Skip to main content

Analysis of long term CD4+CD25highCD127- T-reg cells kinetics in peripheral blood of lung transplant recipients

Abstract

Background

The role of CD4+CD25highCD127 T-reg cells in solid-organ Transplant (Tx) acceptance has been extensively studied. In previous studies on kidney and liver recipients, peripheral T-reg cell counts were associated to graft survival, while in lung Tx, there is limited evidence for similar findings. This study aims to analyze long term peripheral kinetics of T-reg-cells in a cohort of lung recipients and tests its association to several clinical variables.

Methods

From jan 2009 to dec 2014, 137 lung Tx recipients were submitted to an immunological follow up (median: 105.9 months (6.7–310.5)). Immunological follow up consisted of a complete blood peripheral immuno-phenotype, inclusive of CD4+CD25highCD127 T and FOXP3+ cells. We tested the association between T-reg and relevant variables by linear OR regression models for repeated measures, adjusting for time from Tx. Also, by ordered logistic models for panel data, the association between Chronic Lung Allograft Dysfuncton (CLAD) onset/progression and T-reg counts in the previous 3 months was tested.

Results

Among all variables analyzed at multivariate analysis: Bronchiolitis Obliterans Syndrome (OR −6.51, p < 0.001), Restrictive Allograft Syndrome (OR −5.19, p = 0.04) and Extracorporeal photopheresis (OR −5.65, p < 0.001) were significantly associated to T-reg cell. T-reg cell counts progressively decreased according to the severity of CLAD. Furthermore, patients with higher mean T-reg counts in a trimester had a significantly lower risk (OR 0.97, p = 0.012) of presenting CLAD or progressing in the graft dysfunction in the following trimester.

Conclusions

Our present data confirm animal observations on the possible role of T-reg in the evolution of CLAD.

Peer Review reports

Background

Lung transplant (Tx) is the only therapy for patients with end-stage respiratory failure, but despite the use of potent immunosuppressive protocols, the long-term survival of lung graft is hampered by the occurrence of chronic lung allograft dysfunction that occurs in nearly 50% of patients by the 5th post-Tx year [1]. According to recent classification, there are two major clinical phenotypes of chronic irreversible allograft dysfunction (CLAD): the so called obstructive form: bronchiolitis obliterans syndrome (BOS), (nearly 70% of cases) [2], and the newly described restrictive allograft syndrome (RAS) [3, 4]. CLAD is considered a complex multifactorial process [5], which leads to chronic airway inflammation and tissue injuries and ultimates in a fibro-reparative response. This can involve either the airway lumen in BOS, or the interstitial and airways in RAS [6]. Although recent evidence ascribes an important pathogenic role in CLAD to aspecific inflammatory mechanisms [7,8,9,10], the importance of specific immunity can’t be denied. Several previous studies support a crucial role for allo and auto-reactive T cells in BOS pathogenesis [11,12,13,14]. T cell response as well as the production of allo or auto-antibodies have been described, and are dependent on the migration of antigen-presenting cells in secondary lymphoid organs and on the direct stimulation of T cells within graft [15, 16]. All of this experimental and clinical evidence strongly points out the difficulties in achieving immunologic tolerance, either central or peripheral, of lung graft [17,18,19].

Since 1995, when Sakaguchi described a CD4+CD25+T cell subset displaying regulatory properties, several subsets of T-reg cells have been described and classified as natural T-reg, thymus-derived and naturally committed to immunoregulation, and inducible T-reg, generated in the periphery during immune response. So far the best characterised T-reg population is the thymus-derived CD4 subset, constitutively expressing CD25 (at a high rate) and FoxP3, a transcription regulator which controls the maturation and function of T-reg cells. Although a specific and exclusive marker of T-reg activity has not yet been identified, the diminished surface expression of CD127, as well as the expression of CD39 or CD152, are considered characteristic of CD4+CD25+ T-reg cells. [20, 21]These molecules can be expressed, even if transiently, as Foxp3, by other cell types including recently activated effector T cells. However, experimental evidence suggests that most of the Foxp3+ T-reg cells are within the CD4+CD25highCD127 cell population, so that sorting of this subset is actually considered the best method for the isolation and in vitro amplification of these cells [22].

