, 91], suggesting that current biologics avail-able to inhibit these cytokine pathways might beinformative in human lung allograft dysfunction.

IL-12 is important for the promotion of cell-mediatedtype 1 immune response and has been shown to induceIFN-γ during an allogeneic response [92–104]; however,the exact role of the IL-12 family during allograft rejectionremains controversial. Some studies suggest IL-12 causesallo-injury while others do not [92, 93, 97, 103–111]. Withregard to lung allograft dysfunction, overexpression of IL-12 accelerates airway allograft injury [112]. Similarly, in atranslation/longitudinal study of 44 human LTR, those with

Fig. 2 Direct and indirect allorecognition. Full CD4+ T cell activationrequires both a primary signal from the T cell receptor (TCR)recognition of peptide antigens and a second signal from theinteraction of co-stimulatory molecules (CD80/CD86 and CD28). Tcells recognize alloantigens by two distinct pathways known as thedirect and indirect. a Direct allorecognition occurs when therecipient’s CD4+ T cell recognizes intact donor major histocompati-bility complex (MHC) molecules on antigen-presenting cells (APCs)of donor origin. As a result, direct allorecognition between host T cells

and transplanted cells leads to sensitization, proliferation, andcytotoxic response against the graft. This recognition is relevant tothe process of acute rejection. b Indirect allorecognition occurs whenrecipient APCs processes alloantigens in the conventional manner, andthe resultant peptide fragments are presented to recipient’s CD4+ Tcells in the context of self-MHC molecules. Persistent T cellalloresponses to antigens on recipient APCs and to donor APCMHC proteins may be a major driving force for cellular death andchronic allograft rejection

Semin Immunopathol (2011) 33:135–156 141

Page 8: Lung transplantation: infection, inflammation, and the microbiome

elevated levels of IL-12 in BALF were more likely todevelop BOS [113], supporting a role for IL-12 in thepathogenesis of BOS.

IFN-γ is a type 1 cytokine that can be induced by IL-12and inhibited by IL-10 [114–116]. Using a rat heterotopictracheal transplantation model of BOS, IFN-γ is persistent-ly elevated during fibro-obliterative events [117]. However,other animal models of acute and chronic rejectiondemonstrate a few unanticipated results. Using an immu-nosuppressed murine cardiac transplantation model, IFN-γ−/− recipients demonstrate accelerated acute parenchymalrejection [118–120]. Interestingly, these allografts demon-strate decreased chronic transplantation coronary arteryrejection [118–120]. Collectively, these studies suggest arole for IFN-γ, in part, by limiting acute allograft rejectiondue to anti-proliferative effects on T cells and decreasedCTL lytic activity against allogeneic targets. In contrast,IFN-γ secretion by activated infiltrating cells may, in part,worsen chronic rejection by increasing integrin and MHCclass II on the surface of allograft endothelial cells,culminating in the accumulation of activated macrophagessecreting pro-fibrotic mediators.

Pertaining to human lung transplantation, elevatedexpression of IFN-γ from BALF is associated with acuteand refractory lung allograft rejection, both risk factors forthe development of BOS [121, 122]. In addition, asignificant correlation is detected between the presence ofa high expressing human polymorphism at position +874 ofthe IFN-γ gene and BOS [123]. More animal and humanstudies will be required to determine the exact influence ofreceptor/IFN-γ biology during the continuum of early tolate lung allograft dysfunction.

IL-15 is another type 1 pro-inflammatory cytokine that ispredominately produced by activated mononuclear phag-ocytes [124–127]. Under physiological conditions, IL-15 isinvolved in the differentiation and proliferation of NK andT cells [124–132]. Increased intragraft expression of IL-15is associated with human acute rejection in both kidney andcardiac allografts [133–136]. Studies of IL-15 biology areneeded to evaluate its potential role in lung allograftdysfunction.

The role of type 2 cytokines during the pathogenesis oflung allograft dysfunction

IL-10 is a type 2 cytokine with immunomodulatorybioactivity including the inhibition of cytotoxicity and thedown-regulation of MHC class II antigens and pro-inflammatory cytokine production [116, 137–150]. Therole of IL-10 in modulating the response to allograftrejection is unclear [119, 151–160]. Nonetheless, augmen-tation of IL-10 is to be protective in multiple models oflung IRI [161–163]. Similarly, overexpression of IL-10

attenuates acute lung allograft rejection when used in a ratorthotopic single-lung transplantation model with or with-out the addition of cyclosporin [164]. Moreover, neutrali-zation of IL-10 in a rat heterotopic tracheal transplantationmodel accelerates airway obliteration, whereas the admin-istration of physiologic doses of IL-10 attenuates allograftairway fibroplasia [165–168]. With regard to human LTR,the increased IL-10 production genotype (GCC/GCC)protects recipients against acute rejection when comparedwith the intermediate or decreased IL-10 productiongenotypes [169]. The discrepancy in the effect of IL-10during allograft dysfunction may be related to dose(physiologic versus super-physiologic), type (viral versuscellular IL-10), timing of the dose, and the compartment(systemic or local administration) by which IL-10 isdelivered.

IL-13 is a potent type 2 cytokine elevated in the BALFfrom patients with BOS [170, 171]. Importantly, BALF IL-13 levels are biologically active as ex vivo experimentsdemonstrate that IL-13 induces fibroplasia [170, 171].Translational studies in rodent models of BOS demonstratethat inhibition of IL-13 biology attenuates airway allograftobliteration [170, 171]. IL-13 may be a target to investigateregarding the potential to improve long-term survival post-lung transplant.

The role of type 17 cytokines during the pathogenesis oflung allograft dysfunction

IL-6 is a cytokine with both pro-inflammatory and anti-inflammatory properties and has been associated with aTh17 profile and fibrogenesis [16, 21, 22, 172–176]. In arodent lung allograft dysfunction model, IL-6 has a bimodaldistribution (e.g., increased with IRI and then during acuterejection) [177, 178]. Similarly, elevated levels of humanIL-6 in BALF are associated with refractory acute lungallograft rejection as well as BOS [178, 179]. These studiesunderscore the possible role for IL-6 during the pathogen-esis of BOS [123].

While TGF-β has a strong history of immunosuppres-sive activity, it is the most potent inducer of collagensynthesis, fibroblast proliferation, and fibroblast chemotaxisand is recently associated with a Th17 profile [16, 180,181]. Studies involving solid organ transplantation demon-strate that TGF-β has beneficial effects during acuteallograft rejection [182–184]. Conversely, the administra-tion of adenoviral-mediated soluble TGF-β IIIR; a func-tional TGF-β antagonist, attenuates rodent BOS [185].Similar results are seen with Smad3−/− (Smad3 is asignaling pathway that mediate many of the fibrogeniceffects of TGF-β1) recipients in a rodent model of BOS[186]. Human studies involving lung transplantation showaugmented expression of TGF-β to be an early marker of

142 Semin Immunopathol (2011) 33:135–156

Page 9: Lung transplantation: infection, inflammation, and the microbiome

BOS that may correlate with the severity of luminal fibrosis[187–189]. More human studies involving TGF-β and lungtransplantation will be required to determine whether itplays a specific role in BOS.

IL-17 is a pro-inflammatory cytokine that is predomi-nately produced by T cells [190–193]. IL-17 stimulatesmultiple cell types to express IL-6, IL-1β, CXCL8, TNF-α,G-CSF, CCL2, and PGE2 [193, 194]. Excessive amounts ofIL-17 cells are thought to play a key role in autoimmunedisease. A more natural role for IL-17 is suggested bystudies which demonstrate preferential induction of IL-17in cases of host infection with various bacterial and fungalspecies. Th17 cells primarily produce two main members ofthe IL-17 family, IL-17A and IL-17F, which are involved inthe recruitment, activation, and migration of neutrophils[195, 196].

IL-17 stimulates proliferation of alloreactive T cells andthe maturation of dendritic cells (DC) [197]. Burlingham etal. [198] demonstrate that de novo autoimmunity mediatedby collagen (V)-specific Th17 cells may contribute to theprogressive airway obliteration seen in human BOS.Furthermore, in a human cross-sectional study, BALF cellpellet expressions of IL-23 and IL-17 are associated withBOS [199]. Anti-IL-23 antibody impairs the function of IL-23 in inducing IL-17 production in a rat model of airwayallograft rejection [200]. Overall, these studies suggest arole for the type 17 immune response during the pathogen-esis of BOS.

The role of receptors/chemokines during the pathogenesisof lung allograft dysfunction

The hallmark of allograft dysfunction is the infiltration ofleukocytes to the lung allograft. The ability to maintainleukocyte recruitment throughout the continuum of early(IRI/primary graft dysfunction and acute lung allograftrejection) to late (BOS) lung allograft dysfunction despiteaggressive immunosuppression is pivotal in the transitionfrom inflammation/immune responses to fibroplasia of theallograft. This persistent elicitation of mononuclear cellsrequires intercellular communication between infiltratingleukocytes, endothelium, parenchymal cells, and compo-nents of the extracellular matrix. These events are mediatedvia the generation of adhesion molecules, cytokines, andchemokines. The chemokines, by virtue of their specificcell surface receptor expression, can selectively mediate thelocal recruitment/activation of distinct leukocytes/cells,allowing for migration across the endothelium and beyondthe vascular compartment along established chemotacticgradients.

The chemokine superfamily is divided into four sub-families (C, CC, CXC, and CX3C) based on the presence ofa conserved cysteine residue at the NH2-terminus [201–

203]. CXC chemokines have been further subdivided onthe basis of the presence or absence of the sequenceglutamic acid–leucine–arginine (ELR) near the NH2-termi-nal. ELR+ chemokines are neutrophil chemoattractants withangiogenic properties. ELR-CXC chemokines are chemo-attractants of lymphocytes with angiostatic properties [204–208]. CC chemokines predominantly recruit mononuclearcells [201, 209]. The C subfamily consists of XCL1 andXCL2, which attract lymphocytes, while CX3CL1 is theonly member of the CX3C subfamily, and its domain sits ona mucin stalk allowing for cellular adhesion [210–213].

All chemokine action is mediated through seven-transmembrane-spanning G protein-coupled receptors [23].These heterotrimeric G proteins are composed of β- and -subunits. The chemokine receptors generally undergointernalization and phosphorylation following ligand bind-ing. Interaction of a ligand with its receptor leads to theexchange of GTP for GDP and the dissociation of the α-subunit from the β -subunit. The dissociated Gα and Gβ

can activate downstream signal transduction events [214,215].

Receptors/CC chemokines

In humans, elevated levels of CCL5 are associated withacute and chronic lung allograft rejection [216–218]. BothCCL3 and CCL5 are increased in an animal model of IRI,and in vivo neutralization of either chemokine reduces lunginjury [219]. In vivo depletion of CCL5 attenuates acuteand chronic lung allograft rejection in multiple animalmodels [117, 217, 218, 220, 221].

With regard to CCR2/ligand biology, CCL2 is associatedwith the continuum of human acute to chronic lungallograft rejection [220]. Rodent models of lung allograftdysfunction indicate that the inhibition of CCR2/ligandbiology attenuates IRI and acute/chronic lung allograftrejection by reducing allograft mononuclear phagocyteinfiltration [220–222].

Forkhead box P3 (FOXP3) is a gene that encodes aforkhead/winged helix transcription factor Scurfin, which isrequired for Treg development and thus is a marker of Tregcells [24, 223]. Lee et al. [224] evaluated the presence ofTregs (allograft Foxp3 expression) in tolerized (anti-CD154with donor-specific transfusion) allografts as compared tountreated allografts and as compared to isografts and naïvehearts. They found a hierarchy of Foxp3 and CCR4expression (anti-CD154 with donor-specific transfusion>>> untreated allografts >> isografts>naïve hearts). Cardiacallografts from CCR4−/− recipients treated with (anti-CD154with donor-specific transfusion therapy) have significantreductions in Foxp3 expression and reject their cardiac graftsat a normal rate. This suggests that CCR4, in this modelsystem, is critical for Treg recruitment. It will be important to

Semin Immunopathol (2011) 33:135–156 143

Page 10: Lung transplantation: infection, inflammation, and the microbiome

determine the specific detrimental and protective mecha-nisms of CCR4/ligand biology during the pathogenesis oflung allograft dysfunction.

CCR7 and its ligands CCL19 and CCL21 play a role, inpart, in the localization of antigen-loaded DC and antigen-specific T cells within T cell-rich zones, which is critical forthe initiation of an adaptive immune response [225–228].CCR7−/− BALB/c donor hearts to CCR7−/− C57BL/6recipients prolong allograft survival to 14 days and theaddition of low-dose CSA prolongs survival to 16 days[225]. Overall, CCR7 is a key regulator of DC/T cellinteraction within secondary lymphoid tissue/organs forboth the initiation of an antigen-specific T cell response andfor the initiation of alloantigen-induced cytotoxic T cellresponses. More recent human studies demonstrate thatCCR7/ligand interactions are important for Treg recruit-ment to the lung allograft associated with the prevention ofBOS [229, 230].

