Advertisement
Original Article| Volume 18, ISSUE 5, P630-635, September 2019

Pseudomonas aeruginosa colonization causes PD-L1 overexpression on monocytes, impairing the adaptive immune response in patients with cystic fibrosis

Open ArchivePublished:November 12, 2018DOI:https://doi.org/10.1016/j.jcf.2018.11.002

      Highlights

      • P. aeruginosa is associated with PD-L1 overexpression in patients with CF.
      • Endotoxin Tolerance is a CF hallmark in patients colonized by P. aeruginosa.
      • Translocation of P. aeruginosa LPS induces PD-L1 expression and Endotoxin Tolerance.
      • Colistimethate ameliorates the effects of LPS translocation

      Abstract

      Background

      Cystic fibrosis (CF) is an endotoxin tolerance (ET)-related disease. Given that increased PD-L1 has been reported in ET, its expression and physiological effects on cystic fibrosis monocytes should be studied.

      Methods

      We analyzed the phenotype and ex vivo response of immune system cells in 32 patients with CF, 19 of them colonized by Pseudomonas aeruginosa. An in vitro model was developed of Pseudomonas aeruginosa colonization using purified lipopolysaccharides (LPS) from one of the most prevalent strains in patients with CF (a CF-adapted Pseudomonas aeruginosa ST395 clone). Changes in the immune response, including cytokine production and T-lymphocyte proliferation, as well as expression of PD-L1, were evaluated.

      Results

      PD-L1 was overexpressed in the monocytes of patients with CF compared with healthy volunteers, and levels of this immune checkpoint were associated with Pseudomonas aeruginosa colonization. In addition, patients with Pseudomonas aeruginosa colonization showed a patent ET status, including poor inflammatory response, reduced HLA-DR expression and T-lymphocyte proliferation impairment. PD-L1/PD-1 blocking assays reverted the impaired adaptive response. Ultimately, monocytes from healthy volunteers cultured in the presence of the clinically relevant strain of Pseudomonas aeruginosa or serum collected from patients with CF colonized by Pseudomonas aeruginosa reproduced the previous observed features.

      Conclusions

      Pseudomonas aeruginosa colonization in patients with CF was associated with PD-L1 overexpression and impaired T cell response, and LPS from this pathogen induced the observed phenotype. Our findings open new avenues for the use of anti-PD-1/PD-L1 immunotherapy in patients with CF who are colonized by Pseudomonas aeruginosa.

      Keywords

      1. Introduction

      Cystic fibrosis (CF) is a disease resulting from mutations in the gene that codifies the chloride ion channel (CFTR) [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ]. Patients with CF have high infection rates compared with the normal population. Infections in patients with CF are associated with disease complications, and the mortality of these patients is usually the result of chronic lower airway bacterial colonization [
      • Jain K.
      • Smyth A.R.
      Current dilemmas in antimicrobial therapy in cystic fibrosis.
      ]. According to a number of studies, Pseudomonas aeruginosa is the most prevalent Gram-negative bacterial pathogen in patients with CF, reaching rates of up to 49.56% [
      • Salsgiver E.L.
      • Fink A.K.
      • Knapp E.A.
      • Lipuma J.J.
      • Olivier K.N.
      • Marshall B.C.
      • et al.
      Changing epidemiology of the respiratory bacteriology of patients with cystic fibrosis.
      ]. Although chronic Pseudomonas aeruginosa infection rates are decreasing due to advances in clinical care and infection prevention [
      • Crull M.R.
      • Somayaji R.
      • Ramos K.J.
      • Caldwell E.
      • Mayer-Hamblett N.
      • Aitken M.L.
      • et al.
      Changing rates of chronic Pseudomonas aeruginosa infections in cystic fibrosis: a population-based cohort study.
      ], the infection still has a major influence on the clinical outcome of patients with CF [
      • Elborn J.S.
      Cystic fibrosis.
      ,
      • Gilligan P.H.
      Infections in patients with cystic fibrosis: diagnostic microbiology update.
      ,
      • de Dios Caballero J.
      • Vida R.
      • Cobo M.
      • Máiz L.
      • Suárez L.
      • Galeano J.
      • et al.
      Individual patterns of complexity in cystic fibrosis lung microbiota, including predator bacteria, over a 1-year period.
      ]. In fact, current efforts are focused on the development of vaccines against Pseudomonas aeruginosa. Thus, several vaccines dedicated to patients who have not yet been colonized or who have intermittent colonization have been evaluated without success [

      Johansen HK, Gotzsche PC. Vaccines for preventing infection with Pseudomonas aeruginosa in cystic fibrosis. The cochrane database of systematic reviews. 2015:Cd001399.