The role of CD4+CD25highCD127 T-reg cells in Tx acceptance has been studied in experimental models and also in clinical settings. Animal studies demonstrated that CD4+CD25+ Foxp3+ T-reg cells are expanded in tolerized animals, that tolerance can be infectiously transferred to naïve animals, and appears to be a specific and localized phenomenon. These data have been partially confirmed in kidney and liver Tx recipients, showing a positive correlation between graft survival and the number of circulating CD4+CD25+ T-reg cells, as well as a correlation between their peripheral fluctuation and the occurrence of acute and chronic rejection [18, 23, 24].

As for lung Tx, evidence is limited and somewhat contradictory [25]. In previous cross- sectional studies, we showed that lung Tx recipients with BOS had significantly lower peripheral CD4+CD25high T-reg cells than clinically stable lung recipients, and demonstrated their functional regulatory profile, in vitro [26, 27]but subsequesnt studies failed to demonstrate a correlation between T-reg cell counts and long term lung Tx outcome [28,29,30].

To date, however, little is known about the long term evolution of peripheral CD4+CD25highCD127 regulatory T cells in lung Tx. The present study aims at analyzing their long-term kinetic on a cohort of lung recipients and at testing its association with several clinical and pharmacological variables (CLAD, treatment, infections, kidney dysfunction or neoplasia).

Methods

Patients

In our Center, LTx has been performed since 1991. The study was conducted from 1st January 2009 to 31st December 2014. During this period 137 lung recipients entered the immunological follow-up, with a median follow-up of 105.9 months (range 6,7–310,5). All patients were submitted to the assessment of a complete peripheral immune phenotype at least twice a year.

Our immune suppression protocol has undergone some changes over time. No patients underwent induction treatment at time of transplantation. All patients transplanted between 2001 and 2007 were treated with a triple immunosuppressive regimen including cyclosporine, azathioprine, and prednisone, whereas patients transplanted since January 2007 received a modified standard triple regimen, with tacrolimus, mycophenolate mofetil, and prednisone. In case of refractory acute rejection (AR), patients were switched from cyclosporine to tacrolimus and from azathioprine to mycophenolate mofetil. In the presence of documented renal dysfunction, patients were treated with low- dose tacrolimus plus everolimus.

All patients underwent surveillance and on-need bronchoscopies; biopsy-proven episodes of AR following criteria [31] were treated with steroid boluses and, in case of AR recurrence or persistence, with a standard anti-thymoglobulin course and a modulation of the IS regimen. Our surveillance protocol has been reported in previous studies [32]. BOS diagnosis and severity grades has been assessed according to published guidelines [6, 33,34,35]. The CLAD subtype RAS has been retrospectively re-classified for patients diagnosed before 2013, according to radiological (CT scan showing a pattern of persistent interstitial/upper lobe fibrosis) and functional criteria (persistent decline in forced expiratory volume in 1 s (FEV 1) of >20% compared to the best post Tx value and a decline in total lung capacity of >10% compared to baseline) [33,34,35]. In case of a BOS 0p or early RAS diagnosis, patients were prescribed a 3-month course of chronic low-dose azithromycin. At the same time, patients underwent gastro-esophageal reflux assessment and maximization of anti-reflux medical treatment. In case of a further decline consistent with a CLAD diagnosis, since 2003, patients are referred to the Apheresis Unit for compassionate ECP treatment [36]. Severity of CLAD has been graduated according to the degree of functional impairment as described for BOS [6, 33,34,35]. Our cytomegalovirus surveillance protocol has been detailed in previous studies [37].

The ethics review committee of the IRCCS Policlinico San Matteo of Pavia approved the research n° ICS 30.4/RF00.65.