Meloni et al. [231] evaluates CC chemokines in BALF ofpatients in the first post-transplant year and demonstrates thatCCL19, CCL20, and CCL22 levels predict BOS onset. Weigtet al. [71] show that the alteration of BALF levels of CCL2,CCL3, and CCL5 during pulmonary CMV infection of thelung allograft predicts outcomes. Elevated levels of CCL3 inBALF are protective with regards to survival and elevatedlevels of CCL2 in BALF predicts the development of BOS,while elevated levels of CCL5 in BALF predicts an increasein mortality post-lung transplant. These studies suggest apossible utility of BALF chemokines as biomarkers forguiding risk assessment post-lung transplantation.

CX3CL1/CX3CR1

CX3CL1 is involved in direct leukocyte activation, chemo-taxis, and adhesion through its interaction with CX3CR1[210–213, 232–236]. Although there are currently nostudies of CX3CL1 and lung rejection, studies of CX3CL1show its importance during murine cardiac rejection. Inthese studies, CX3CL1 localize to the endothelium, epicar-dium, endocardium, myocardium, and infiltrating mononu-clear cells in allografts [210, 237]. In vivo antibodydepletion of CX3CL1 increases allograft survival withoutgross disruption in graft leukocyte infiltration [210].Interestingly, when CX3CR1

−/− recipients are used, thereis no effect on allograft survival, even though there is areduction in graft infiltrating NK cells [237]. The additionof CSA to the CX3CR1

−/− mice prolongs graft survival by18 days. Together, these studies suggest that NK cells of theinnate immune system and T cells of the adaptive immunesystem act synergistically to induce rejection. Also,CX3CL1 (both soluble and tethered from recipient/donorcells)/CX3CR1 interactions are important for the recruit-

ment of NK cells to the allograft. Future studies involvingCX3CL1/CX3CR1 interactions may demonstrate its impor-tance in lung allograft dysfunction.

Receptor/CXC chemokines

Interferon-inducible ELR- CXC chemokines are importantin the pathogenesis of rejection due to their chemoattractionto activated T cells [238–243]. These chemokines (CXCL9,CXCL10, and CXCL11) bind and activate through theirshared receptor, CXCR3 [205, 244, 245]. IncreasedCXCR3/ligand biology is associated with human androdent lung allograft dysfunction. Multiple experimentalmodels of allograft injury demonstrate that down-regulationof this chemokine biology inhibits allograft injury, predom-inately by reducing the recruitment of T cells and NK cellsto the allograft [121, 246–256].

Thus far, CXCL13 is a CXCR5 exclusive ligand [257].In vivo neutralization of CXCL13 leads to a ninefoldreduction of cardiac allograft infiltrating double-negative(DN) Tregs, while their homing into the spleen and lymphnodes appears to be unaffected [258]. Overall, this studydemonstrates that the CXCR5/CXCL13 biological axisplays an important role in DN Treg homing into cardiacallografts, but not secondary lymphoid organs. Overall,these data suggest that this axis may have an important rolein the generation of tolerance.

CXCR2/ligands are important in neutrophil recruitmentand angiogenesis. In human studies, CXCR2/ligands areimportant in the continuum of primary graft dysfunction toacute and chronic lung allograft rejection [187, 259–267].Similar results are found in rodent studies. The CXCR2/ligands are important for neutrophilic-dependent earlyallograft dysfunction and the vascular remodeling thatsupports BOS [259, 260].

In summary, the above human studies demonstrate theimportance of receptors/cytokines/chemokines in transplan-tation allograft dysfunction. Also, studies in animal modelsof allograft dysfunction indicate proof of principle thatreceptor/cytokine/chemokine interactions are crucial inmediating leukocyte infiltration that contribute to allograftdysfunction. Future studies of other receptors/cytokines/chemokines may foster greater understanding in thepathogenesis of lung allograft dysfunction.

The role of Tregs during the pathogenesis of lung allograftdysfunction

A decreased percentage of CD4+FOXP3+ Tregs in the BALfrom LTR is associated with the development of BOS [268].A more distinct population of Tregs (CCR7+CD3+CD4+CD25(hi)FOXP3+CD45RA− lymphocytes) is protective against the

144 Semin Immunopathol (2011) 33:135–156

Page 11: Lung transplantation: infection, inflammation, and the microbiome

development of BOS [229, 230]. Respiratory viruses induceTreg apoptosis both in humans and murine models of chronicrejection [69]. A decreased number of Tregs correlates withmore rapid onset of BOS [69]. The therapeutic application ofTregs with a potential for increased long-term survival post-lung transplantation remains to be investigated.

Microbiome

Definition

The microbiome is defined as the totality of microbes, theirgenetic elements (genomes), and environmental interactionsin a specific environment. Traditional microbiology hasfocused on the study of individual species as isolated units.However, many organisms have not been successfullycultured, identified, or otherwise characterized becausetheir growth is dependent upon a specific microenviron-ment that cannot be reproduced experimentally. Advancesin DNA sequencing technologies have created a new fieldof research, termed metagenomics, allowing comprehensiveexamination of microbial communities, even those com-prising uncultivable organisms. The metagenomic approachfacilitates comprehensive characterization of the humanmicrobiota and analysis of its role in human health anddisease.

Immune response and microbiome

Although the immune system is classically thought to haveevolved to protect from infection by microbial pathogens,humans and living organisms peacefully coexist with a vastand complex microbiota that extensively interact with theimmune system. Basic development features and functions ofthe immune system depend on interactions with the micro-biome [269]. If some bacteria are critical for shaping ahealthy immune system, the absence of these organisms maylead to disease. The impact of the microbiota on human healthis best exemplified by studies of inflammatory bowel disease(IBD), such as Crohn’s disease and ulcerative colitis [270–272]. IBD has classically been associated with increases inpro-inflammatory cytokines such as TNF-α and IFN-γ.Recently, Th17 cells and Tregs have been implicated in thepathogenesis of colitis [273–275]. Germ-free animals havedefective Th17 cell development in the small intestine, andthe reduction in IL-17 production is associated with areciprocal increase in the number of CD4+Foxp3+ Tregs[276]. Treatment of mice with colitis with probiotic bacteriaincreases the production of IL-10 and the number of Tregs[277, 278]. The concomitant reduction in intestinal inflam-mation with probiotic administration suggests a link between

a decrease in a particular species from microbiota and thedevelopment of disease

Microbiome in patients with asthma and COPD (analysisof bronchoscopic samples)

Regarding the microbiome in lung, recent studiesindicate that changes in the airway microbiome mayimpact the development of allergic inflammation andCOPD exacerbations [279, 280]. An analysis of theairway microbiota by bronchoscopic examination inpatients with asthma, COPD, and controls indicates thatthe bronchial tree is not sterile. Pathogenic proteobac-teria, particularly Haemophilus spp., are more frequent inpatients with asthma and COPD than controls. Converse-ly, Bacteroidetes, particularly Prevotella spp., are morefrequent in controls [279]. Analysis of BAL by 16SrRNA PhyloChip analysis, a culture-independent micro-array, from COPD patients who were being managed forsevere respiratory exacerbations revealed a much greaterdiversity of bacteria than has previously been appreciat-ed. Members of Pseudomonadaceae, Enterobacteria-ceae, and Helicobacteraceae are present in the airwayof COPD patients. The identification of a diverse airwaybacterial community in respiratory samples from COPDpatients suggests that the pathogenesis of these eventsmay involve a polymicrobial process [280]. The specificbacterial species remain unknown and further studies arenecessary.

Microbiome in patients with cystic fibrosis (analysis of sputumsamples)

CF is one of the most important indications for lungtransplantation, and an understanding of the microbiomeof airway infection in CF patients may improve themanagement of lung transplantation. Although only thesputum of CF patients have been studied to date, recentdata derived from molecular culture-independent analysesindicate the presence of a previously underestimatedcomplex microbial community in CF. Chronic infectionsare typically associated with a few bacterial pathogenssuch as P. aeruginosa, S. aureus, and B. cepacia complex.However, the CF airways are colonized by polymicrobialcommunities composed of a number of additional organ-isms, many of which are considered members of thenormal oropharyngeal microbiota. The recent moleculartechnologies for the accurate identification or detection ofbacteria and for the evaluation of microbial diversity haveincreased the list of emerging pathogens associated withCF patients. These recent data suggest the concept that themicrobiome ecology in CF patients is more complex and

Semin Immunopathol (2011) 33:135–156 145

Page 12: Lung transplantation: infection, inflammation, and the microbiome

Fig. 3 Interactions between T helper 17 and T regulatory cells in themicrobiome of alveolar space. Alveolar epithelial cells have roles as astructural barrier and in the facilitation of mucociliary clearance inalveolar space contact with bacterial components and signals. Pro-inflammatory signals produced by pathobionts (organisms withpotential functions to induce pathology) activate innate immuneresponses and Th17 cells producing IL-17, a pro-inflammatory

cyotokine. Specific molecules produced by symbionts (organismswith known health-promoting functions) are recognized by antigen-presenting cells (APCs). This recognition activates naïve CD4+ T cellsand induces T regulatory cells (Tregs), producing IL-10, an anti-inflammatory cytokine. IL-10 suppresses the production of pro-inflammatory cytokines including IL-17

Fig. 4 Balance between T helper17 and T regulatory cells in themicrobiome. a A healthy microbiota contains a balanced compositionof symbionts, commensals, and pathobionts. Symbionts are organismswith known health-promoting functions. Commensals are thought toprovide no benefit or be a detriment to the host. Pathobionts have thepotential to induce pathology. The balance of Th17 and T regulatory

cells (Tregs) is also maintained in this situation. b Lung transplant caninduce changes in the balance between symbionts and pathobionts inthe allograft microbiota due to its infectious susceptibility, which maycause imbalance of Th17/Tregs and the development of lung allograftrejection

146 Semin Immunopathol (2011) 33:135–156

Page 13: Lung transplantation: infection, inflammation, and the microbiome

that CF may be a considered a polymicrobial infectiousdisease [281–284].

Interaction between Th17 and Tregs in the allograftmicrobiome

Interactions between the microbiome and immune re-sponse in lung transplant allografts consist of a delicatebalance due to the infectious susceptibility of allograftand the complexity of host defense response to theallograft. A model of the microbiota and interactionbetween Th17 and Tregs has been proposed [285].Signals produced by pathobionts, organisms with potentialfunctions to induce pathology, activate innate immunity aswell as Th17 cells. IL-17, a pro-inflammatory cytokine,from Th17 cells leads to injury or inflammation of tissue(Fig. 3). On the other hand, specific molecules producedby symbionts, organisms with known health-promotingfunctions, are taken up and presented to CD4+ T cells byAPCs. The recognition of the molecules by naïve CD4+ Tcells induces Tregs producing IL-10, an anti-inflammatorycytokine. IL-10 suppresses the production of IL-17 andprotects tissue damage. Whether inflammation occurs inthe lung may depend upon the responses of Th17 cells andTregs to organisms. Thus, the Th17/Tregs balance con-trolled by the microbiota is one of the most importantregulatory factors in disease. Healthy microbiota containsa balanced composition of symbionts, pathobionts, andcommensals, organisms thought to provide no benefit orbe a detriment to the host. Lung transplant can induce achange in the balance between symbionts and pathobiontsdue to multiple factors, including invasive surgery and useof antibiotics and immunosuppressant drugs. The decreaseof symbionts or increase of pathobionts may precipitateimbalance of Th17/Tregs and the development of allograftrejection (Fig. 4).

Conclusion

Despite improvements in surgical techniques and immuno-suppressive therapy, long-term outcomes after lung trans-plantation remain disappointing due to BOS. Thepathophysiology of BOS is multifactorial and complicated.Although insufficient immunosuppression results in allo-graft rejection, BOS is also strongly linked to airwayinfection caused by over-immunosuppression. Both hostimmune responses and pathogen attack need to becontrolled after lung transplant to prevent BOS. CD4+ Tcells have an important role in the immune responses to anallograft. Among CD4+ T cells, the interaction betweenTh17 cells and Tregs should be considered. Because Tregsattenuate inflammation, immunosuppressant drugs which

suppress activity of not only effector T cells but also Tregsmust be used with caution for BOS treatment or prevention.An additional caution for immunosuppression is the effecton the microbiome (the full collection of microbes thatnaturally exist in the lung). As mentioned previously,various pathogens including bacteria, fungi, and virus areinvolved in BOS. However, these pathogens constitute onlya small portion of whole microbes. New technologies allowcomprehensive examination of microbial communities andreveal previously unknown organisms. Recently, a relation-ship between the microbiome and diseases has been shown.The interaction between the changes in the microbiome andthe balance of Th17/Tregs may be closely linked to thepathogenesis of BOS. Further clarification of the micro-biome component and recognition of the changes in themicrobiome after lung transplantation are critical tounderstand the pathogenesis of allograft rejection.