      ,
      • Talwalkar J.S.
      • Murray T.S.
      The approach to pseudomonas aeruginosa in cystic fibrosis.
      ]. Therefore, antibiotics appear to be the only option of treatment available to control the bacterial population [
      • Stefani S.
      • Campana S.
      • Cariani L.
      • Carnovale V.
      • Colombo C.
      • Lleo M.M.
      • et al.
      Relevance of multidrug-resistant pseudomonas aeruginosa infections in cystic fibrosis.
      ,
      • S W.S.Y.
      • Li J.
      • Patel K.
      • Wilson J.W.
      • Dooley M.J.
      • George J.
      • et al.
      Pulmonary and systemic pharmacokinetics of inhaled and intravenous colistin methanesulfonate in cystic fibrosis patients: targeting advantage of inhalational administration.
      ].
      The high frequency of pathogen colonization suggests a significant deficiency in the patient's innate immune system [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ,
      • Bonfield T.
      • Chmiel J.F.
      Impaired innate immune cells in cystic fibrosis: is it really a surprise?.
      ]. Therefore, identifying those patients with a suppressed immune system would be advantageous in terms of choosing treatment and preventing infection-derived complications. Although most studies have been focused on immune system changes at a local pulmonary level [
      • Bonfield T.L.
      • Panuska J.R.
      • Konstan M.W.
      • Hilliard K.A.
      • Hilliard J.B.
      • Ghnaim H.
      • et al.
      Inflammatory cytokines in cystic fibrosis lungs.
      ,
      • Reynolds C.J.
      • Quigley K.
      • Cheng X.
      • Suresh A.
      • Tahir S.
      • Ahmed-Jushuf F.
      • et al.
      Lung defense through interleukin-8 carries a cost of chronic lung remodeling and impaired function.
      ], we have previously reported the role of peripheral cells in patients with CF. Due to the translocation of bacterial products, mainly lipopolysaccharides (LPS) [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ], blood monocytes isolated from these patients exhibited a patent refractory status, also known as endotoxin tolerance (ET) [
      • del Fresno C.
      • Gomez-Pina V.
      • Lores V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Rojo B.
      • et al.
      Monocytes from cystic fibrosis patients are locked in an LPS tolerance state: down-regulation of TREM-1 as putative underlying mechanism.
      ,
      • del Fresno C.
      • Garcia-Rio F.
      • Gomez-Pina V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Jurado T.
      • et al.
      Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients.
      ,
      • Biswas S.K.
      • Lopez-Collazo E.
      Endotoxin tolerance: new mechanisms, molecules and clinical significance.
      ,
      • Lopez-Collazo E.
      • del Fresno C.
      Pathophysiology of endotoxin tolerance: mechanisms and clinical consequences.
      ]. These cells were unable to orchestrate an inflammatory response, and they show impaired antigen presentation when they are stimulated ex vivo [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ,
      • del Fresno C.
      • Gomez-Pina V.
      • Lores V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Rojo B.
      • et al.
      Monocytes from cystic fibrosis patients are locked in an LPS tolerance state: down-regulation of TREM-1 as putative underlying mechanism.
      ,
      • del Fresno C.
      • Garcia-Rio F.
      • Gomez-Pina V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Jurado T.
      • et al.
      Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients.
      ]. In fact, a clinical trial has been approved to examine the potential of circulating blood cell monocytes as a predictive biomarker of osteoporosis in patients with CF (NCT03492567).
      In recent years, crosstalk between programmed cell death ligand 1 (PD-L1) and its receptor, PD-1, has emerged as an important mechanism in the immune response, not only in cancer but also in infectious diseases. Several studies have indicated the PD-L1/PD-1 axis as a promising therapy in an increasing number of clinical contexts [
      • Topalian S.L.
      • Hodi F.S.
      • Brahmer J.R.
      • Gettinger S.N.
      • Smith D.C.
      • McDermott D.F.
      • et al.
      Safety, activity, and immune correlates of anti-PD-1 antibody in cancer.
      ,
      • El Annan J.
      • Goyal S.
      • Zhang Q.
      • Freeman G.J.
      • Sharpe A.H.
      • Dana R.
      Regulation of T-cell chemotaxis by programmed death-ligand 1 (PD-L1) in dry eye-associated corneal inflammation.
      ]. For example, Patil et al. have shown that Pseudomonas aeruginosa colonization increased expression of PD-L1 on splenic antigen presenting cells [
      • Patil N.K.
      • Luan L.
      • Bohannon J.K.
      • Hernandez A.
      • Guo Y.
      • Sherwood E.R.
      Frontline science: anti-PD-L1 protects against infection with common bacterial pathogens after burn injury.
      ]. In addition, treatment with anti-PD-L1 antibodies protects against infection due to increased interferon-γ secretion in CD8+ T cells and wound-draining lymph nodes after burn wound sepsis [
      • Patil N.K.
      • Luan L.
      • Bohannon J.K.
      • Hernandez A.
      • Guo Y.
      • Sherwood E.R.
      Frontline science: anti-PD-L1 protects against infection with common bacterial pathogens after burn injury.
      ]. We have demonstrated that PD-L1 is a robust ET marker, and its control appears to be mediated by hypoxia-inducible factor-1α in a cohort of patients with sepsis [
      • Avendano-Ortiz J.
      • Maroun-Eid C.
      • Martin-Quiros A.
      • Toledano V.
      • Cubillos-Zapata C.
      • Gomez-Campelo P.
      • et al.
      PD-L1 Overexpression during endotoxin tolerance impairs the adaptive immune response in septic patients via HIF1alpha.
      ]. However, PD-L1 expression in the context of CF has not been explored, especially in those patients colonized by Pseudomonas aeruginosa.
      Herein, we have examined the PD-L1 and PD-1 levels on monocytes and lymphocytes, respectively, in 32 patients with CF. We have also analyzed the potential association between the expression of PD-L1 and the clinical parameters available, such as Pseudomonas aeruginosa colonization and type of infection. Finally, we have corroborated the results obtained in patients using an in vitro model.

      2. Materials and methods

      2.1 Study design

      The study was conducted in accordance with the ethical guidelines of the 1975 Declaration of Helsinki and was approved by the Committee for Human Subjects of La Paz University Hospital. All the participants provided written consent for the study. Patients with CF were prospectively enrolled from the Pneumology Department of La Paz University Hospital, Madrid, Spain. Patients were nonsmoking adults who fulfilled the diagnostic criteria for CF (clinical phenotype, sweat testing, and CFTR genotyping [
      • Elborn J.S.
      Cystic fibrosis.
      ]). Exclusion criteria included a history of chronic obstructive pulmonary disease, asthma, or other active lung disease, mental or physical handicap, or other significant diseases such as diabetes mellitus, congestive heart failure, ischemic or valvular cardiopathy, or neuromuscular disease. None of the patients had experienced a lung exacerbation of respiratory tract infection within the previous 4 weeks, and none had had received oral corticosteroid therapy for at least 3 months before the study. The clinical data on the patients included in the study are summarized in Supplementary Table S1. Age and sex-matched healthy volunteers (HVs) were also included as controls.

      2.2 Reagents

      Roswell Park Memorial Institute (RPMI) medium (Invitrogen) was used for the cell cultures. The LPS, from Escherichia coli O111:B4, was obtained from Sigma-Aldrich. Carboxyfluorescein succinimidyl ester (CFSE) for the proliferation assays was purchased from Thermo Fisher. The CF-adapted ST395 P. aeruginosa clone was isolated from a patient who had been colonized during 13 years. LPS was obtained as previously described [
      • Davis Jr., M.R.
      • Goldberg J.B.
      Purification and visualization of lipopolysaccharide from Gram-negative bacteria by hot aqueous-phenol extraction.
      ]. Colistimethate Sodium (polymyxin E) was purchased from Genéricos Españoles (GES) and dissolved in RPMI medium. The lymphocyte stimulus pokeweed (PWD) was purchased from Sigma-Aldrich. To inhibit PD-L1/PD-1 interaction, a fully human IgG4 (S228P) anti-PD-1 receptor-blocking monoclonal antibody was used (Bristol-Myers Squibb). All the reagents used for the cell cultures were endotoxin-free, as assayed with the Limulus amebocyte lysate test (Cambrex).