Flow cytometric determination of peripheral lymphocyte subsets

Flow cytometry was performed on a Beckman Coulter Navios using Kaluza software (Beckman Coulter). Briefly, 50 μl of fresh whole blood was incubated with the appropriate amounts of fluorochrome-labeled monoclonal antibodies CD45 APC Alexa Fluor 750 (Allophycocyanin-Alexa Fluor 750; clone J33), CD4 APC (Allophycocyanin; clone 13B8.2), CD69ECD (R Phycoerythrin-Texas Red; clone TP1.55.3), CD25 PE (R Phycoerythrin; clone B1.49.9), CD127 PC5 (R Phycoerythrin-Cyanine 5.1; clone R34.34) for the T-reg and CD45 APC Alexa Fluor 750, CD4 APC, CD3 FITC (Fluorescein isothiocyanate; clone UCHT1), CD8 PE (R Phycoerythrin; clone B9.11), CD56 PC5 (; clone N901), CD16 PC5 (PC5.1; clone 3G8), CD19 PC7 (PC 7; clone J3–119) for the lymphocytic population (Beckman Coulter) at room temperature in the dark for 15 min using appropriate mouse immunoglobulin isotypes as a control. Following incubation, 1 ml erythrocyte lysing solution (VersaLyse, Beckman Coulter) was added to the samples and incubated under the same conditions for 20 min. In some samples, peripheral blood mononuclear cells (PBMC) were stained with CD4 APC, CD25 FITC (clone B1.49.9), and CD127 Alexa Fluor 647 (clone HIL-7R-M21) (BD Pharmingen), fixed and permeabilized, followed by intracellular staining with Foxp3 PE (clone PCH101) or control IgG1 (Human Regulatory T cell Staining kit, eBioscience) for 30 min. Finally, the cells were characterized by flow cytometry analysis.

T-reg cell analysis

Peripheral blood assessment of regulatory T cell subsets has significantly evolved in the last years, even more so during the realization of this study. For this reason, we started the protocol performing the analysis of peripheral CD4+CD25highCD127 cell subset, and later on we also started to quantify FOXP3+ cells.

Therefore, we could determine that peripheral CD4+CD25highCD127 T-reg cell subset included a mean of 93.15% (±4.34) FOXP3+ cells (Fig. 1). We also found a high significant correlation between peripheral CD4+CD25highCD127 T-reg cell counts and FOXP3+ cell counts over the whole cohort. For this reason, and in accordance with published evidence [20] we included CD4+CD25highCD127 cell determinations in the final statistical analysis, expressed this subset as absolute number (n°/μl peripheral blood) and named these cells approvedCD4+CD25highCD127 T-reg cells. Analysis performed with CD4+CD25highCD127 T-reg cells expressed as percentage of the whole CD4 subset, gave analogous results.

Fig. 1
figure 1

a. Representative dot plot of intracellular staining Foxp3 in CD4 + CD25highCD127 - T-reg cell.b. Representative dot plot of intracellular staining Foxp3 in CD4 + CD25-CD127pos (0%) T-reg cell. c. Representative dot plot of isotope control

Statistical analysis

Descriptive statistics were produced for demographic, clinical and laboratory characteristics of cases. Mean and standard deviation (SD) are shown for normally distributed variables, and median and interquartile range (IQR) for non-normally distributed variables, numbers and percentages for categorical variables.

The association between T-reg and a number of predictors was explored by means of bivariate and multivariate linear mixed models, with patient and time since Tx as random effects and predictors and time (also) as fixed effects. Clinical and Immunological variables that were included in the analysis are shown in Table 1.

Table 1 demographic and clinical features of the patients enrolled in this study

To assess whether CD4+CD25highCD127 T-reg cell count was associated to CLAD occurrence or higher CLAD grade in the subsequent 3 months, we fit ordered logistic models for panel data. When a dependent variable has more than two categories and the values of each category have a meaningful sequential order where a value is indeed ‘higher’ than the previous one, then we can use ordinal logistic regression. In this instance, we are comparing patients in CLAD grade > k versus ≤k. The interpretation is: for a one unit change in the predictor variable (e.g. T-reg cell count in previous trimester), likelihood of having CLAD grade > k instead of ≤k is an OR times higher.

In all cases, tests were two-tailed, and the p-value cut-off for significance was set at 0.05.

Stata computer software version 14.0 (Stata Corporation, 4905 Lakeway Drive, College Station, Texas 77,845, USA) was used for statistical analysis.