Acknowledgments We thank Ko-Wei Lin PhD for a critical reviewof the manuscript. This work was supported by R01-HL077900 andR01-AI075317.

References

1. Christie JD, Edwards LB, Kucheryavaya AY, Aurora P, DobbelsF, Kirk R, Rahmel AO, Stehlik J, Hertz MI (2010) The Registryof the International Society for Heart and Lung Transplantation:twenty-seventh official adult lung and heart–lung transplantreport—2010. J Heart Lung Transplant 29(10):1104–1118

2. Verleden GM (2001) Chronic allograft rejection (obliterativebronchiolitis). Semin Respir Crit Care Med 22(5):551–558

3. Verleden GM, Dupont LJ, Van Raemdonck DE (2005) Is itbronchiolitis obliterans syndrome or is it chronic rejection: areappraisal? Eur Respir J 25(2):221–224

4. Vanaudenaerde BM, Meyts I, Vos R, Geudens N, De Wever W,Verbeken EK, Van Raemdonck DE, Dupont LJ, Verleden GM(2008) A dichotomy in bronchiolitis obliterans syndrome afterlung transplantation revealed by azithromycin therapy. EurRespir J 32(4):832–843. doi:10.1183/09031936.00134307

5. Estenne M, Hertz MI (2002) Bronchiolitis obliterans after humanlung transplantation. Am J Respir Crit Care Med 166(4):440–444

6. Estenne M, Maurer JR, Boehler A, Egan JJ, Frost A, Hertz M,Mallory GB, Snell GI, Yousem S (2002) Bronchiolitis obliteranssyndrome 2001: an update of the diagnostic criteria. J HeartLung Transplant 21(3):297–310

7. Knoop C, Estenne M (2006) Acute and chronic rejection afterlung transplantation. Semin Respir Crit Care Med 27(5):521–533

8. Belperio JA, Lake K, Tazelaar H, Keane MP, Strieter RM, LynchJP 3rd (2003) Bronchiolitis obliterans syndrome complicatinglung or heart–lung transplantation. Semin Respir Crit Care Med24(5):499–530. doi:10.1055/s-2004-815601

9. Stewart S, Fishbein MC, Snell GI, Berry GJ, Boehler A, BurkeMM, Glanville A, Gould FK, Magro C, Marboe CC, McNeilKD, Reed EF, Reinsmoen NL, Scott JP, Studer SM, TazelaarHD, Wallwork JL, Westall G, Zamora MR, Zeevi A, Yousem SA(2007) Revision of the 1996 working formulation for thestandardization of nomenclature in the diagnosis of lungrejection. J Heart Lung Transplant 26(12):1229–1242

Semin Immunopathol (2011) 33:135–156 147

Page 14: Lung transplantation: infection, inflammation, and the microbiome

10. Glanville AR, Aboyoun CL, Havryk A, Plit M, Rainer S, MaloufMA (2008) Severity of lymphocytic bronchiolitis predicts longterm outcome after lung transplantation. Am J Respir Crit CareMed 177:1033–1040

11. Husain AN, Siddiqui MT, Holmes EW, Chandrasekhar AJ,McCabe M, Radvany R, Garrity ER (1999) Analysis of riskfactors for the development of bronchiolitis obliterans syndrome.Am J Respir Crit Care Med 159(3):829–833

12. Hachem RR, Khalifah AP, Chakinala MM, Yusen RD, AloushAA, Mohanakumar T, Patterson GA, Trulock EP, Walter MJ(2005) The significance of a single episode of minimal acuterejection after lung transplantation. Transplantation 80(10):1406–1413

13. Hopkins PM, Aboyoun CL, Chhajed PN, Malouf MA, Plit ML,Rainer SP, Glanville AR (2004) Association of minimal rejectionin lung transplant recipients with obliterative bronchiolitis. Am JRespir Crit Care Med 170(9):1022–1026

14. Burton CM, Iversen M, Scheike T, Carlsen J, Andersen CB(2008) Is lymphocytic bronchiolitis a marker of acute rejection?An analysis of 2,697 transbronchial biopsies after lung trans-plantation. J Heart Lung Transplant 27(10):1128–1134.doi:10.1016/j.healun.2008.06.014

15. Weigt SS, Wallace WD, Derhovanessian A, Saggar R, Lynch JP,Belperio JA (2010) Chronic allograft rejection: epidemiology,diagnosis, pathogenesis, and treatment. Semin Respir Crit CareMed 31(2):189–207. doi:10.1055/s-0030-1249116

16. Akdis M (2010) The cellular orchestra in skin allergy; aredifferences to lung and nose relevant? Curr Opin Allergy ClinImmunol 10(5):443–451. doi:10.1097/ACI.0b013e32833d7d48

17. Gordon S (2003) Alternative activation of macrophages. Nat RevImmunol 3(1):23–35

18. Zingoni A, Sornasse T, Cocks BG, Tanaka Y, Santoni A, LanierLL (2005) NK cell regulation of T cell-mediated responses. MolImmunol 42(4):451–454

19. Mosmann TR, Sad S (1996) The expanding universe of T-cellsubsets: Th1, Th2 and more. Immunol Today 17(3):138–146

20. Mantovani A, Allavena P, Sica A (2004) Tumour-associatedmacrophages as a prototypic type II polarised phagocytepopulation: role in tumour progression. Eur J Cancer 40(11):1660–1667

21. Zhai Y, Kupiec-Weglinski JW (1999) What is the role ofregulatory T cells in transplantation tolerance? Curr OpinImmunol 11(5):497–503

22. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, CoffmanRL (1986) Two types of murine helper T cell clone I. Definitionaccording to profiles of lymphokine activities and secretedproteins. J Immunol 136(7):2348–2357

23. Zlotnik A, Yoshie O (2000) Chemokines: a new classificationsystem and their role in immunity. Immunity 12(2):121–127

24. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programsthe development and function of CD4+CD25+ regulatory T cells.Nat Immunol 4(4):330–336

25. Speich R, van der Bij W (2001) Epidemiology and managementof infections after lung transplantation. Clin Infect Dis 33(Suppl1):S58–S65. doi:10.1086/320906

26. Mulligan MS (2001) Endoscopic management of airway com-plications after lung transplantation. Chest Surg Clin N Am 11(4):907–915

27. Kshettry VR, Kroshus TJ, Hertz MI, Hunter DW, Shumway SJ,Bolman RM 3rd (1997) Early and late airway complications afterlung transplantation: incidence and management. Ann ThoracSurg 63(6):1576–1583

28. Yankaskas JR, Mallory GB Jr (1998) Lung transplantation incystic fibrosis: consensus conference statement. Chest 113(1):217–226

29. Flume PA, Egan TM, Paradowski LJ, Detterbeck FC, ThompsonJT, Yankaskas JR (1994) Infectious complications of lungtransplantation. Impact of cystic fibrosis. Am J Respir Crit CareMed 149(6):1601–1607

30. de Leval MR, Smyth R, Whitehead B, Scott JP, Elliott MJ,Sharples L, Caine N, Helms P, Martin IR, Higenbottam T (1991)Heart and lung transplantation for terminal cystic fibrosis. A 4 1/2-year experience. J Thorac Cardiovasc Surg 101(4):633–641,discussion 641–632

31. Snell GI, de Hoyos A, Krajden M, Winton T, Maurer JR (1993)Pseudomonas cepacia in lung transplant recipients with cysticfibrosis. Chest 103(2):466–471

32. Sun L, Jiang RZ, Steinbach S, Holmes A, Campanelli C,Forstner J, Sajjan U, Tan Y, Riley M, Goldstein R (1995) Theemergence of a highly transmissible lineage of cbl+ Pseudomo-nas (Burkholderia) cepacia causing CF centre epidemics inNorth America and Britain. Nat Med 1(7):661–666

33. Hadjiliadis D, Sporn TA, Perfect JR, Tapson VF, Davis RD,Palmer SM (2002) Outcome of lung transplantation in patientswith mycetomas. Chest 121(1):128–134

34. Olivier KN, Weber DJ, Lee JH, Handler A, Tudor G, Molina PL,Tomashefski J, Knowles MR (2003) Nontuberculous mycobac-teria. II: nested-cohort study of impact on cystic fibrosis lungdisease. Am J Respir Crit Care Med 167(6):835–840

35. Fishman JA, Rubin RH (1998) Infection in organ-transplantrecipients. N Engl J Med 338(24):1741–1751

36. Kuo CJ, Chung J, Fiorentino DF, Flanagan WM, Blenis J,Crabtree GR (1992) Rapamycin selectively inhibits interleukin-2activation of p70 S6 kinase. Nature 358(6381):70–73

37. Price DJ, Grove JR, Calvo V, Avruch J, Bierer BE (1992)Rapamycin-induced inhibition of the 70-kilodalton S6 proteinkinase. Science 257(5072):973–977

38. King-Biggs MB, Dunitz JM, Park SJ, Kay Savik S, Hertz MI(2003) Airway anastomotic dehiscence associated with use ofsirolimus immediately after lung transplantation. Transplantation75(9):1437–1443

39. Snell GI, Valentine VG, Vitulo P, Glanville AR, McGiffin DC,Loyd JE, Roman A, Aris R, Sole A, Hmissi A, Pirron U (2006)Everolimus versus azathioprine in maintenance lung transplantrecipients: an international, randomized, double-blind clinicaltrial. Am J Transplant 6(1):169–177

40. Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J,Kaupper T, Roncarolo MG (2006) Rapamycin promotes expan-sion of functional CD4+CD25+FOXP3+ regulatory T cells ofboth healthy subjects and type 1 diabetic patients. J Immunol177(12):8338–8347

41. Segundo DS, Ruiz JC, Izquierdo M, Fernandez-Fresnedo G,Gomez-Alamillo C, Merino R, Benito MJ, Cacho E, Rodrigo E,Palomar R, Lopez-Hoyos M, Arias M (2006) Calcineurininhibitors, but not rapamycin, reduce percentages ofCD4+CD25+FOXP3+ regulatory T cells in renal transplantrecipients. Transplantation 82(4):550–557

42. Aguilar-Guisado M, Givalda J, Ussetti P, Ramos A, Morales P,Blanes M, Bou G, de la Torre-Cisneros J, Roman A, Borro JM,Lama R, Cisneros JM (2007) Pneumonia after lung transplanta-tion in the RESITRA Cohort: a multicenter prospective study.Am J Transplant 7(8):1989–1996. doi:10.1111/j.1600-6143.2007.01882.x

43. Valentine VG, Bonvillain RW, Gupta MR, Lombard GA,LaPlace SG, Dhillon GS, Wang G (2008) Infections in lungallograft recipients: ganciclovir era. J Heart Lung Transplant 27(5):528–535

44. Sharples LD, McNeil K, Stewart S, Wallwork J (2002) Riskfactors for bronchiolitis obliterans: a systematic review of recentpublications. J Heart Lung Transplant 21(2):271–281

148 Semin Immunopathol (2011) 33:135–156

Page 15: Lung transplantation: infection, inflammation, and the microbiome

45. Mattner F, Fischer S, Weissbrodt H, Chaberny IF, Sohr D,Gottlieb J, Welte T, Henke-Gendo C, Gastmeier P, Strueber M(2007) Post-operative nosocomial infections after lung and hearttransplantation. J Heart Lung Transplant 26(3):241–249.doi:10.1016/j.healun.2006.12.009

46. Zeglen S, Wojarski J, Wozniak-Grygiel E, Siola M, JastrzebskiD, Kucewicz-Czech E, Zembala M (2009) Frequency ofPseudomonas aeruginosa colonizations/infections in lung trans-plant recipients. Transplant Proc 41(8):3222–3224. doi:10.1016/j.transproceed.2009.07.063

47. Bonvillain RW, Valentine VG, Lombard G, LaPlace S, DhillonG, Wang G (2007) Post-operative infections in cystic fibrosis andnon-cystic fibrosis patients after lung transplantation. J HeartLung Transplant 26(9):890–897. doi :10.1016/ j .healun.2007.07.002

48. Gottlieb J, Mattner F, Weissbrodt H, Dierich M, Fuehner T,Strueber M, Simon A, Welte T (2009) Impact of graftcolonization with Gram-negative bacteria after lung transplanta-tion on the development of bronchiolitis obliterans syndrome inrecipients with cystic fibrosis. Respir Med 103(5):743–749.doi:10.1016/j.rmed.2008.11.015

49. Botha P, Archer L, Anderson RL, Lordan J, Dark JH, Corris PA,Gould K, Fisher AJ (2008) Pseudomonas aeruginosa coloniza-tion of the allograft after lung transplantation and the risk ofbronchiolitis obliterans syndrome. Transplantation 85(5):771–774

50. Vos R, Vanaudenaerde BM, Geudens N, Dupont LJ, VanRaemdonck DE, Verleden GM (2008) Pseudomonal airwaycolonisation: risk factor for bronchiolitis obliterans syndromeafter lung transplantation? Eur Respir J 31(5):1037–1045