      2.3 Cytokine production in ex vivo LPS stimulation

      Fresh blood from patients with CF and paired HV were diluted in RPMI medium in a 1:1 proportion. The cells were then stimulated with 10 ng/mL of LPS. After 3 and 6 h, the cultures were centrifuged at 18,000 g for 10 min and supernatants were collected for their analysis by cytometric bead array (CBA). Tumor necrosis factor alpha (TNFα), interleukin (IL)10, and IL6 protein levels in the culture supernatants were determined using the Human Inflammatory CBA kit (BD Biosciences), following the manufacturer's protocol. Supernatants were analyzed by flow cytometry using a BD FACSCalibur flow cytometer (BD Biosciences).

      2.4 Peripheral blood mononuclear cell isolation and flow cytometry analysis

      The peripheral blood mononuclear cells (PBMCs) from patients with CF and the controls were isolated using Ficoll-Plus gradient (GE Healthcare Bio-Sciences) [
      • del Fresno C.
      • Garcia-Rio F.
      • Gomez-Pina V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Jurado T.
      • et al.
      Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients.
      ]. The cells were labeled with allophycocyanin (APC)-conjugated anti-human CD14, fluorescein isothiocyanate-conjugated anti-human HLA-DR, APC-conjugated anti-human CD3 (all from Immunostep, Spain); and phycoerythrin-conjugated anti-human PD-L1 (Miltenyi Biotec, USA). Matched isotype antibodies were used as negative controls. The cells were incubated for 30 min at 4 °C in the dark. The data were acquired by flow cytometry using a BD FACSCalibur flow cytometer (BD Biosciences) and analyzed with FlowJo vX.0.7 software (FlowJo, LLC). Gating strategy is show on Supplementary Fig. S1.

      2.5 T cell proliferation assays

      Proliferation was analyzed by flow cytometry of CFSE labeled cells as reported previously [
      • Avendano-Ortiz J.
      • Maroun-Eid C.
      • Martin-Quiros A.
      • Toledano V.
      • Cubillos-Zapata C.
      • Gomez-Campelo P.
      • et al.
      PD-L1 Overexpression during endotoxin tolerance impairs the adaptive immune response in septic patients via HIF1alpha.
      ]. Briefly, PBMCs were isolated using Ficoll-Plus gradient (GE Healthcare Bio-Sciences) [
      • del Fresno C.
      • Garcia-Rio F.
      • Gomez-Pina V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Jurado T.
      • et al.
      Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients.
      ]. The PBMCs were then labeled and cultured in a 96-well plate with fresh RPMI medium and were stimulated or not with PWD (2.5 μg/mL) and treated with 5 μg/mL of a fully human IgG4 (S228P) anti-PD-1 receptor-blocking monoclonal antibody (Bristol-Myers Squibb).

      2.6 Statistical analysis

      The number of experiments analyzed is indicated in each figure. In summary, statistical significance was calculated using an unpaired Mann–Whitney U test or a paired Wilcoxon t-test, as appropriate, depending on the specific assay. The statistical significance was set at p < .05, and the statistical analyses were conducted using Prism 6.0 (GraphPad) and SPSS version 23 (IBM) software.

      3. Results

      3.1 PD-L1 on monocytes and PD-1 on lymphocytes are overexpressed in patients with cystic fibrosis

      Given that circulating monocytes from patients with CF have traditionally been associated with an ET phenotype [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ,
      • del Fresno C.
      • Gomez-Pina V.
      • Lores V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Rojo B.
      • et al.
      Monocytes from cystic fibrosis patients are locked in an LPS tolerance state: down-regulation of TREM-1 as putative underlying mechanism.
      ,
      • del Fresno C.
      • Garcia-Rio F.
      • Gomez-Pina V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Jurado T.
      • et al.
      Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients.
      ], and PD-L1 overexpression is one of the hallmarks of ET [
      • Avendano-Ortiz J.
      • Maroun-Eid C.
      • Martin-Quiros A.
      • Toledano V.
      • Cubillos-Zapata C.
      • Gomez-Campelo P.
      • et al.
      PD-L1 Overexpression during endotoxin tolerance impairs the adaptive immune response in septic patients via HIF1alpha.
      ], we evaluated the expression of this immune checkpoint ligand. We found PD-L1 overexpression on CF monocytes as well as high levels of sPD-L1 in plasma from CF patients (Fig. 1A ). PD-1 was also overexpressed in both CD4 and CD8 lymphocytes from patients (Fig. 1B). In full agreement, T lymphocyte (CD3) proliferation was significantly reduced in cells from patients with CF compared with HVs (Fig. 1C). This effect was negated when PD-L1/PD-1 crosstalk was blocked using an anti-PD-1 antibody (Fig. 1C).
      Fig. 1
      Fig. 1PD-L1 on monocytes and PD-1 on T lymphocytes are overexpressed in patients with CF, corresponding to an impaired T-cell response.
      (A) PD-L1 expression on monocytes from patients with CF and HVs was measured by flow cytometry. Percentage of PD-L1+ on CD14+ gated cells is shown on left panel. Soluble PD-L1 (sPD-L1) in sera from patients with CF and HVs was measured by ELISA and summarized on right panel. (n = 32 patients with CF; n = 15 HVs). (B) PD-1 expression on T lymphocytes from patients with CF and HVs were measured by flow cytometry. Percentages of PD-1+ on both CD4+ and CD8+ gated cells are shown (n = 32 patients with CF; n = 15 HVs). (C) T cell proliferation in patients with CF and HVs was measured with CFSE proliferation assay by flow cytometry. Percentage of proliferation (defined as CFSEdim) on gated CD3+ cells is shown. (n = 16 patients with CF randomly selected; n = 7 HVs) *, p < .05; **, p < .01; ***, p < .001 vs. HVs using an unpaired t-test. Data are expressed as the mean ± SD.