Results

Patients

Overall 137 patients were included in this retrospective study and followed-up for a median of 105.9 months (6.7–310.5). Demographics and clinical features of included patients are listed in Table 1, including gender, age at Tx, Tx indication, type of Tx, length of follow-up and type of immunosuppressive drugs used. Some patients (27%) were enrolled in the immunological follow- up at time of Tx, while the others entered the study later in the FU period (median follow-up months at first determination in the latter group: 82,4 months, range 14,0–275,7) this, as stated above, was considered in the statistical analysis.

Being a prospective immunological FU, the overall number of included samples is high: n° 1943 with a median of 14 sample/patient.

Variables associated to T-reg cell counts

Results of bivariate and multivariate analysis are shown in Table 2. As for acute rejection, the limited number of samples obtained during an episode of acute cellular (Agrade ≥2, any B grade) or humoral rejection (globally < than 15) did not allow us to assess any statistical association with peripheral T-reg cell number. All tested immunological variables (CD3+,CD4+, CD8+, CD19+ and CD16 + CD56+ cells) resulted positively correlated associated to T-reg (Table 2) at the initial bivariate analysis. Among clinical variables only the presence of CLAD, treatment with azathioprine and ECP were significantly associated to peripheral CD4+CD25highCD127 T-reg cell counts. Given the lack of association with T-reg cell counts with the majority of tested variables only azathioprine, ECP, and CLAD, as well as BOS or RAS and CLAD severity grades were included in the multivariate model. At multivariate analysis, only the association with azathioprine did not retain its significance (Table 2). Furthermore, when BOS severity grade was considered, a significant progression of CD4+CD25highCD127 T-reg cell decline was observed (Fig. 2). Of note, no difference was detectable between the 2 CLAD phenotypes, BOS and RAS (Fig. 3). The significant negative association of T-reg cell counts with ECP was confounded on the presence and severity of CLAD, and thus was not considered clinically relevant.

Table 2 bi- and multi-variate linear regression analysis per CD4+CD25highCD127
Fig. 2
figure 2

Relation between T-reg cells count and BOS grade (calculated according to BOS severity classification [30]). Figure is purely descriptive. Median, IQR and min/max are depicted. *p < 0.001; §p = 0.01

Fig. 3
figure 3

Relation between T-reg cells count and stable, BOS (p < 0.001) or RAS (p = 0.01) patients’ (BOS was diagnosed according to published guidelines [6, 30,31,32]. RAS has been retrospectively re-classified according to radiological and functional criteria [30,31,32]).). Figure is purely descriptive. Median, IQR and min/max are depicted

Prediction of CLAD

In patients with higher mean peripheral CD4+CD25highCD127 T-reg counts, the risk of presenting CLAD or progressing in the graft dysfunction in the subsequent trimester was significantly lower (OR 0.97, 95%CI 0.95–0.99, p = 0.012). Variables were included in the model if clinically relevant or statistically significant (at the 0.05 level) at bivariate analysis, without further selection (Table 3).

Table 3 ordered logistic regression analysis for association between CLAD grade and a number of predictors, including T-reg in the previous trimester

Discussion

On the basis of our results we can confirm and extend our previous observation on the role of CD4 + CD25high T-reg cells in lung graft acceptance/rejection [27]. To our knowledge, this is the first longitudinal study reporting the long-term kinetics of peripheral CD4+CD25highCD127 T-reg cells in lung recipients, and the first observation, that variation of peripheral T-reg cell counts can predict CLAD onset/progression.

Although, during the study period, a methodological evolution in the identification of T-reg cells subsets occurred, we clearly demonstrated that CD4+CD25highCD127 subset is significantly enriched with FOXP3+ cells, thus inferring that it is endowed with regulatory functions. Based on these observations, we have referred to these cells as CD4+CD25highCD127 T-reg cells.

Experimental evidence in animals have strongly suggested that lung graft acceptance is associated to an intra-graft [16, 38] and peripheral [39] T-reg cell expansion and that adoptive transfer of “in vitro” generated and expanded T-reg cells attenuates airway obliteration in a rat OB model [40].