51. Chaparro C, Maurer J, Gutierrez C, Krajden M, Chan C, WintonT, Keshavjee S, Scavuzzo M, Tullis E, Hutcheon M, Kesten S(2001) Infection with Burkholderia cepacia in cystic fibrosis:outcome following lung transplantation. Am J Respir Crit CareMed 163(1):43–48

52. Meachery G, De Soyza A, Nicholson A, Parry G, Hasan A,Tocewicz K, Pillay T, Clark S, Lordan JL, Schueler S, Fisher AJ,Dark JH, Gould FK, Corris PA (2008) Outcomes of lungtransplantation for cystic fibrosis in a large UK cohort. Thorax63(8):725–731. doi:10.1136/thx.2007.092056

53. Boussaud V, Guillemain R, Grenet D, Coley N, Souilamas R,Bonnette P, Stern M (2008) Clinical outcome following lungtransplantation in patients with cystic fibrosis colonised withBurkholderia cepacia complex: results from two French centres.Thorax 63(8):732–737. doi:10.1136/thx.2007.089458

54. Alexander BD, Petzold EW, Reller LB, Palmer SM, Davis RD,Woods CW, Lipuma JJ (2008) Survival after lung transplantationof cystic fibrosis patients infected with Burkholderia cepaciacomplex. Am J Transplant 8(5):1025–1030. doi:10.1111/j.1600-6143.2008.02186.x

55. Murray S, Charbeneau J, Marshall BC, LiPuma JJ (2008) Impactof Burkholderia infection on lung transplantation in cysticfibrosis. Am J Respir Crit Care Med 178(4):363–371.doi:10.1164/rccm.200712-1834OC

56. Kesten S, Chaparro C (1999) Mycobacterial infections in lungtransplant recipients. Chest 115(3):741–745

57. Malouf MA, Glanville AR (1999) The spectrum of mycobacte-rial infection after lung transplantation. Am J Respir Crit CareMed 160(5 Pt 1):1611–1616

58. Kotloff RM, Ahya VN, Crawford SW (2004) Pulmonarycomplications of solid organ and hematopoietic stem celltransplantation. Am J Respir Crit Care Med 170(1):22–48

59. Segal BH, Walsh TJ (2006) Current approaches to diagnosis andtreatment of invasive aspergillosis. Am J Respir Crit Care Med173(7):707–717

60. Sole A, Morant P, Salavert M, Peman J, Morales P (2005)Aspergillus infections in lung transplant recipients: risk factorsand outcome. Clin Microbiol Infect 11(5):359–365

61. Marik PE (2006) Fungal infections in solid organ transplantation.Expert Opin Pharmacother 7(3):297–305

62. Singh N, Husain S (2003) Aspergillus infections after lungtransplantation: clinical differences in type of transplant andimplications for management. J Heart Lung Transplant 22(3):258–266

63. Alexander BD, Tapson VF (2001) Infectious complications oflung transplantation. Transpl Infect Dis 3(3):128–137

64. Weigt SS, Elashoff RM, Huang C, Ardehali A, Gregson AL,Kubak B, Fishbein MC, Saggar R, Keane MP, Lynch JP 3rd,Zisman DA, Ross DJ, Belperio JA (2009) Aspergillus coloniza-tion of the lung allograft is a risk factor for bronchiolitisobliterans syndrome. Am J Transplant 9(8):1903–1911.doi:10.1111/j.1600-6143.2009.02635.x

65. Cahill BC, Hibbs JR, Savik K, Juni BA, Dosland BM, Edin-Stibbe C, Hertz MI (1997) Aspergillus airway colonization andinvasive disease after lung transplantation. Chest 112(5):1160–1164

66. Monforte V, Roman A, Gavalda J, Bravo C, Tenorio L, Ferrer A,Maestre J, Morell F (2001) Nebulized amphotericin B prophy-laxis for Aspergillus infection in lung transplantation: study ofrisk factors. J Heart Lung Transplant 20(12):1274–1281

67. Paradowski LJ (1997) Saprophytic fungal infections and lungtransplantation—revisited. J Heart Lung Transplant 16(5):524–531

68. Khalifah AP, Hachem RR, Chakinala MM, Schechtman KB,Patterson GA, Schuster DP, Mohanakumar T, Trulock EP, WalterMJ (2004) Respiratory viral infections are a distinct risk forbronchiolitis obliterans syndrome and death. Am J Respir CritCare Med 170(2):181–187

69. Bharat A, Kuo E, Saini D, Steward N, Hachem R, Trulock EP,Patterson GA, Meyers BF, Mohanakumar T (2010) Respiratoryvirus-induced dysregulation of T-regulatory cells leads to chronicrejection. Ann Thorac Surg 90(5):1637–1644, discussion 1644

70. Zamora MR, Davis RD, Leonard C (2005) Management ofcytomegalovirus infection in lung transplant recipients:evidence-based recommendations. Transplantation 80(2):157–163

71. Weigt SS, Elashoff RM, Keane MP, Strieter RM, Gomperts BN,Xue YY, Ardehali A, Gregson AL, Kubak B, Fishbein MC,Saggar R, Ross DJ, Lynch JP 3rd, Zisman DA, Belperio JA(2008) Altered levels of CC chemokines during pulmonaryCMV predict BOS and mortality post-lung transplantation. Am JTransplant 8:1512–1522

72. Bridges ND, Spray TL, Collins MH, Bowles NE, Towbin JA(1998) Adenovirus infection in the lung results in graft failureafter lung transplantation. J Thorac Cardiovasc Surg 116(4):617–623

73. Palmer SM Jr, Henshaw NG, Howell DN, Miller SE, Davis RD,Tapson VF (1998) Community respiratory viral infection in adultlung transplant recipients. Chest 113(4):944–950

74. Simsir A, Greenebaum E, Nuovo G, Schulman LL (1998) Latefatal adenovirus pneumonitis in a lung transplant recipient.Transplantation 65(4):592–594

75. Aris RM, Maia DM, Neuringer IP, Gott K, Kiley S, Gertis K,Handy J (1996) Post-transplantation lymphoproliferative disor-der in the Epstein–Barr virus-naive lung transplant recipient. AmJ Respir Crit Care Med 154(6 Pt 1):1712–1717

76. Montone KT, Litzky LA,Wurster A, Kaiser L, Bavaria J, Kotloff R,Palevsky H, Pietra GG, Tomaszewski JE (1996) Analysis ofEpstein–Barr virus-associated posttransplantation lymphoprolifer-ative disorder after lung transplantation. Surgery 119(5):544–551

Semin Immunopathol (2011) 33:135–156 149

Page 16: Lung transplantation: infection, inflammation, and the microbiome

77. Sherman LA, Chattopadhyay S (1993) The molecular basis ofallorecognition. Annu Rev Immunol 11:385–402

78. Sayegh MH, Turka LA (1998) The role of T-cell costimulatoryactivation pathways in transplant rejection. N Engl J Med 338(25):1813–1821

79. Ciubotariu R, Liu Z, Colovai AI, Ho E, Itescu S, Ravalli S,Hardy MA, Cortesini R, Rose EA, Suciu-Foca N (1998)Persistent allopeptide reactivity and epitope spreading in chronicrejection of organ allografts. J Clin Invest 101(2):398–405

80. Vella JP, Spadafora-Ferreira M, Murphy B, Alexander SI,Harmon W, Carpenter CB, Sayegh MH (1997) Indirect allor-ecognition of major histocompatibility complex allopeptides inhuman renal transplant recipients with chronic graft dysfunction.Transplantation 64(6):795–800

81. Dinarello CA (1996) Biologic basis for interleukin-1 in disease.Blood 87(6):2095–2147

82. Arend WP, Welgus HG, Thompson RC, Eisenberg SP (1990)Biological properties of recombinant human monocyte-derivedinterleukin 1 receptor antagonist. J Clin Invest 85(5):1694–1697

83. Beutler BA (1999) The role of tumor necrosis factor in healthand disease. J Rheumatol 26(Suppl 57):16–21

84. Ostensen ME, Thiele DL, Lipsky PE (1987) Tumor necrosisfactor-alpha enhances cytolytic activity of human natural killercells. J Immunol 138(12):4185–4191

85. Piguet PF, Collart MA, Grau GE, Kapanci Y, Vassalli P (1989)Tumor necrosis factor/cachectin plays a key role in bleomycin-induced pneumopathy and fibrosis. J Exp Med 170(3):655–663

86. He Y, Liu W (1996) The preliminary research on the relationshipbetween TNF- and egg-induced granuloma and hepatic fibrosisof schistosomiasis japonica. J Tongji Med Univ 16(4):205–208

87. Krishnadasan B, Naidu BV, Byrne K, Fraga C, Verrier ED,Mulligan MS (2003) The role of proinflammatory cytokines inlung ischemia–reperfusion injury. J Thorac Cardiovasc Surg 125(2):261–272

88. DeMeester SR, Rolfe MW, Kunkel SL, Swiderski DL, LincolnPM, Deeb GM, Strieter RM (1993) The bimodal expression oftumor necrosis factor-alpha in association with rat lung reim-plantation and allograft rejection. J Immunol 150(6):2494–2505

89. Aris RM, Walsh S, Chalermskulrat W, Hathwar V, Neuringer IP(2002) Growth factor upregulation during obliterative bronchio-litis in the mouse model. Am J Respir Crit Care Med 166(3):417–422

90. Smith CR, Jaramillo A, Lu KC, Higuchi T, Kaleem Z,Mohanakumar T (2001) Prevention of obliterative airwaydisease in HLA-A2-transgenic tracheal allografts by neutral-ization of tumor necrosis factor. Transplantation 72(9):1512–1518

91. Rizzo M, SivaSai KS, Smith MA, Trulock EP, Lynch JP,Patterson GA, Mohanakumar T (2000) Increased expression ofinflammatory cytokines and adhesion molecules by alveolarmacrophages of human lung allograft recipients with acuterejection: decline with resolution of rejection. J Heart LungTransplant 19(9):858–865

92. Gately MK, Renzetti LM, Magram J, Stern AS, Adorini L,Gubler U, Presky DH (1998) The interleukin-12/interleukin-12-receptor system: role in normal and pathologic immuneresponses. Annu Rev Immunol 16:495–521

93. Trinchieri G (1995) Interleukin-12: a proinflammatory cytokinewith immunoregulatory functions that bridge innate resistanceand antigen-specific adaptive immunity. Annu Rev Immunol13:251–276

94. Seder RA, Gazzinelli R, Sher A, Paul WE (1993) Interleukin12 acts directly on CD4+ T cells to enhance priming forinterferon gamma production and diminishes interleukin 4inhibition of such priming. Proc Natl Acad Sci USA 90(21):10188–10192

95. McKnight AJ, Zimmer GJ, Fogelman I, Wolf SF, Abbas AK(1994) Effects of IL-12 on helper T cell-dependent immuneresponses in vivo. J Immunol 152(5):2172–2179

96. Marshall JD, Secrist H, DeKruyff RH, Wolf SF, Umetsu DT(1995) IL-12 inhibits the production of IL-4 and IL-10 inallergen-specific human CD4+ T lymphocytes. J Immunol 155(1):111–117

97. Via CS, Rus V, Gately MK, Finkelman FD (1994) IL-12stimulates the development of acute graft-versus-host disease inmice that normally would develop chronic, autoimmune graft-versus-host disease. J Immunol 153(9):4040–4047

98. Hsieh CS, Macatonia SE, Tripp CS, Wolf SF, O'Garra A,Murphy KM (1993) Development of TH1 CD4+ T cells throughIL-12 produced by Listeria-induced macrophages. Science 260(5107):547–549

99. Oswald IP, Caspar P, Jankovic D, Wynn TA, Pearce EJ, Sher A(1994) IL-12 inhibits Th2 cytokine responses induced by eggs ofSchistosoma mansoni. J Immunol 153(4):1707–1713

100. Pearlman E, Heinzel FP, Hazlett FE Jr, Kazura JW (1995) IL-12modulation of T helper responses to the filarial helminth, Brugiamalayi. J Immunol 154(9):4658–4664

101. Gazzinelli RT, Giese NA, Morse HC 3rd (1994) In vivotreatment with interleukin 12 protects mice from immuneabnormalities observed during murine acquired immunodeficien-cy syndrome (MAIDS). J Exp Med 180(6):2199–2208

102. Zhou P, Sieve MC, Bennett J, Kwon-Chung KJ, Tewari RP,Gazzinelli RT, Sher A, Seder RA (1995) IL-12 preventsmortality in mice infected with Histoplasma capsulatum throughinduction of IFN-gamma. J Immunol 155(2):785–795

103. Rosenberg AS, Singer A (1992) Cellular basis of skin allograftrejection: an in vivo model of immune-mediated tissue destruc-tion. Annu Rev Immunol 10:333–358

104. Piccotti JR, Li K, Chan SY, Eichwald EJ, Bishop DK (1999)Interleukin-12 (IL-12)-driven alloimmune responses in vitro andin vivo: requirement for beta1 subunit of the IL-12 receptor.Transplantation 67(11):1453–1460