      3.2 PD-L1 overexpression on monocytes and T cell proliferation impairment is associated with Pseudomonas aeruginosa colonization in patients with CF

      Patients were classified in subgroups according to relevant parameters: sex, CFTR mutation, age at diagnosis, number of infections in the last year, airflow limitation, body mass index and microbiological data. We then evaluated the influence of these parameters on the PD-L1 expression on their monocytes. No correlations were found with sex, mutations, age at diagnosis, airflow limitation, number of infections in the last year, and body mass index (Supplementary Fig. S2). In contrast, colonization by Pseudomonas aeruginosa correlated not only with PD-L1 overexpression on monocytes (Fig. 2A) but also for sPD-L1 levels in serum (Fig. 2B). Note that patients with Staphylococcus aureus colonization but not Pseudomonas aeruginosa did not exhibit PD-L1 overexpression (Supplementary Fig. S3). In agreement, T cell proliferation impairment was greater in cells from patients with Pseudomonas aeruginosa than in patients free from this pathogen. As expected, an anti-PD-1 antibody reverted the adaptive immune response impairment (Fig. 2C).

      3.3 Monocytes from patients with CF colonized by Pseudomonas aeruginosa exhibited a patent ET phenotype

      Previously, we reported that the monocytes of patients with CF are locked into a refractory state in which they are unable to respond to an endotoxin stimulus [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ,
      • del Fresno C.
      • Gomez-Pina V.
      • Lores V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Rojo B.
      • et al.
      Monocytes from cystic fibrosis patients are locked in an LPS tolerance state: down-regulation of TREM-1 as putative underlying mechanism.
      ,
      • del Fresno C.
      • Garcia-Rio F.
      • Gomez-Pina V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Jurado T.
      • et al.
      Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients.
      ]. Here, we have explored the main ET hallmarks in monocytes from patients with CF classified according to the presence of Pseudomonas aeruginosa colonization. Although TNFα and IL6 showed a patent downregulation after an ex vivo LPS challenge of these cells isolated from colonized patients (Fig. 3A and B), IL10 exhibited significant activation (Fig. 3C). Analysis of HLA-DR levels revealed a diminished expression in colonized patients (Fig. 3D). In order to corroborate whether the ET phenotype and PD-L1 overexpression occurs in the same cell population, we assessed a Spearman correlation test between basal PD-L1 and HLA-DR increment after stimulation. As Supplementary Fig. S4 illustrates, we have observed a negative correlation between these two parameters, in agreement to the observed in sepsis [
      • Avendano-Ortiz J.
      • Maroun-Eid C.
      • Martin-Quiros A.
      • Toledano V.
      • Cubillos-Zapata C.
      • Gomez-Campelo P.
      • et al.
      PD-L1 Overexpression during endotoxin tolerance impairs the adaptive immune response in septic patients via HIF1alpha.
      ]. Note that no differences in plasma cytokines were found between groups, ruling out the possibility of chronic basal inflammation in patients (Supplementary Fig. S5). Collectively, these data suggest a deeper refractory status in those patients with CF colonized by Pseudomonas aeruginosa. Note that none of the other clinical parameters have been shown to have a clear association with cytokine production (data not shown).

      3.4 LPS isolated from a clinically relevant Pseudomonas aeruginosa strain and sera from patients colonized by this pathogen induced PD-L1 overexpression on naïve monocytes

      To evaluate the role of LPS in the observed phenomenon, we isolated LPS from a clinically relevant Pseudomonas aeruginosa strain. Next, human monocytes from healthy donors were challenged with the isolated LPS and PD-L1 overexpression was verified (Fig. 4A ). Similar results were obtained in the presence of sera from patients colonized by Pseudomonas aeruginosa (Fig. 4B, left panel). The effect was negated when colistimethate (polymyxin E; PmE, an LPS scavenger [
      • P M.
      • S S.
      • C D.
      • K P.
      • C S.
      • E L.
      • et al.
      Colistin reduces LPS-triggered inflammation in a human sepsis model in vivo: a randomized controlled trial.
      ]) was added to the culture (Fig. 4B, right panel). In line, sPD-L1 levels detected in sera from patients colonized by Pseudomonas aeruginosa were lower in those under inhaled colistimethate sodium (Fig. 5) suggesting a role for colistimethate in the Pseudomonas aeruginosa LPS translocation to peripheral blood.
      Fig. 2
      Fig. 2Pseudomonas aeruginosa colonization in patients with CF is associated with higher PD-L1 overexpression.
      (A) PD-L1 expression on monocytes from patients with CF by flow cytometry; patients were classified according the presence of Pseudomonas aeruginosa in their sputum. Percentage of PD-L1+ on CD14+ gated cells is shown (n = 32 patients with CF; n = 19 CF patients with Pseudomonas aeruginosa; n = 15 HVs). (B) Soluble PD-L1 (sPD-L1) in sera from patients with CF and HVs was measured by ELISA (n = 32 patients with CF; n = 19 CF patients with Pseudomonas aeruginosa; n = 15 HVs). (C) T cell proliferation in patients with CF was measured with CFSE proliferation assay. Percentage of proliferation (defined as CFSEdim) on gated CD3+ cells is shown (n = 16 patients with CF randomly selected and n = 7 HVs). “+”, patients with Pseudomonas aeruginosa colonization; “-”, patients without Pseudomonas aeruginosa colonization. *, p < .05 vs. noncolonized patients, using an unpaired t-test. Data are expressed as the mean ± SD.
      Fig. 3
      Fig. 3Monocytes from patients with CF colonized by Pseudomonas aeruginosa exhibit a more pronounced ET phenotype.
      Whole blood from patients with CF was stimulated with 10 ng/mL of LPS for 3 h. Inflammatory cytokine production was measured by CBA. HLA-DR expression was analyzed by flow cytometry. Concentrations of TNFα (A), IL-6 (B) and IL-10 (C) supernatants are shown. HLA-DR expression data as fold (ratio between mean intensity of fluorescence (MIF) on LPS stimulated cells divided by MIF on unstimulated cells) are show (D). “+”, patients with Pseudomonas aeruginosa colonization (n = 19); “-”, patients without Pseudomonas aeruginosa colonization (n = 13). *, p < .05 vs. noncolonized patients, using an unpaired t-test. Data are expressed as the mean ± SD.
      Fig. 4
      Fig. 4LPS and sera from patients with CF colonized by Pseudomonas aeruginosa induce PD-L1 expression in naïve monocytes, which is reverted by polymyxin E.
      (A) Monocytes from HVs were stimulated with 100 ng/mL of LPS from a Pseudomonas aeruginosa ST395 clone, and PD-L1 expression was analyzed by flow cytometry. Percentage of PD-L1+ on gated CD14+ cells is shown. (B) Monocytes from HVs were stimulated with 10% patient sera pretreated or not with polymyxin E (150 IU/mL); PD-L1 expressions were analyzed by flow cytometry. Percentage of PD-L1+ on gated CD14+ cells is shown. “+”, treated with sera from patients with Pseudomonas aeruginosa colonization (n = 5); “-” treated with sera from patients without Pseudomonas aeruginosa colonization (n = 5). *, p < .05; ***, p < .001 using a paired t-test. The data are pooled from four independent experiments and are expressed as the mean ± SEM.
      Fig. 5
      Fig. 5Soluble PD-L1 levels are low in sera from patients colonized by Pseudomonas aeruginosa treated with colistimethate.
      Soluble PD-L1 in sera from patients with CF classified according to Pseudomonas aeruginosa colonization and inhaled colistimethate sodium treatment was measured by ELISA (n = 32 patients with CF; n = 15 HVs). **: p < .01 versus noncolonized patients, using an unpaired t-test.