Conversely, human studies have provided, to date, sub-optimal evidence on the protective role of T-reg cells towards chronic graft rejection. Most studies, in fact, have explored peripheral CD4 + CD25+ T-reg cell counts early after Tx, suggesting that a low number of these effectors might predict subclinical or clinical acute kidney rejection [41, 42], and can be significantly affected by the type of immunosuppressive regimen [43, 44]. A few studies are available on lung Tx recipients with conflicting results. Neujahr and colleagues found higher BAL CD4 + CD25high cell counts in patients with biopsy-proven AR, but failed to find a correlation with peripheral blood findings [45]. On the contrary, lower CD4 + CD25high T cell counts were detected both at peripheral and BAL level of BOS patients with respect to stable lung recipients [27, 46].Moreover, an increase in BAL CCR7(+) T-reg cell percentage was found to correlate to a reduced risk of BOS by Gregson and colleagues [47]. Krustrup and colleagues, analyzing CD4 + CD25 + FOXP3+ cell at tissue level, found higher frequencies during acute rejection episodes [29], but failed to demonstrate an association between these cells and the risk of CLAD occurrence [30]. We believe that prediction studies on peripheral T-reg in Tx recipients are invalidated by the high fluctuation of blood-T-reg cell in time, possibly in relation to a number of confounding variables including drugs, infectious complications, neoplasia, kidney failure and ECP treatment.

Thanks to the large number of determinations that we included in this long term study, we took into account this variability in the analysis, thus we succeeded in demonstrating that: 1) peripheral counts of CD4+CD25highCD127 T-reg cells significantly decrease in CLAD patients; 2) the degree of their decrease is associated with the severity of BOS and, most noteworthy; 3) CLAD onset or GRADE is significantly associated to mean T-reg cell counts in the previous trimester.

Finally, we also tested the association of CD4+CD25highCD127 T-reg cell counts with a number of clinical variables, including also infections and different immunosuppressive drugs. Interestingly, only azathioprine and ECP were found to be significantly associated to them at the univariate analysis, while, in the multivariate model, only the ECP effect was significant. However, the ECP effect was confounded on CLAD and CLAD severity, and is therefore to be considered irrelevant from the clinical point of view.

Interestingly, unlike previous evidence in literature [26, 48]we could not detect any significant variation of T-reg cell counts with respect to other specific immunosuppressive drugs such as cyclosporine A, tacrolimus or everolimus. The effect of maintenance drugs on T-reg cell counts has been poorly evaluated in humans, the role of CsA and tacrolimus has been studied in mice with controversial results [44] while everolimus has been shown to enhance the number of peripheral T-reg cells in liver recipients following a tacrolimus to everolimus conversion [48]. However, it was recently observed that sirolimus, another mTor inhibitor, did not expand peripheral T-reg cells in a cohort of de novo kidney recipients [49], in analogy to our present observation. Finally, in the present study we could not assess the possible role of lympho-depleting strategies, since induction treatment is not performed at our center.

We must acknowledge a number of limitations. First, although larger than other cohorts, our sample is not sufficiently large to identify small associations, especially in some subgroups. Second, patients started FU at different times from Tx; although this was taken into account in the analysis, we cannot be certain that bias would not impact on results. Third, although data were collected prospectively, this study was designed retrospectively as an exploratory analysis; therefore, patients were sampled at irregularly spaced times, and sometimes upon clinical basis, so there might be unobserved variables that might have affected results. In addition, we performed all the analysis using both the absolute number and the percentage of T-reg in peripheral blood, but, although both had the same trends over time and were significantly correlated with CLAD, we decided to show only the results concerning absolute number analysis because they had the best correlation with the analyzed variables. Finally, a relevant issue, from the biological point of view, is that graft tolerance is a results of a balance between regulatory and effector clones. In this study we couldn’t take into account the weight of allospecific effectors at different time points.

Conclusion

In conclusion, given all the above limitations, we could find an association between T-reg cell counts and CLAD onset/progression, including both BOS and RAS phenotype, thus confirming animal observations on the protective role of T-reg cells with respect to CLAD.