105. Williamson E, Garside P, Bradley JA, More IA, Mowat AM(1997) Neutralizing IL-12 during induction of murine acutegraft-versus-host disease polarizes the cytokine profile toward aTh2-type alloimmune response and confers long term protectionfrom disease. J Immunol 159(3):1208–1215

106. Orr DJ, Bolton EM, Bradley JA (1998) Neutralising IL-12activity as a strategy for prolonging allograft survival andpreventing graft-versus-host disease. Scott Med J 43(4):109–111

107. Welniak LA, Blazar BR, Wiltrout RH, Anver MR, Murphy WJ(2001) Role of interleukin-12 in acute graft-versus-host disease(1). Transplant Proc 33(1–2):1752–1753

108. Williamson E, Garside P, Bradley JA, Mowat AM (1996) IL-12is a central mediator of acute graft-versus-host disease in mice. JImmunol 157(2):689–699

109. Piccotti JR, Chan SY, Goodman RE, Magram J, Eichwald EJ,Bishop DK (1996) IL-12 antagonism induces T helper 2responses, yet exacerbates cardiac allograft rejection. Evidenceagainst a dominant protective role for T helper 2 cytokines inalloimmunity. J Immunol 157(5):1951–1957

110. Konieczny BT, Dai Z, Elwood ET, Saleem S, Linsley PS,Baddoura FK, Larsen CP, Pearson TC, Lakkis FG (1998) IFN-gamma is critical for long-term allograft survival induced byblocking the CD28 and CD40 ligand T cell costimulationpathways. J Immunol 160(5):2059–2064

111. Tau GZ, von der Weid T, Lu B, Cowan S, Kvatyuk M, Pernis A,Cattoretti G, Braunstein NS, Coffman RL, Rothman PB (2000)Interferon gamma signaling alters the function of T helper type 1cells. J Exp Med 192(7):977–986

112. Mikols CL, Yan L, Norris JY, Russell TD, Khalifah AP, HachemRR, Chakinala MM, Yusen RD, Castro M, Kuo E, Patterson GA,

150 Semin Immunopathol (2011) 33:135–156

Page 17: Lung transplantation: infection, inflammation, and the microbiome

Mohanakumar T, Trulock EP, Walter MJ (2006) IL-12 p80 is aninnate epithelial cell effector that mediates chronic allograftdysfunction. Am J Respir Crit Care Med 174(4):461–470

113. Meloni F, Vitulo P, Cascina A, Oggionni T, Bulgheroni A,Paschetto E, Klersy C, D'Armini AM, Fietta A, Bianco AM,Arbustini E, Vigano M (2004) Bronchoalveolar lavage cytokineprofile in a cohort of lung transplant recipients: a predictive roleof interleukin-12 with respect to onset of bronchiolitis obliteranssyndrome. J Heart Lung Transplant 23(9):1053–1060

114. Farrar MA, Schreiber RD (1993) The molecular cell biology ofinterferon-gamma and its receptor. Annu Rev Immunol 11:571–611

115. Thierfelder WE, van Deursen JM, Yamamoto K, Tripp RA,Sarawar SR, Carson RT, Sangster MY, Vignali DA, Doherty PC,Grosveld GC, Ihle JN (1996) Requirement for Stat4 ininterleukin-12-mediated responses of natural killer and T cells.Nature 382(6587):171–174

116. Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, OG A(1991) IL-10 inhibits cytokine production by activated macro-phages. J Immunol 147(11):3815–3822

117. Boehler A, Bai XH, Liu M, Cassivi S, Chamberlain D, SlutskyAS, Keshavjee S (1999) Upregulation of T-helper 1 cytokinesand chemokine expression in post-transplant airway obliteration.Am J Respir Crit Care Med 159(6):1910–1917

118. Nagano H, Mitchell RN, Taylor MK, Hasegawa S, Tilney NL,Libby P (1997) Interferon-gamma deficiency prevents coronaryarteriosclerosis but not myocardial rejection in transplantedmouse hearts. J Clin Invest 100(3):550–557

119. Raisanen-Sokolowski A, Mottram PL, Glysing-Jensen T, Satos-kar A, Russell ME (1997) Heart transplants in interferon-gamma,interleukin 4, and interleukin 10 knockout mice. Recipientenvironment alters graft rejection. J Clin Invest 100(10):2449–2456

120. Raisanen-Sokolowski A, Glysing-Jensen T, Russell ME (1998)Leukocyte-suppressing influences of interleukin (IL)-10 incardiac allografts: insights from IL-10 knockout mice. Am JPathol 153(5):1491–1500

121. Moudgil A, Bagga A, Toyoda M, Nicolaidou E, Jordan SC, RossD (1999) Expression of gamma-IFN mRNA in bronchoalveolarlavage fluid correlates with early acute allograft rejection in lungtransplant recipients. Clin Transplant 13(2):201–207

122. Iacono A, Dauber J, Keenan R, Spichty K, Cai J, Grgurich W,Burckart G, Smaldone G, Pham S, Ohori NP, Yousem S,Williams P, Griffith B, Zeevi A (1997) Interleukin 6 andinterferon-gamma gene expression in lung transplant recipientswith refractory acute cellular rejection: implications for monitor-ing and inhibition by treatment with aerosolized cyclosporine.Transplantation 64(2):263–269

123. Lu KC, Jaramillo A, Lecha RL, Schuessler RB, Aloush A,Trulock EP, Mendeloff EN, Huddleston CB, Alexander PattersonG, Mohanakumar T (2002) Interleukin-6 and interferon-gammagene polymorphisms in the development of bronchiolitisobliterans syndrome after lung transplantation. Transplantation74(9):1297–1302

124. Grabstein KH, Eisenman J, Shanebeck K, Rauch C, SrinivasanS, Fung V, Beers C, Richardson J, Schoenborn MA, Ahdieh M etal (1994) Cloning of a T cell growth factor that interacts with thebeta chain of the interleukin-2 receptor. Science 264(5161):965–968

125. Lodolce JP, Boone DL, Chai S, Swain RE, Dassopoulos T,Trettin S, Ma A (1998) IL-15 receptor maintains lymphoidhomeostasis by supporting lymphocyte homing and proliferation.Immunity 9(5):669–676

126. Kennedy MK, Glaccum M, Brown SN, Butz EA, Viney JL,Embers M, Matsuki N, Charrier K, Sedger L, Willis CR, BraselK, Morrissey PJ, Stocking K, Schuh JC, Joyce S, Peschon JJ

(2000) Reversible defects in natural killer and memory CD8 Tcell lineages in interleukin 15-deficient mice. J Exp Med 191(5):771–780

127. Doherty TM, Seder RA, Sher A (1996) Induction and regulationof IL-15 expression in murine macrophages. J Immunol 156(2):735–741

128. Alleva DG, Kaser SB, Monroy MA, Fenton MJ, Beller DI(1997) IL-15 functions as a potent autocrine regulator ofmacrophage proinflammatory cytokine production: evidence fordifferential receptor subunit utilization associated with stimula-tion or inhibition. J Immunol 159(6):2941–2951

129. Fehniger TA, Suzuki K, Ponnappan A, VanDeusen JB, CooperMA, Florea SM, Freud AG, Robinson ML, Durbin J, CaligiuriMA (2001) Fatal leukemia in interleukin 15 transgenic micefollows early expansions in natural killer and memory phenotypeCD8+ T cells. J Exp Med 193(2):219–231

130. Carson WE, Giri JG, Lindemann MJ, Linett ML, Ahdieh M,Paxton R, Anderson D, Eisenmann J, Grabstein K, Caligiuri MA(1994) Interleukin (IL) 15 is a novel cytokine that activateshuman natural killer cells via components of the IL-2 receptor. JExp Med 180(4):1395–1403

131. Kanegane H, Tosato G (1996) Activation of naive and memory Tcells by interleukin-15. Blood 88(1):230–235

132. Bulfone-Paus S, Ungureanu D, Pohl T, Lindner G, Paus R,Ruckert R, Krause H, Kunzendorf U (1997) Interleukin-15protects from lethal apoptosis in vivo. Nat Med 3(10):1124–1128

133. Pavlakis M, Strehlau J, Lipman M, Shapiro M, Maslinski W,Strom TB (1996) Intragraft IL-15 transcripts are increased inhuman renal allograft rejection. Transplantation 62(4):543–545

134. Strehlau J, Pavlakis M, Lipman M, Shapiro M, Vasconcellos L,Harmon W, Strom TB (1997) Quantitative detection of immuneactivation transcripts as a diagnostic tool in kidney transplanta-tion. Proc Natl Acad Sci USA 94(2):695–700

135. van Gelder T, Baan CC, Balk AH, Knoop CJ, Holweg CT, vander Meer P, Mochtar B, Zondervan PE, Niesters HG, Weimar W(1998) Blockade of the interleukin (IL)-2/IL-2 receptor pathwaywith a monoclonal anti-IL-2 receptor antibody (BT563) does notprevent the development of acute heart allograft rejection inhumans. Transplantation 65(3):405–410

136. Baan CC, Knoop CJ, van Gelder T, Holweg CT, Niesters HG,Smeets TJ, van der Ham F, Zondervan PE, Maat LP, Balk AH,Weimar W (1999) Anti-CD25 therapy reveals the redundancy ofthe intragraft cytokine network after clinical heart transplanta-tion. Transplantation 67(6):870–876

137. de Waal MR, Abrams J, Bennett B, Figdor CG, de Vries JE(1991) Interleukin 10(IL-10) inhibits cytokine synthesis byhuman monocytes: an autoregulatory role of IL-10 produced bymonocytes. J Exp Med 174(5):1209–1220

138. de Waal MR, Haanen J, Spits H, Roncarolo MG, te Velde A,Figdor C, Johnson K, Kastelein R, Yssel H, de Vries JE (1991)Interleukin 10 (IL-10) and viral IL-10 strongly reduce antigen-specific human T cell proliferation by diminishing the antigen-presenting capacity of monocytes via downregulation of class IImajor histocompatibility complex expression. J Exp Med 174(4):915–924

139. de Waal MR, Yssel H, Roncarolo MG, Spits H, de Vries JE(1992) Interleukin-10. Curr Opin Immunol 4(3):314–320

140. de Waal Malefyt R, Yssel H, de Vries JE (1993) Direct effects ofIL-10 on subsets of human CD4+ T cell clones and resting Tcells. Specific inhibition of IL-2 production and proliferation. JImmunol 150(11):4754–4765

141. te Velde AA, de Waal Malefijt R, Huijbens RJ, de Vries JE,Figdor CG (1992) IL-10 stimulates monocyte Fc gamma Rsurface expression and cytotoxic activity. Distinct regulation ofantibody-dependent cellular cytotoxicity by IFN-gamma, IL-4,and IL-10. J Immunol 149(12):4048–4052

Semin Immunopathol (2011) 33:135–156 151

Page 18: Lung transplantation: infection, inflammation, and the microbiome

142. Fiorentino DF, Zlotnik A, Vieira P, Mosmann TR, Howard M,Moore KW, OG A (1991) IL-10 acts on the antigen-presentingcell to inhibit cytokine production by Th1 cells. J Immunol 146(10):3444–3451

143. Bogdan C, Vodovotz Y, Nathan C (1991) Macrophage deactiva-tion by interleukin 10. J Exp Med 174(6):1549–1555

144. Ralph P, Nakoinz I, Sampson-Johannes A, Fong S, Lowe D, MinHY, Lin L (1992) IL-10, T lymphocyte inhibitor of human bloodcell production of IL-1 and tumor necrosis factor. J Immunol 148(3):808–814

145. Hsu DH, de Waal MR, Fiorentino DF, Dang MN, Vieira P, deVries J, Spits H, Mosmann TR, Moore KW (1990) Expression ofinterleukin-10 activity by Epstein–Barr virus protein BCRF1.Science 250(4982):830–832

146. Hsu DH, Moore KW, Spits H (1992) Differential effects of IL-4and IL-10 on IL-2-induced IFN-gamma synthesis andlymphokine-activated killer activity. Int Immunol 4(5):563–569

147. Chen WF, Zlotnik A (1991) IL-10: a novel cytotoxic T celldifferentiation factor. J Immunol 147(2):528–534

148. MacNeil IA, Suda T, Moore KW, Mosmann TR, Zlotnik A(1990) IL-10, a novel growth cofactor for mature and immatureT cells. J Immunol 145(12):4167–4173

149. Hu S, Chao CC, Ehrlich LC, Sheng WS, Sutton RL, RockswoldGL, Peterson PK (1999) Inhibition of microglial cell RANTESproduction by IL-10 and TGF-beta. J Leukoc Biol 65(6):815–821

150. Bejarano MT, de Waal MR, Abrams JS, Bigler M, Bacchetta R,de Vries JE, Roncarolo MG (1992) Interleukin 10 inhibitsallogeneic proliferative and cytotoxic T cell responses generatedin primary mixed lymphocyte cultures. Int Immunol 4(12):1389–1397