      4. Discussion

      Infections remain the major cause of morbidity and mortality among patients with CF [
      • Jain K.
      • Smyth A.R.
      Current dilemmas in antimicrobial therapy in cystic fibrosis.
      ,
      • Ralhan A.
      • Laval J.
      • Lelis F.
      • Ballbach M.
      • Grund C.
      • Hector A.
      • et al.
      Current concepts and controversies in innate immunity of cystic fibrosis lung disease.
      ]. The high frequency of pathogen colonization in patients with CF suggests a significant deficiency in their innate immune system [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ,
      • Bonfield T.
      • Chmiel J.F.
      Impaired innate immune cells in cystic fibrosis: is it really a surprise?.
      ,
      • Hartl D.
      • Gaggar A.
      • Bruscia E.
      • Hector A.
      • Marcos V.
      • Jung A.
      • et al.
      Innate immunity in cystic fibrosis lung disease.
      ]. Thus, identifying those patients with immunosuppression could confer an advantage in choosing optimal treatment and preventing infection-derived complications. Although basal chronic inflammation and neutrophilia in the lungs of CF patients clearly leads to a decline in lung function [
      • Bonfield T.L.
      • Panuska J.R.
      • Konstan M.W.
      • Hilliard K.A.
      • Hilliard J.B.
      • Ghnaim H.
      • et al.
      Inflammatory cytokines in cystic fibrosis lungs.
      ,

      Reynolds CJ, Quigley K, Cheng X, Suresh A, Tahir S, Ahmed-Jushuf F, et al. Lung defense through interleukin-8 carries a cost of chronic lung remodeling and impaired function. Am J Respir Cell Mol Biol[0:null].