151. Bromberg JS (1995) IL-10 immunosuppression in transplanta-tion. Curr Opin Immunol 7(5):639–643

152. Zuo XJ, Matsumura Y, Prehn J, Saito R, Marchevesky A,Matloff J, Jordan SC (1995) Cytokine gene expression inrejecting and tolerant rat lung allograft models: analysis by RT-PCR. Transpl Immunol 3(2):151–161

153. Maeda H, Takata M, Takahashi S, Ogoshi S, Fujimoto S (1994)Adoptive transfer of a Th2-like cell line prolongs MHC class IIantigen disparate skin allograft survival in the mouse. IntImmunol 6(6):855–862

154. Zheng XX, Steele AW, Nickerson PW, Steurer W, Steiger J,Strom TB (1995) Administration of noncytolytic IL-10/Fc inmurine models of lipopolysaccharide-induced septic shockand allogeneic islet transplantation. J Immunol 154(10):5590–5600

155. Lee MS, Wogensen L, Shizuru J, Oldstone MB, Sarvetnick N(1994) Pancreatic islet production of murine interleukin-10 doesnot inhibit immune-mediated tissue destruction. J Clin Invest 93(3):1332–1338

156. Qian S, Li W, Li Y, Fu F, Lu L, Fung JJ, Thomson AW (1996)Systemic administration of cellular interleukin-10 can exacerbatecardiac allograft rejection in mice. Transplantation 62(12):1709–1714

157. Mulligan MS, Warner RL, McDuffie JE, Bolling SF, Sarma JV,Ward PA (2000) Regulatory role of Th-2 cytokines, IL-10 andIL-4, in cardiac allograft rejection. Exp Mol Pathol 69(1):1–9

158. Oshima K, Sen L, Cui G, Tung T, Sacks BM, Arellano-Kruse A,Laks H (2002) Localized interleukin-10 gene transfer inducesapoptosis of alloreactive T cells via FAS/FASL pathway,improves function, and prolongs survival of cardiac allograft.Transplantation 73(7):1019–1026

159. Qin L, Chavin KD, Ding Y, Tahara H, Favaro JP, Woodward JE,Suzuki T, Robbins PD, Lotze MT, Bromberg JS (1996)Retrovirus-mediated transfer of viral IL-10 gene prolongs murinecardiac allograft survival. J Immunol 156(6):2316–2323

160. Qin L, Ding Y, Pahud DR, Robson ND, Shaked A, Bromberg JS(1997) Adenovirus-mediated gene transfer of viral interleukin-10inhibits the immune response to both alloantigen and adenoviralantigen. Hum Gene Ther 8(11):1365–1374

161. de Perrot M, Fischer S, Liu M, Imai Y, Martins S, Sakiyama S,Tabata T, Bai XH, Waddell TK, Davidson BL, Keshavjee S(2003) Impact of human interleukin-10 on vector-inducedinflammation and early graft function in rat lung transplantation.Am J Respir Cell Mol Biol 28(5):616–625

162. Kozower BD, Kanaan SA, Tagawa T, Suda T, Grapperhaus K,Daddi N, Crouch EC, Doerschuk CM, Patterson GA (2002)Intramuscular gene transfer of interleukin-10 reduces neutrophilrecruitment and ameliorates lung graft ischemia–reperfusioninjury. Am J Transplant 2(9):837–842

163. Martins S, de Perrot M, Imai Y, Yamane M, Quadri SM, SegallL, Dutly A, Sakiyama S, Chaparro A, Davidson BL, WaddellTK, Liu M, Keshavjee S (2004) Transbronchial administration ofadenoviral-mediated interleukin-10 gene to the donor improvesfunction in a pig lung transplant model. Gene Ther 11(24):1786–1796

164. Pierog J, Gazdhar A, Stammberger U, Gugger M, Hyde S,Mathiesen I, Grodzki T, Schmid RA (2005) Synergistic effect oflow dose cyclosporine A and human interleukin 10 over-expression on acute rejection in rat lung allotransplantation.Eur J Cardiothorac Surg 27(6):1030–1035

165. Naidu B, Krishnadasan B, Whyte RI, Warner RL, Ward PA,Mulligan MS (2002) Regulatory role of IL-10 in experimentalobliterative bronchiolitis in rats. Exp Mol Pathol 73(3):164–170

166. Yonemitsu Y, Kitson C, Ferrari S, Farley R, Griesenbach U,Judd D, Steel R, Scheid P, Zhu J, Jeffery PK, Kato A, HasanMK, Nagai Y, Masaki I, Fukumura M, Hasegawa M, GeddesDM, Alton EW (2000) Efficient gene transfer to airwayepithelium using recombinant Sendai virus. Nat Biotechnol 18(9):970–973

167. Shoji F, Yonemitsu Y, Okano S, Yoshino I, Nakagawa K,Nakashima Y, Hasegawa M, Sugimachi K, Sueishi K (2003)Airway-directed gene transfer of interleukin-10 using recombi-nant Sendai virus effectively prevents post-transplant fibrousairway obliteration in mice. Gene Ther 10(3):213–218

168. Salgar SK, Yang D, Ruiz P, Miller J, Tzakis AG (2004) Viralinterleukin-10-engineered autologous hematopoietic stem celltherapy: a novel gene therapy approach to prevent graft rejection.Hum Gene Ther 15(2):131–144

169. Zheng HX, Burckart GJ, McCurry K, Webber S, Ristich J,Iacono A, Dauber J, McDade K, Grgurich W, Zaldonis D, PillageG, Griffith BP, Zeevi A (2004) Interleukin-10 productiongenotype protects against acute persistent rejection after lungtransplantation. J Heart Lung Transplant 23(5):541–546

170. Keane MP, Gomperts BN, Weigt S, Xue YY, Burdick MD,Nakamura H, Zisman DA, Ardehali A, Saggar R, Lynch JP 3rd,Hogaboam C, Kunkel SL, Lukacs NW, Ross DJ, Grusby MJ,Strieter RM, Belperio JA (2007) IL-13 is pivotal in the fibro-obliterative process of bronchiolitis obliterans syndrome. JImmunol 178(1):511–519

171. Lama VN, Harada H, Badri LN, Flint A, Hogaboam CM,McKenzie A, Martinez FJ, Toews GB, Moore BB, Pinsky DJ(2006) Obligatory role for interleukin-13 in obstructive lesiondevelopment in airway allografts. Am J Pathol 169(1):47–60

172. Kishimoto T (1992) Interleukin-6 and its receptor in autoimmu-nity. J Autoimmun 5(Suppl A):123–132

173. Hirano T, Matsuda T, Turner M, Miyasaka N, Buchan G, TangB, Sato K, Shimizu M, Maini R, Feldmann M et al (1988)Excessive production of interleukin 6/B cell stimulatory factor-2in rheumatoid arthritis. Eur J Immunol 18(11):1797–1801

174. Taga T, Kishimoto T (1997) Gp130 and the interleukin-6 familyof cytokines. Annu Rev Immunol 15:797–819

152 Semin Immunopathol (2011) 33:135–156

Page 19: Lung transplantation: infection, inflammation, and the microbiome

175. Shahar I, Fireman E, Topilsky M, Grief J, Kivity S, Spirer Z,Ben Efraim S (1996) Effect of IL-6 on alveolar fibroblastproliferation in interstitial lung diseases. Clin Immunol Immu-nopathol 79(3):244–251

176. Horii Y, Muraguchi A, Iwano M, Matsuda T, Hirayama T,Yamada H, Fujii Y, Dohi K, Ishikawa H, Ohmoto Y et al (1989)Involvement of IL-6 in mesangial proliferative glomerulonephri-tis. J Immunol 143(12):3949–3955

177. Rolfe MW, Kunkel S, Lincoln P, Deeb M, Lupinetti F, Strieter R(1993) Lung allograft rejection: role of tumor necrosis factor-alpha and interleukin-6. Chest 103(2 Suppl):133S

178. Farivar AS, Merry HE, Fica-Delgado MJ, McCourtie AS,Mackinnon-Patterson BC, Mulligan MS (2006) Interleukin-6regulation of direct lung ischemia reperfusion injury. Ann ThoracSurg 82(2):472–478

179. Scholma J, Slebos DJ, Boezen HM, van den Berg JW, van derBij W, de Boer WJ, Koeter GH, Timens W, Kauffman HF,Postma DS (2000) Eosinophilic granulocytes and interleukin-6level in bronchoalveolar lavage fluid are associated with thedevelopment of obliterative bronchiolitis after lung transplanta-tion. Am J Respir Crit Care Med 162(6):2221–2225

180. Penttinen RP, Kobayashi S, Bornstein P (1988) Transforminggrowth factor beta increases mRNA for matrix proteins both inthe presence and in the absence of changes in mRNA stability.Proc Natl Acad Sci USA 85(4):1105–1108

181. Roberts AB, Sporn MB, Assoian RK, Smith JM, Roche NS,Wakefield LM, Heine UI, Liotta LA, Falanga V, Kehrl JH et al(1986) Transforming growth factor type beta: rapid induction offibrosis and angiogenesis in vivo and stimulation of collagenformation in vitro. Proc Natl Acad Sci USA 83(12):4167–4171

182. Mora BN, Boasquevisque CH, Boglione M, Ritter JM, ScheuleRK, Yew NS, Debruyne L, Qin L, Bromberg JS, Patterson GA(2000) Transforming growth factor-beta1 gene transfer amelio-rates acute lung allograft rejection. J Thorac Cardiovasc Surg 119(5):913–920

183. Qin L, Chavin KD, Ding Y, Woodward JE, Favaro JP, Lin J,Bromberg JS (1994) Gene transfer for transplantation. Prolon-gation of allograft survival with transforming growth factor-beta1. Ann Surg 220(4):508–518, discussion 518–509

184. Yasufuku K, Heidler KM, O'Donnell PW, Smith GN Jr,Cummings OW, Foresman BH, Fujisawa T, Wilkes DS (2001)Oral tolerance induction by type V collagen downregulates lungallograft rejection. Am J Respir Cell Mol Biol 25(1):26–34

185. Liu M, Suga M, Maclean AA, St George JA, Souza DW,Keshavjee S (2002) Soluble transforming growth factor-betatype III receptor gene transfection inhibits fibrous airwayobliteration in a rat model of bronchiolitis obliterans. Am JRespir Crit Care Med 165(3):419–423

186. Massague J, Wotton D (2000) Transcriptional control by theTGF-beta/Smad signaling system. EMBO J 19(8):1745–1754

187. Elssner A, Jaumann F, Dobmann S, Behr J, Schwaiblmair M,Reichenspurner H, Furst H, Briegel J, Vogelmeier C (2000)Elevated levels of interleukin-8 and transforming growth factor-beta in bronchoalveolar lavage fluid from patients with bron-chiolitis obliterans syndrome: proinflammatory role of bronchialepithelial cells. Munich Lung Transplant Group. Transplantation70(2):362–367

188. El-Gamel A, Sim E, Hasleton P, Hutchinson J, Yonan N, Egan J,Campbell C, Rahman A, Sheldon S, Deiraniya A, Hutchinson IV(1999) Transforming growth factor beta (TGF-beta) and obliter-ative bronchiolitis following pulmonary transplantation. J HeartLung Transplant 18(9):828–837

189. Charpin JM, Valcke J, Kettaneh L, Epardeau B, Stern M, Israel-Biet D (1998) Peaks of transforming growth factor-beta mRNAin alveolar cells of lung transplant recipients as an early markerof chronic rejection. Transplantation 65(5):752–755

190. Shin HC, Benbernou N, Esnault S, Guenounou M (1999)Expression of IL-17 in human memory CD45RO+ T lympho-cytes and its regulation by protein kinase A pathway. Cytokine11(4):257–266

191. Rouvier E, Luciani MF, Mattei MG, Denizot F, Golstein P(1993) CTLA-8, cloned from an activated T cell, bearing AU-rich messenger RNA instability sequences, and homologous to aherpesvirus saimiri gene. J Immunol 150(12):5445–5456

192. Infante-Duarte C, Horton HF, Byrne MC, Kamradt T (2000)Microbial lipopeptides induce the production of IL-17 in Thcells. J Immunol 165(11):6107–6115

193. Kennedy J, Rossi DL, Zurawski SM, Vega F Jr, Kastelein RA,Wagner JL, Hannum CH, Zlotnik A (1996) Mouse IL-17: acytokine preferentially expressed by alpha beta TCR+ CD4-CD8-T cells. J Interferon Cytokine Res 16(8):611–617

194. Jovanovic DV, Di Battista JA, Martel-Pelletier J, Jolicoeur FC,He Y, Zhang M, Mineau F, Pelletier JP (1998) IL-17 stimulatesthe production and expression of proinflammatory cytokines, IL-beta and TNF-alpha, by human macrophages. J Immunol 160(7):3513–3521

195. Aggarwal S, Gurney AL (2002) IL-17: prototype member of anemerging cytokine family. J Leukoc Biol 71(1):1–8

196. Moseley TA, Haudenschild DR, Rose L, Reddi AH (2003)Interleukin-17 family and IL-17 receptors. Cytokine GrowthFactor Rev 14(2):155–174