      ], the immune response of blood cells is still controversial [
      • Ralhan A.
      • Laval J.
      • Lelis F.
      • Ballbach M.
      • Grund C.
      • Hector A.
      • et al.
      Current concepts and controversies in innate immunity of cystic fibrosis lung disease.
      ].
      Here, we have observed no basal inflammatory differences comparing the sera of patients and HVs regarding IL6 and TNFα cytokine levels in sera, which is in accordance with others' observations [
      • Cantin A.M.
      • Bilodeau G.
      • Larivee P.
      • Richter M.V.
      Plasma biomarkers and cystic fibrosis lung disease.
      ,
      • Paats M.S.
      • Bergen I.M.
      • Bakker M.
      • Hoek R.A.
      • Nietzman-Lammering K.J.
      • Hoogsteden H.C.
      • et al.
      Cytokines in nasal lavages and plasma and their correlation with clinical parameters in cystic fibrosis.
      ]. However, to evaluate immune status properly, we must study not only basal immunological status but also the capability to generate an appropriate immune response against an insult. We have previously demonstrated that the monocytes of patients with CF are in an ET status in which they were unable to respond properly against LPS stimulus [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ,
      • del Fresno C.
      • Gomez-Pina V.
      • Lores V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Rojo B.
      • et al.
      Monocytes from cystic fibrosis patients are locked in an LPS tolerance state: down-regulation of TREM-1 as putative underlying mechanism.
      ,
      • del Fresno C.
      • Garcia-Rio F.
      • Gomez-Pina V.
      • Soares-Schanoski A.
      • Fernandez-Ruiz I.
      • Jurado T.
      • et al.
      Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients.
      ,
      • Biswas S.K.
      • Lopez-Collazo E.
      Endotoxin tolerance: new mechanisms, molecules and clinical significance.
      ,
      • Lopez-Collazo E.
      • del Fresno C.
      Pathophysiology of endotoxin tolerance: mechanisms and clinical consequences.
      ]. We have corroborated these data and have added to our study the recently discovered ET marker PD-L1 [
      • Avendano-Ortiz J.
      • Maroun-Eid C.
      • Martin-Quiros A.
      • Toledano V.
      • Cubillos-Zapata C.
      • Gomez-Campelo P.
      • et al.
      PD-L1 Overexpression during endotoxin tolerance impairs the adaptive immune response in septic patients via HIF1alpha.
      ]. According to our findings, PD-L1 overexpression on monocytes appears to be a novel characteristic of the CF circulating immune system. Moreover, the data regarding T-cell proliferation with anti PD-1 monoclonal antibodies not only confirms the functional implication of this observation, but also opens the possibility of evaluating this novel therapeutic option for treating chronic infections in some patients with CF, increasing their clinical management. Ingersoll's et al. observed how a PD-L1 upregulation on CF neutrophils did not produce T cell suppression [
      • Ingersoll S.A.
      • Laval J.
      • Forrest O.A.
      • Preininger M.
      • Brown M.R.
      • Arafat D.
      • et al.
      Mature cystic fibrosis airway neutrophils suppress T cell function: evidence for a role of arginase 1 but not programmed death-ligand 1.
      ]. On the contrary, our data illustrated a crucial role of PD-L1 in T cell suppression. This inconsistency took place when the cohort was divided into two groups, according to Pseudomonas aeruginosa colonization (please compare Fig. 2C and Fig. 1C). Moreover, monocytes, but not neutrophils, were able to express antigen presentation molecules such as HLA-DR, which we have shown to be deregulated in this pathology. Additionally, it has been reported that PWD-induced T cell proliferation requires monocytes activation [
      • Pryjma J.
      • Ernst M.
      • Fetting R.
      • Woloszyn M.
      • Zembala M.
      • Flad H.D.
      The role of monocytes in the induction and regulation of IFN-gamma production by lectin-activated human T lymphocytes.
      ,
      • Gallart T.
      • Angel De La Fuente M.
      • Josep Barcelo J.
      • Alberola-Ila J.
      • Lozano F.
      Desialylation of T lymphocytes overcomes the monocyte dependency of pokeweed mitogen-induced T-cell activation.
      ]. Altogether, these data indicate that not all patients, but rather those with P. aeruginosa colonization would be beneficiaries of anti-PD-1 therapy. Along these lines, PD-L1 levels on monocytes could be better predictors for immunotherapy efficacy than PD-L1 expression on neutrophils, which is in line with what is observed in cancer [
      • Krieg C.
      • Nowicka M.
      • Guglietta S.
      • Schindler S.
      • Hartmann F.J.
      • Weber L.M.
      • et al.
      High-dimensionalsingle-cell analysis predicts response to anti-PD-1 immunotherapy.
      ].
      In a previous study with a smaller cohort including all patients with Gram-negative colonization, we have proposed that LPS translocation to the bloodstream is a putative explanation for the ET observed in CF [
      • del Campo R.
      • Martinez E.
      • del Fresno C.
      • Alenda R.
      • Gomez-Pina V.
      • Fernandez-Ruiz I.
      • et al.
      Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
      ]. Herein, we have a larger cohort including a wider variety of colonizing bacteria, which has allowed us to classify patients with CF into various subgroups according to the microorganism in their sputum.
      On one hand, our results showed that only Pseudomonas aeruginosa colonization, and no other clinical parameters, exhibited a strong association with PD-L1 levels in CF. Moreover, the patients with Pseudomonas aeruginosa colonization showed a greater ET. Although the immune system effects of some intrinsic features of CF such CFTR mutations cannot be totally ruled out [
      • Tarique A.A.
      • Sly P.D.
      • Holt P.G.
      • Bosco A.
      • Ware R.S.
      • Logan J.
      • et al.
      CFTR-dependent defect in alternatively-activated macrophages in cystic fibrosis.
      ], our data indicated the relevance of the Pseudomonas aeruginosa colonization in this context. In agreement, other groups have reported altered adaptive responses in P. aeruginosa + patients mainly characterized by disrupted balance of Th17 and Th2 responses [
      • Moser C.
      • Kjaergaard S.
      • Pressler T.
      • Kharazmi A.
      • Koch C.
      • Hoiby N.
      The immune response to chronic Pseudomonas aeruginosa lung infection in cystic fibrosis patients is predominantly of the Th2 type.
      ,
      • Tiringer K.
      • Treis A.
      • Fucik P.
      • Gona M.
      • Gruber S.
      • Renner S.
      • et al.
      A Th17- and Th2-skewed cytokine profile in cystic fibrosis lungs represents a potential risk factor for Pseudomonas aeruginosa infection.
      ,
      • Kushwah R.
      • Gagnon S.
      • Sweezey N.B.
      T cell unresponsiveness in a pediatric cystic fibrosis patient: a case report.
      ,
      • Casaulta C.
      • Schoni M.H.
      • Weichel M.
      • Crameri R.
      • Jutel M.
      • Daigle I.
      • et al.
      IL-10 controls aspergillus fumigatus- and pseudomonas aeruginosa-specific T-cell response in cystic fibrosis.
      ,
      • Hector A.
      • Schafer H.
      • Poschel S.
      • Fischer A.
      • Fritzsching B.
      • Ralhan A.
      • et al.
      Regulatory T-cell impairment in cystic fibrosis patients with chronic pseudomonas infection.
      ]. On the other hand, our in vitro model reinforced the idea of LPS translocation as an explanation of the features found in patients. Monocytes from HVs stimulated with isolated P. aeruginosa LPS increased PD-L1 expression in a time-dependent manner. Therefore, sera from patients with Pseudomonas aeruginosa were shown to induce more PD-L1 expression in HV monocytes than in sera from patients without colonization; this induction was reduced when the samples were pretreated with the LPS scavenger polymyxin E. Accordingly, we detected lower levels of sPD-L1 sera from patients treated with inhaled sodium colistimethate (polymyxin E) than from those treated with other antibiotics. This last datum suggests that colistimethate could reduce the effects of LPS translocation in patients with CF who have Pseudomonas aeruginosa colonization, although a prospective, randomized, double-blind clinical trial must be performed to confirm this hypothesis.
      The results presented here support the hypothesis that translocated LPS on Pseudomonas aeruginosa-colonized patients with CF causes both PD-L1 overexpression and an ET phenotype. Moreover, the antibody-mediated blockade of PD-L1 and PD-1 in these patients reversed their impaired T cell response. Altogether, our results suggest the possibility of studying antibiotics and antiPD-1/PD-L1 antibody combination as a new potential treatment to provide clinical benefits to patients with P. aeruginosa colonization.

      Funding

      This work was supported by grants from the “Instituto de Salud Carlos III” (ISCIII), “Fondos de Investigación Sanitarias” (FIS) and FEDER [PI14/01234 and PIE15/00065] to ELC, and a grant from “Comunidad de Madrid” [PEJ15/BIO/AI-0021] to JAO.
      The following are the supplementary data related to this article.
      • Supplementary Fig. S2

        PD-L1 expression in CF patient's subgroups.

        PD-L1 expression on monocytes from CF patients and HV was measured by flow cytometry. Percentages of PD-L1+ on CD14+ gated cells from patients classified according their sex (A), type of CFTR mutation (B), age at diagnosis (C), airflow limitation (D), number of infections during last year (E) and body mass index (F) are shown (n = 32 CF patients; n = 15 HV). Airflow limitation was defined as FE1V/FVC ratio < 70. Data are expressed as the mean ± SD.

      • Supplementary Fig. S3

        PD-L1 expression on monocytes from patients with CF according to their bacterial colonization.

        PD-L1 expression on monocytes from CF patients and HV was measured by flow cytometry. Patients were classified in four groups according the presence of Staphylococcus aureus and Pseudomonas aeruginosa in their sputum. “None”, CF patients without bacteria colonization; “SA”, patients with Staphylococcus aureus but not Pseudomonas aeruginosa colonization; “PA”, patients with Pseudomonas aeruginosa but not Staphylococcus aureus colonization; “Both”, patients with both Pseudomonas aeruginosa and Staphylococcus aureus colonization. *: p < .05; ***, p < .001 and n. s., no significant using an unpaired t-test. Data are expressed as the mean ± SD.

      • Supplementary Fig. S4

        PD-L1 expression on monocytes from patients with CF correlates with endotoxin tolerance phenotype.

        Percentages of PD-L1+ cells and HLA-DR expression on gated CD14+ cells from patients with CF (n = 32) were analyzed by FACS. The PD-L1 data is basal in non-stimulated cells. HLA-DR expression data are shown as fold (ratio between mean intensity of fluorescence (MIF) on LPS stimulated cells divided by MIF on unstimulated cells). The correlation between PD-L1 and Fold of HLA-DR is shown.

      • Supplementary Fig. S5

        Basal cytokines levels on plasma from patients with CF.

        Cytokines levels on plasma from CF patients were measured. Concentrations of TNFα (A), IL-6 (B) and IL-10 (C) are shown (n = 32 patients with CF; n = 19 CF patients with Pseudomonas aeruginosa; n = 15 HVs). “+”, patients with Pseudomonas aeruginosa colonization; “-”, patients without Pseudomonas aeruginosa colonization. Data are expressed as the mean ± SD.

      • Supplementary Table S1

        Summary of clinical data in all patients included in the study. M, Male; F, Female; −, not included on medical history; MSSA, methicillin-sensitive Staphylococcus aureus; MRSA, methicillin-resistent Staphylococcus aureus.

      Conflict of interest statement

      The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

      Acknowledgements

      We thank the blood donor service from La Paz University Hospital; Aurora Muñoz for technical assistance; Ana Sierra from Cytostatic Service for her collaboration; and ServingMed.com for the editing of the manuscript.

      References

        • del Campo R.
        • Martinez E.
        • del Fresno C.
        • Alenda R.
        • Gomez-Pina V.
        • Fernandez-Ruiz I.
        • et al.
        Translocated LPS might cause endotoxin tolerance in circulating monocytes of cystic fibrosis patients.
        PLoS One. 2011; 6
        • Jain K.
        • Smyth A.R.
        Current dilemmas in antimicrobial therapy in cystic fibrosis.
        Expert Rev Respir Med. 2012; 6: 407-422
        • Salsgiver E.L.
        • Fink A.K.
        • Knapp E.A.
        • Lipuma J.J.
        • Olivier K.N.
        • Marshall B.C.
        • et al.
        Changing epidemiology of the respiratory bacteriology of patients with cystic fibrosis.
        Chest. 2016; 149: 390-400
        • Crull M.R.
        • Somayaji R.
        • Ramos K.J.
        • Caldwell E.
        • Mayer-Hamblett N.
        • Aitken M.L.
        • et al.
        Changing rates of chronic Pseudomonas aeruginosa infections in cystic fibrosis: a population-based cohort study.
        in: Clinical infectious diseases: an official publication of the infectious diseases society of america. 2018
        • Elborn J.S.
        Cystic fibrosis.
        Lancet (London, England). 2016; 388: 2519-2531
        • Gilligan P.H.
        Infections in patients with cystic fibrosis: diagnostic microbiology update.
        Clin Lab Med. 2014; 34: 197-217
        • de Dios Caballero J.
        • Vida R.
        • Cobo M.
        • Máiz L.
        • Suárez L.
        • Galeano J.
        • et al.
        Individual patterns of complexity in cystic fibrosis lung microbiota, including predator bacteria, over a 1-year period.
        MBio. 2017; 8
      1. Johansen HK, Gotzsche PC. Vaccines for preventing infection with Pseudomonas aeruginosa in cystic fibrosis. The cochrane database of systematic reviews. 2015:Cd001399.

        • Talwalkar J.S.
        • Murray T.S.
        The approach to pseudomonas aeruginosa in cystic fibrosis.
        Clin Chest Med. 2016; 37: 69-81
        • Stefani S.
        • Campana S.
        • Cariani L.
        • Carnovale V.
        • Colombo C.
        • Lleo M.M.
        • et al.
        Relevance of multidrug-resistant pseudomonas aeruginosa infections in cystic fibrosis.
        Int J Med Microbiol. 2017; 307: 353-362
        • S W.S.Y.
        • Li J.
        • Patel K.
        • Wilson J.W.
        • Dooley M.J.
        • George J.
        • et al.
        Pulmonary and systemic pharmacokinetics of inhaled and intravenous colistin methanesulfonate in cystic fibrosis patients: targeting advantage of inhalational administration.
        Antimicrob Agents Chemother. 2014; 58: 2570-2579
        • Bonfield T.
        • Chmiel J.F.
        Impaired innate immune cells in cystic fibrosis: is it really a surprise?.
        J Cyst Fibros. 2017; 16: 433-435
        • Bonfield T.L.
        • Panuska J.R.
        • Konstan M.W.
        • Hilliard K.A.
        • Hilliard J.B.
        • Ghnaim H.
        • et al.
        Inflammatory cytokines in cystic fibrosis lungs.
        Am J Respir Crit Care Med. 1995; 152: 2111-2118
        • Reynolds C.J.
        • Quigley K.
        • Cheng X.
        • Suresh A.
        • Tahir S.
        • Ahmed-Jushuf F.
        • et al.
        Lung defense through interleukin-8 carries a cost of chronic lung remodeling and impaired function.
        Am J Respir Cell Mol Biol. 2018; 59: 557-571
        • del Fresno C.
        • Gomez-Pina V.
        • Lores V.
        • Soares-Schanoski A.
        • Fernandez-Ruiz I.
        • Rojo B.
        • et al.
        Monocytes from cystic fibrosis patients are locked in an LPS tolerance state: down-regulation of TREM-1 as putative underlying mechanism.
        PLoS One. 2008; 3e2667
        • del Fresno C.
        • Garcia-Rio F.
        • Gomez-Pina V.
        • Soares-Schanoski A.
        • Fernandez-Ruiz I.
        • Jurado T.
        • et al.
        Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients.
        J Immunol. 2009; 182: 6494-6507
        • Biswas S.K.
        • Lopez-Collazo E.
        Endotoxin tolerance: new mechanisms, molecules and clinical significance.
        Trends Immunol. 2009; 30: 475-487
        • Lopez-Collazo E.
        • del Fresno C.
        Pathophysiology of endotoxin tolerance: mechanisms and clinical consequences.
        Crit Care. 2013; 17: 242
        • Topalian S.L.
        • Hodi F.S.
        • Brahmer J.R.
        • Gettinger S.N.
        • Smith D.C.
        • McDermott D.F.
        • et al.
        Safety, activity, and immune correlates of anti-PD-1 antibody in cancer.
        N Engl J Med. 2012; 366: 2443-2454
        • El Annan J.
        • Goyal S.
        • Zhang Q.
        • Freeman G.J.
        • Sharpe A.H.
        • Dana R.
        Regulation of T-cell chemotaxis by programmed death-ligand 1 (PD-L1) in dry eye-associated corneal inflammation.
        Invest Ophthalmol Vis Sci. 2010; 51: 3418-3423
        • Patil N.K.
        • Luan L.
        • Bohannon J.K.
        • Hernandez A.
        • Guo Y.
        • Sherwood E.R.
        Frontline science: anti-PD-L1 protects against infection with common bacterial pathogens after burn injury.
        J Leukoc Biol. 2018; 103: 23-33
        • Avendano-Ortiz J.
        • Maroun-Eid C.
        • Martin-Quiros A.
        • Toledano V.
        • Cubillos-Zapata C.
        • Gomez-Campelo P.
        • et al.
        PD-L1 Overexpression during endotoxin tolerance impairs the adaptive immune response in septic patients via HIF1alpha.
        J Infect Dis. 2018; 217: 393-404
        • Davis Jr., M.R.
        • Goldberg J.B.
        Purification and visualization of lipopolysaccharide from Gram-negative bacteria by hot aqueous-phenol extraction.
        J Vis Exp. 2012; (3916)
        • P M.
        • S S.
        • C D.
        • K P.
        • C S.
        • E L.
        • et al.
        Colistin reduces LPS-triggered inflammation in a human sepsis model in vivo: a randomized controlled trial.
        Clin Pharmacol Ther. 2017; 101: 773-781
        • Ralhan A.
        • Laval J.
        • Lelis F.
        • Ballbach M.
        • Grund C.
        • Hector A.
        • et al.
        Current concepts and controversies in innate immunity of cystic fibrosis lung disease.
        J Innate Immun. 2016; 8: 531-540
        • Hartl D.
        • Gaggar A.
        • Bruscia E.
        • Hector A.
        • Marcos V.
        • Jung A.
        • et al.
        Innate immunity in cystic fibrosis lung disease.
        J Cyst Fibros. 2012; 11: 363-382
      2. Reynolds CJ, Quigley K, Cheng X, Suresh A, Tahir S, Ahmed-Jushuf F, et al. Lung defense through interleukin-8 carries a cost of chronic lung remodeling and impaired function. Am J Respir Cell Mol Biol[0:null].