197. Antonysamy MA, Fanslow WC, Fu F, Li W, Qian S, Troutt AB,Thomson AW (1999) Evidence for a role of IL-17 in organallograft rejection: IL-17 promotes the functional differentiationof dendritic cell progenitors. J Immunol 162(1):577–584

198. Burlingham WJ, Love RB, Jankowska-Gan E, Haynes LD, XuQ, Bobadilla JL, Meyer KC, Hayney MS, Braun RK, GreenspanDS, Gopalakrishnan B, Cai J, Brand DD, Yoshida S, CummingsOW, Wilkes DS (2007) IL-17-dependent cellular immunity tocollagen type V predisposes to obliterative bronchiolitis inhuman lung transplants. J Clin Invest 117(11):3498–3506.doi:10.1172/JCI28031

199. Vanaudenaerde BM, De Vleeschauwer SI, Vos R, Meyts I,Bullens DM, Reynders V, Wuyts WA, Van Raemdonck DE,Dupont LJ, Verleden GM (2008) The role of the IL23/IL17 axisin bronchiolitis obliterans syndrome after lung transplantation.Am J Transplant 8(9):1911–1920. doi:10.1111/j.1600-6143.2008.02321.x

200. Cao H, Lan Q, Shi Q, Zhou X, Liu G, Liu J, Tang G, Qiu C, XuJ, Fan H, Liu Z (2010) Anti-IL-23 antibody blockade of IL-23/IL-17 pathway attenuates airway obliteration in rat orthotopictracheal transplantation. Int Immunopharmacol. doi:10.1016/j.intimp.2010.11.007

201. Rollins BJ (1997) Chemokines. Blood 90(3):909–928202. Luster AD (1998) Mechanisms of disease: chemokines—

chemotactic cytokines that mediate inflammation. N Engl JMed 338(7):436–445

203. Strieter RM, Kunkel SL (1997) Chemokines and the lung. In:Crystal R, West J, Weibel E, Barnes P (eds) Lung: scientificfoundations, 2nd edn. Raven, New York, pp 155–186

204. Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer TA(1996) A highly efficacious lymphocyte chemoattractant, stromalcell-derived factor 1 (SDF-1). J Exp Med 184(3):1101–1109 (seecomments)

205. Loetscher M, Gerber B, Loetscher P, Jones SA, Piali L, Clark-Lewis I, Baggiolini M, Moser B (1996) Chemokine receptorspecific for IP10 and Mig: structure, function, and expression inactivated T-lymphocytes. J Exp Med 184(3):963–969 (seecomments)

206. Keane MP, Belperio JA, Arenberg DA, Burdick MD, Xu ZJ, XueYY, Strieter RM (1999) IFN-gamma-inducible protein-10 attenu-ates bleomycin-induced pulmonary fibrosis via inhibition of

Semin Immunopathol (2011) 33:135–156 153

Page 20: Lung transplantation: infection, inflammation, and the microbiome

angiogenesis. J Immunol 163(10):5686–5692 (in process cita-tion)

207. Keane MP, Belperio JA, Moore TA, Moore BB, Arenberg DA,Smith RE, Burdick MD, Kunkel SL, Strieter RM (1999)Neutralization of the CXC chemokine, macrophage inflammato-ry protein-2, attenuates bleomycin-induced pulmonary fibrosis. JImmunol 162(9):5511–5518

208. Moore BB, Arenberg DA, Addison CL, Keane MP, Strieter RM(1998) Tumor angiogenesis is regulated by CXC chemokines. JLab Clin Med 132(2):97–103

209. Baggiolini M, Dewald B, Moser B (1997) Human chemokines:an update. Annu Rev Immunol 15:675–705

210. Robinson LA, Nataraj C, Thomas DW, Howell DN, Griffiths R,Bautch V, Patel DD, Feng L, Coffman TM (2000) A role forfractalkine and its receptor (CX3CR1) in cardiac allograftrejection. J Immunol 165(11):6067–6072

211. Fong AM, Robinson LA, Steeber DA, Tedder TF, Yoshie O,Imai T, Patel DD (1998) Fractalkine and CX3CR1 mediate anovel mechanism of leukocyte capture, firm adhesion, andactivation under physiologic flow. J Exp Med 188(8):1413–1419

212. Pan Y, Lloyd C, Zhou H, Dolich S, Deeds J, Gonzalo JA, Vath J,Gosselin M, Ma J, Dussault B, Woolf E, Alperin G, Culpepper J,Gutierrez-Ramos JC, Gearing D (1997) Neurotactin, amembrane-anchored chemokine upregulated in brain inflamma-tion. Nature 387(6633):611–617 (published erratum appears inNature 1997 Sep 4;389(6646):100)

213. Bazan JF, Bacon KB, Hardiman G, Wang W, Soo K, Rossi D,Greaves DR, Zlotnik A, Schall TJ (1997) A new class ofmembrane-bound chemokine with a CX3C motif. Nature 385(6617):640–644

214. Murphy PM, Baggiolini M, Charo IF, Hebert CA, Horuk R,Matsushima K, Miller LH, Oppenheim JJ, Power CA (2000)International union of pharmacology. XXII. Nomenclature forchemokine receptors. Pharmacol Rev 52(1):145–176

215. Segerer S, Nelson PJ, Schlondorff D (2000) Chemokines,chemokine receptors, and renal disease: from basic science topathophysiologic and therapeutic studies. J Am Soc Nephrol 11(1):152–176

216. Monti G, Magnan A, Fattal M, Rain B, Humbert M, Mege JL,Noirclerc M, Dartevelle P, Cerrina J, Simonneau G, Galanaud P,Emilie D (1996) Intrapulmonary production of RANTES duringrejection and CMV pneumonitis after lung transplantation.Transplantation 61(12):1757–1762

217. Belperio JA, Burdick MD, Keane MP, Xue YY, Lynch JP 3rd,Daugherty BL, Kunkel SL, Strieter RM (2000) The role of theCC chemokine, RANTES, in acute lung allograft rejection. JImmunol 165(1):461–472

218. Suga M, Maclean AA, Keshavjee S, Fischer S, Moreira JM, LiuM (2000) RANTES plays an important role in the evolution ofallograft transplant-induced fibrous airway obliteration. Am JRespir Crit Care Med 162(5):1940–1948

219. Krishnadasan B, Farivar AS, Naidu BV, Woolley SM, Byrne K,Fraga CH, Mulligan MS (2004) Beta-chemokine function inexperimental lung ischemia–reperfusion injury. Ann Thorac Surg77(3):1056–1062

220. Belperio JA, Keane MP, Burdick MD, Lynch JP 3rd, Xue YY,Berlin A, Ross DJ, Kunkel SL, Charo IF, Strieter RM (2001)Critical role for the chemokine MCP-1/CCR2 in the pathogen-esis of bronchiolitis obliterans syndrome. J Clin Invest 108(4):547–556

221. Farivar AS, Krishnadasan B, Naidu BV, Woolley SM, MulliganMS (2003) The role of the beta chemokines in experimentalobliterative bronchiolitis. Exp Mol Pathol 75(3):210–216

222. Gelman AE, Okazaki M, Sugimoto S, Li W, Kornfeld CG, Lai J,Richardson SB, Kreisel FH, Huang HJ, Tietjens JR, ZinselmeyerBH, Patterson GA, Miller MJ, Krupnick AS, Kreisel D (2010)

CCR2 regulates monocyte recruitment as well as CD4 T1allorecognition after lung transplantation. Am J Transplant 10(5):1189–1199. doi:10.1111/j.1600-6143.2010.03101.x

223. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory Tcell development by the transcription factor Foxp3. Science 299(5609):1057–1061

224. Lee I, Wang L, Wells AD, Dorf ME, Ozkaynak E, Hancock WW(2005) Recruitment of Foxp3+ T regulatory cells mediatingallograft tolerance depends on the CCR4 chemokine receptor. JExp Med 201(7):1037–1044

225. Alferink J, Lieberam I, Reindl W, Behrens A, Weiss S, Huser N,Gerauer K, Ross R, Reske-Kunz AB, Ahmad-Nejad P, WagnerH, Forster I (2003) Compartmentalized production of CCL17 invivo: strong inducibility in peripheral dendritic cells contrastsselective absence from the spleen. J Exp Med 197(5):585–599

226. Forster R, Schubel A, Breitfeld D, Kremmer E, Renner-Muller I,Wolf E, Lipp M (1999) CCR7 coordinates the primary immuneresponse by establishing functional microenvironments in sec-ondary lymphoid organs. Cell 99(1):23–33

227. Sallusto F, Mackay CR, Lanzavecchia A (2000) The role ofchemokine receptors in primary, effector, and memory immuneresponses. Annu Rev Immunol 18:593–620

228. Cyster JG (1999) Chemokines and cell migration in secondarylymphoid organs. Science 286(5447):2098–2102

229. Gregson AL, Hoji A, Saggar R, Ross DJ, Kubak BM, JamiesonBD, Weigt SS, Lynch JP 3rd, Ardehali A, Belperio JA, Yang OO(2008) Bronchoalveolar immunologic profile of acute humanlung transplant allograft rejection. Transplantation 85(7):1056–1059. doi:10.1097/TP.0b013e318169bd85

230. Gregson AL, Hoji A, Palchevskiy V, Hu S, Weigt SS, Liao E,Derhovanessian A, Saggar R, Song S, Elashoff R, Yang OO,Belperio JA (2010) Protection against bronchiolitis obliteranssyndrome is associated with allograft CCR7+CD45RA− Tregulatory cells. PLoS One 5(6):e11354. doi:10.1371/journal.pone.0011354

231. Meloni F, Solari N, Miserere S, Morosini M, Cascina A, KlersyC, Arbustini E, Pellegrini C, Vigano M, Fietta AM (2008)Chemokine redundancy in BOS pathogenesis. A possible rolealso for the CC chemokines: MIP3-beta, MIP3-alpha, MDC andtheir specific receptors. Transpl Immunol 18(3):275–280.doi:10.1016/j.trim.2007.08.004

232. Imai T, Hieshima K, Haskell C, Baba M, Nagira M, NishimuraM, Kakizaki M, Takagi S, Nomiyama H, Schall TJ, Yoshie O(1997) Identification and molecular characterization of fractal-kine receptor CX3CR1, which mediates both leukocyte migra-tion and adhesion. Cell 91(4):521–530

233. Combadiere C, Gao J, Tiffany HL, Murphy PM (1998) Genecloning, RNA distribution, and functional expression ofmCX3CR1, a mouse chemotactic receptor for the CX3Cchemokine fractalkine. Biochem Biophys Res Commun 253(3):728–732

234. Maciejewski-Lenoir D, Chen S, Feng L, Maki R, Bacon KB(1999) Characterization of fractalkine in rat brain cells: migra-tory and activation signals for CX3CR-1-expressing microglia. JImmunol 163(3):1628–1635

235. Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNamaraRK, Streit WJ, Salafranca MN, Adhikari S, Thompson DA, BottiP, Bacon KB, Feng L (1998) Role for neuronally derivedfractalkine in mediating interactions between neurons andCX3CR1-expressing microglia. Proc Natl Acad Sci USA 95(18):10896–10901

236. Feng L, Chen S, Garcia GE, Xia Y, Siani MA, Botti P, WilsonCB, Harrison JK, Bacon KB (1999) Prevention of crescenticglomerulonephritis by immunoneutralization of the fractalkinereceptor CX3CR1 rapid communication. Kidney Int 56(2):612–620

154 Semin Immunopathol (2011) 33:135–156

Page 21: Lung transplantation: infection, inflammation, and the microbiome

237. Haskell CA, Hancock WW, Salant DJ, Gao W, Csizmadia V,Peters W, Faia K, Fituri O, Rottman JB, Charo IF (2001)Targeted deletion of CX(3)CR1 reveals a role for fractalkine incardiac allograft rejection. J Clin Invest 108(5):679–688

238. Taub DD, Lloyd AR, Conlon K, Wang JM, Ortaldo JR,Harada A, Matsushima K, Kelvin DJ, Oppenheim JJ (1993)Recombinant human interferon-inducible protein 10 is achemoattractant for human monocytes and T lymphocytesand promotes T cell adhesion to endothelial cells. J Exp Med177(6):1809–1814

239. Sallusto F, Lenig D, Mackay CR, Lanzavecchia A (1998)Flexible programs of chemokine receptor expression on humanpolarized T helper 1 and 2 lymphocytes. J Exp Med 187(6):875–883

240. Bonecchi R, Bianchi G, Bordignon PP, D'Ambrosio D, Lang R,Borsatti A, Sozzani S, Allavena P, Gray PA, Mantovani A,Sinigaglia F (1998) Differential expression of chemokinereceptors and chemotactic responsiveness of type 1 T helpercells (Th1s) and Th2s. J Exp Med 187(1):129–134