        • Cantin A.M.
        • Bilodeau G.
        • Larivee P.
        • Richter M.V.
        Plasma biomarkers and cystic fibrosis lung disease.
        Clin Invest Med. 2012; 35: E173-E181
        • Paats M.S.
        • Bergen I.M.
        • Bakker M.
        • Hoek R.A.
        • Nietzman-Lammering K.J.
        • Hoogsteden H.C.
        • et al.
        Cytokines in nasal lavages and plasma and their correlation with clinical parameters in cystic fibrosis.
        J Cyst Fibros. 2013; 12: 623-629
        • Ingersoll S.A.
        • Laval J.
        • Forrest O.A.
        • Preininger M.
        • Brown M.R.
        • Arafat D.
        • et al.
        Mature cystic fibrosis airway neutrophils suppress T cell function: evidence for a role of arginase 1 but not programmed death-ligand 1.
        J Immunol. 2015; 194: 5520-5528
        • Pryjma J.
        • Ernst M.
        • Fetting R.
        • Woloszyn M.
        • Zembala M.
        • Flad H.D.
        The role of monocytes in the induction and regulation of IFN-gamma production by lectin-activated human T lymphocytes.
        Eur Cytokine Netw. 1991; 2: 273-279
        • Gallart T.
        • Angel De La Fuente M.
        • Josep Barcelo J.
        • Alberola-Ila J.
        • Lozano F.
        Desialylation of T lymphocytes overcomes the monocyte dependency of pokeweed mitogen-induced T-cell activation.
        Immunology. 1997; 90: 57-65
        • Krieg C.
        • Nowicka M.
        • Guglietta S.
        • Schindler S.
        • Hartmann F.J.
        • Weber L.M.
        • et al.
        High-dimensionalsingle-cell analysis predicts response to anti-PD-1 immunotherapy.
        Nat Med. 2018; 24: 144-153
        • Tarique A.A.
        • Sly P.D.
        • Holt P.G.
        • Bosco A.
        • Ware R.S.
        • Logan J.
        • et al.
        CFTR-dependent defect in alternatively-activated macrophages in cystic fibrosis.
        J Cyst Fibros. 2017; 16: 475-482
        • Moser C.
        • Kjaergaard S.
        • Pressler T.
        • Kharazmi A.
        • Koch C.
        • Hoiby N.
        The immune response to chronic Pseudomonas aeruginosa lung infection in cystic fibrosis patients is predominantly of the Th2 type.
        APMIS. 2000; 108: 329-335
        • Tiringer K.
        • Treis A.
        • Fucik P.
        • Gona M.
        • Gruber S.
        • Renner S.
        • et al.
        A Th17- and Th2-skewed cytokine profile in cystic fibrosis lungs represents a potential risk factor for Pseudomonas aeruginosa infection.
        Am J Respir Crit Care Med. 2013; 187: 621-629
        • Kushwah R.
        • Gagnon S.
        • Sweezey N.B.
        T cell unresponsiveness in a pediatric cystic fibrosis patient: a case report.
        Allergy Asthma Clin Immunol. 2014; 10: 2
        • Casaulta C.
        • Schoni M.H.
        • Weichel M.
        • Crameri R.
        • Jutel M.
        • Daigle I.
        • et al.
        IL-10 controls aspergillus fumigatus- and pseudomonas aeruginosa-specific T-cell response in cystic fibrosis.
        Pediatr Res. 2003; 53: 313-319
        • Hector A.
        • Schafer H.
        • Poschel S.
        • Fischer A.
        • Fritzsching B.
        • Ralhan A.
        • et al.
        Regulatory T-cell impairment in cystic fibrosis patients with chronic pseudomonas infection.
        Am J Respir Crit Care Med. 2015; 191: 914-923