241. Maghazachi AA, Skalhegg BS, Rolstad B, Al-Aoukaty A (1997)Interferon-inducible protein-10 and lymphotactin induce thechemotaxis and mobilization of intracellular calcium in naturalkiller cells through pertussis toxin-sensitive and -insensitiveheterotrimeric G-proteins. FASEB J 11(10):765–774

242. Liao F, Rabin RL, Yannelli JR, Koniaris LG, Vanguri P, FarberJM (1995) Human Mig chemokine: biochemical and functionalcharacterization. J Exp Med 182(5):1301–1314

243. Rabin RL, Park MK, Liao F, Swofford R, Stephany D, FarberJM (1999) Chemokine receptor responses on T cells are achievedthrough regulation of both receptor expression and signaling. JImmunol 162(7):3840–3850

244. Qin S, Rottman JB, Myers P, Kassam N, Weinblatt M, LoetscherM, Koch AE, Moser B, Mackay CR (1998) The chemokinereceptors CXCR3 and CCR5 mark subsets of T cells associatedwith certain inflammatory reactions. J Clin Invest 101(4):746–754

245. Piali L, Weber C, LaRosa G, Mackay CR, Springer TA, Clark-Lewis I, Moser B (1998) The chemokine receptor CXCR3mediates rapid and shear-resistant adhesion-induction of effectorT lymphocytes by the chemokines IP10 and Mig. Eur J Immunol28(3):961–972

246. Zhai Y, Shen XD, Hancock WW, Gao F, Qiao B, Lassman C,Belperio JA, Strieter RM, Busuttil RW, Kupiec-Weglinski JW(2006) CXCR3+CD4+ T cells mediate innate immune function inthe pathophysiology of liver ischemia/reperfusion injury. JImmunol 176(10):6313–6322

247. Zhao DX, Hu Y, Miller GG, Luster AD, Mitchell RN, Libby P(2002) Differential expression of the IFN-gamma-inducibleCXCR3-binding chemokines, IFN-inducible protein 10, mono-kine induced by IFN, and IFN-inducible T cell alpha chemo-attractant in human cardiac allografts: association with cardiacallograft vasculopathy and acute rejection. J Immunol 169(3):1556–1560

248. Melter M, Exeni A, Reinders ME, Fang JC, McMahon G, GanzP, Hancock WW, Briscoe DM (2001) Expression of the chemo-kine receptor CXCR3 and its ligand IP-10 during human cardiacallograft rejection. Circulation 104(21):2558–2564

249. Kao J, Kobashigawa J, Fishbein MC, MacLellan WR, BurdickMD, Belperio JA, Strieter RM (2003) Elevated serum levels ofthe CXCR3 chemokine ITAC are associated with the develop-ment of transplant coronary artery disease. Circulation 107(15):1958–1961

250. Hancock WW, Lu B, Gao W, Csizmadia V, Faia K, King JA,Smiley ST, Ling M, Gerard NP, Gerard C (2000) Requirement ofthe chemokine receptor CXCR3 for acute allograft rejection. JExp Med 192(10):1515–1520

251. Hancock WW, Gao W, Csizmadia V, Faia KL, Shemmeri N,Luster AD (2001) Donor-derived IP-10 initiates development ofacute allograft rejection. J Exp Med 193(8):975–980

252. Miura M, Morita K, Kobayashi H, Hamilton TA, Burdick MD,Strieter RM, Fairchild RL (2001) Monokine induced by IFN-gamma is a dominant factor directing T cells into murine cardiacallografts during acute rejection. J Immunol 167(6):3494–3504

253. Baker MS, Chen X, Rotramel AR, Nelson JJ, Lu B, Gerard C,Kanwar Y, Kaufman DB (2003) Genetic deletion of chemokinereceptor CXCR3 or antibody blockade of its ligand IP-10modulates posttransplantation graft-site lymphocytic infiltratesand prolongs functional graft survival in pancreatic islet allograftrecipients. Surgery 134(2):126–133

254. Agostini C, Calabrese F, Rea F, Facco M, Tosoni A, Loy M,Binotto G, Valente M, Trentin L, Semenzato G (2001) Cxcr3 andits ligand CXCL10 are expressed by inflammatory cellsinfiltrating lung allografts and mediate chemotaxis of T cells atsites of rejection. Am J Pathol 158(5):1703–1711

255. Belperio JA, Keane MP, Burdick MD, Lynch JP 3rd, Xue YY, LiK, Ross DJ, Strieter RM (2002) Critical role for CXCR3chemokine biology in the pathogenesis of bronchiolitis obliter-ans syndrome. J Immunol 169(2):1037–1049

256. Medoff BD, Wain JC, Seung E, Jackobek R, Means TK, GinnsLC, Farber JM, Luster AD (2006) CXCR3 and its ligands in amurine model of obliterative bronchiolitis: regulation andfunction. J Immunol 176(11):7087–7095

257. Ebert LM, Schaerli P, Moser B (2005) Chemokine-mediatedcontrol of T cell traffic in lymphoid and peripheral tissues. MolImmunol 42(7):799–809

258. Lee BP, Chen W, Shi H, Der SD, Forster R, Zhang L (2006)CXCR5/CXCL13 interaction is important for double-negativeregulatory T cell homing to cardiac allografts. J Immunol 176(9):5276–5283

259. Belperio JA, Keane MP, Arenberg DA, Addison CL, Ehlert JE,Burdick MD, Strieter RM (2000) CXC chemokines in angio-genesis. J Leukoc Biol 68(1):1–8

260. Belperio JA, Keane MP, Burdick MD, Gomperts B, Xue YY,Hong K, Mestas J, Ardehali A, Mehrad B, Saggar R, Lynch JP,Ross DJ, Strieter RM (2005) Role of CXCR2/CXCR2 ligands invascular remodeling during bronchiolitis obliterans syndrome. JClin Invest 115(5):1150–1162

261. Fisher AJ, Donnelly SC, Hirani N, Haslett C, Strieter RM, DarkJH, Corris PA (2001) Elevated levels of interleukin-8 in donorlungs is associated with early graft failure after lung transplan-tation. Am J Respir Crit Care Med 163(1):259–265

262. De Perrot M, Sekine Y, Fischer S, Waddell TK, McRae K, LiuM, Wigle DA, Keshavjee S (2002) Interleukin-8 release duringearly reperfusion predicts graft function in human lung trans-plantation. Am J Respir Crit Care Med 165(2):211–215

263. Riise GC, Williams A, Kjellstrom C, Schersten H, AnderssonBA, Kelly FJ (1998) Bronchiolitis obliterans syndrome in lungtransplant recipients is associated with increased neutrophilactivity and decreased antioxidant status in the lung. Eur RespirJ 12(1):82–88

264. Riise GC, Andersson BA, Kjellstrom C, Martensson G, NilssonFN, Ryd W, Schersten H (1999) Persistent high BAL fluidgranulocyte activation marker levels as early indicators ofbronchiolitis obliterans after lung transplant. Eur Respir J 14(5):1123–1130

265. Zheng L, Walters EH, Ward C, Wang N, Orsida B, Whitford H,Williams TJ, Kotsimbos T, Snell GI (2000) Airway neutrophiliain stable and bronchiolitis obliterans syndrome patients follow-ing lung transplantation. Thorax 55(1):53–59

266. Elssner A, Vogelmeier C (2001) The role of neutrophils in thepathogenesis of obliterative bronchiolitis after lung transplanta-tion. Transpl Infect Dis 3(3):168–176

Semin Immunopathol (2011) 33:135–156 155

Page 22: Lung transplantation: infection, inflammation, and the microbiome

267. DiGiovine B, Lynch JP 3rd, Martinez FJ, Flint A, Whyte RI,Iannettoni MD, Arenberg DA, Burdick MD, Glass MC, WilkeCA, Morris SB, Kunkel SL, Strieter RM (1996) Bronchoalveolarlavage neutrophilia is associated with obliterative bronchiolitisafter lung transplantation: role of IL-8. J Immunol 157(9):4194–4202

268. Bhorade SM, Chen H, Molinero L, Liao C, Garrity ER,Vigneswaran WT, Shilling R, Sperling A, Chong A, AlegreML (2010) Decreased percentage of CD4+FoxP3+ cells inbronchoalveolar lavage from lung transplant recipients correlateswith development of bronchiolitis obliterans syndrome. Trans-plantation 90(5):540–546

269. Macpherson AJ, Harris NL (2004) Interactions between com-mensal intestinal bacteria and the immune system. Nat RevImmunol 4(6):478–485. doi:10.1038/nri1373

270. Podolsky DK (2002) The current future understanding ofinflammatory bowel disease. Best Pract Res Clin Gastroenterol16(6):933–943

271. Shanahan F (2002) Crohn's disease. Lancet 359(9300):62–69.doi:10.1016/S0140-6736(02)07284-7

272. Targan SR, Karp LC (2005) Defects in mucosal immunityleading to ulcerative colitis. Immunol Rev 206:296–305.doi:10.1111/j.0105-2896.2005.00286.x

273. Schmechel S, Konrad A, Diegelmann J, Glas J, Wetzke M,Paschos E, Lohse P, Goke B, Brand S (2008) Linking geneticsusceptibility to Crohn's disease with Th17 cell function: IL-22serum levels are increased in Crohn's disease and correlate withdisease activity and IL23R genotype status. Inflamm Bowel Dis14(2):204–212. doi:10.1002/ibd.20315

274. Kobayashi T, Okamoto S, Hisamatsu T, Kamada N, Chinen H,Saito R, Kitazume MT, Nakazawa A, Sugita A, Koganei K,Isobe K, Hibi T (2008) IL23 differentially regulates the Th1/Th17 balance in ulcerative colitis and Crohn's disease. Gut 57(12):1682–1689. doi:10.1136/gut.2007.135053

275. Fontenot JD, Rudensky AY (2005) A well adapted regulatorycontrivance: regulatory T cell development and the forkheadfamily transcription factor Foxp3. Nat Immunol 6(4):331–337.doi:10.1038/ni1179

276. Ivanov II, Frutos Rde L, Manel N, Yoshinaga K, Rifkin DB,Sartor RB, Finlay BB, Littman DR (2008) Specific microbiotadirect the differentiation of IL-17-producing T-helper cells in the

mucosa of the small intestine. Cell Host Microbe 4(4):337–349.doi:10.1016/j.chom.2008.09.009

277. Di Giacinto C, Marinaro M, Sanchez M, Strober W, Boirivant M(2005) Probiotics ameliorate recurrent Th1-mediated murinecolitis by inducing IL-10 and IL-10-dependent TGF-beta-bearing regulatory cells. J Immunol 174(6):3237–3246

278. O'Mahony C, Scully P, O'Mahony D, Murphy S, O'Brien F,Lyons A, Sherlock G, MacSharry J, Kiely B, Shanahan F,O'Mahony L (2008) Commensal-induced regulatory T cellsmediate protection against pathogen-stimulated NF-kappaBactivation. PLoS Pathog 4(8):e1000112. doi:10.1371/journal.ppat.1000112

279. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C,Davies J, Ervine A, Poulter L, Pachter L, Moffatt MF, CooksonWO (2010) Disordered microbial communities in asthmaticairways. PLoS One 5(1):e8578. doi:10.1371/journal.pone.0008578

280. Huang YJ, Kim E, Cox MJ, Brodie EL, Brown R, Wiener-Kronish JP, Lynch SV (2010) A persistent and diverse airwaymicrobiota present during chronic obstructive pulmonary diseaseexacerbations. OMICS 14(1):9–59. doi:10.1089/omi.2009.0100

281. Bittar F, Richet H, Dubus JC, Reynaud-Gaubert M, Stremler N,Sarles J, Raoult D, Rolain JM (2008) Molecular detection ofmultiple emerging pathogens in sputa from cystic fibrosis patients.PLoS ONE 3(8):e2908. doi:10.1371/journal.pone.0002908

282. Armougom F, Bittar F, Stremler N, Rolain JM, Robert C, DubusJC, Sarles J, Raoult D, La Scola B (2009) Microbial diversity inthe sputum of a cystic fibrosis patient studied with 16S rDNApyrosequencing. Eur J Clin Microbiol Infect Dis 28(9):1151–1154. doi:10.1007/s10096-009-0749-x

283. Harris JK, De Groote MA, Sagel SD, Zemanick ET, Kapsner R,Penvari C, Kaess H, Deterding RR, Accurso FJ, Pace NR (2007)Molecular identification of bacteria in bronchoalveolar lavagefluid from children with cystic fibrosis. Proc Natl Acad Sci USA104(51):20529–20533. doi:10.1073/pnas.0709804104

284. Sibley CD, Rabin H, Surette MG (2006) Cystic fibrosis: apolymicrobial infectious disease. Future Microbiol 1:53–61.doi:10.2217/17460913.1.1.53

285. Round JL, Mazmanian SK (2009) The gut microbiota shapesintestinal immune responses during health and disease. Nat RevImmunol 9(5):313–323

156 Semin Immunopathol (2011) 33:135–156