Abbreviations

AR:

Acute rejection

BAL:

Bronchoalveolar lavage

BOS:

Bronchiolitis obliterans syndrome

CLAD:

Chronic lung allograft dysfunction

CsA:

Cyclosporine A

CT:

Computed tomography

ECP:

Extracorporeal photopheresis

FEV1:

Forced expiratory volume in 1 second

FU:

Follow-up

GER:

Gastro esophageal reflux

IS:

Immunosuppressant

LTx:

Lung transplant

RAS:

Restrictive allograft syndrome

T-reg:

T regulatory cells

Tx:

Transplant

References

  1. Yusen RD, Edwards LB, Kucheryavaya AY, et al. International Society for Heart and Lung Transplantation. The registry of the International Society for Heart and Lung Transplantation: thirty-first adult lung and heart-lung transplant report--2014; focus theme: retransplantation. J Heart Lung Transplant. 2014;33(10):1009–24.

    Article  PubMed  Google Scholar 

  2. Sato M, Ohmori-Matsuda K, Saito T, et al. Time-dependent changes in the risk of death in pure bronchiolitis obliterans syndrome (BOS). J Heart Lung Transplant. 2013;32(5):484–91.

    Article  PubMed  Google Scholar 

  3. Sato M, Hwang DM, Waddell TK, Singer LG, Keshavjee S. Progression pattern of restrictive allograft syndrome after lung transplantation. J Heart Lung Transplant. 2013;32(1):23–30.

    Article  PubMed  Google Scholar 

  4. Ofek E, Sato M, Saito T, et al. Restrictive allograft syndrome post lung transplantation is characterized by pleuroparenchymal fibroelastosis. Mod Pathol. 2013;26(3):350–6.

    Article  CAS  PubMed  Google Scholar 

  5. Weigt SS, DerHovanessian A, Wallace WD, Lynch JP 3rd, Belperio JA. Bronchiolitis obliterans syndrome: the Achilles’ heel of lung transplantation. Semin Respir Crit Care Med. 2013;34(3):336–51.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Verleden SE, Vasilescu DM, McDonough JE, et al. Linking clinical phenotypes of chronic lung allograft dysfunction to changes in lung structure. Eur Respir J. 2015;46(5):1430–9.

  7. Vanaudenaerde BM, De Vleeschauwer SI, Vos R, et al. The role of the IL23/IL17 axis in bronchiolitis obliterans syndrome after lung transplantation. Am J Transplant. 2008;8(9):1911–20.

    Article  CAS  PubMed  Google Scholar 

  8. Evers A, Atanasova S, Fuchs-Moll G, et al. Adaptive and innate immune responses in a rat orthotopic lung transplant model of chronic lung allograft dysfunction. Transpl Int. 2015;28(1):95–107.

    Article  CAS  PubMed  Google Scholar 

  9. Kreisel D, Goldstein DR. Innate immunity and organ transplantation: focus on lung transplantation. Transpl Int. 2013;26(1):2–10.

    Article  CAS  PubMed  Google Scholar 

  10. Verleden SE, Ruttens D, Vos R, et al. Differential cytokine, chemokine and growth factor expression in phenotypes of chronic lung allograft dysfunction. Transplantation. 2015;99(1):86–93.

    Article  CAS  PubMed  Google Scholar 

  11. Grossman EJ, Shilling RA. Bronchiolitis obliterans in lung transplantation: the good, the bad, and the future. Transl Res. 2009;153(4):153–65.

    Article  PubMed  Google Scholar 

  12. Snell GI, Westall GP, Paraskeva MA. Immunosuppression and allograft rejection following lung transplantation: evidence to date. Drugs. 2013;73(16):1793–813.

    Article  CAS  PubMed  Google Scholar 

  13. Angaswamy N, Tiriveedhi V, Sarma NJ, et al. Interplay between immune responses to HLA and non-HLA self-antigens in allograft rejection. Hum Immunol. 2013;74(11):1478–85.

    Article  CAS  PubMed  Google Scholar 

  14. Bharat A, Mohanakumar T. Autoimmunity and lung transplantation. Front Biosci (Elite Ed). 2012;4:2378–88. Review

    Article  Google Scholar 

  15. Gracon AS, Wilkes DS. Lung transplantation: chronic allograft dysfunction and establishing immune tolerance. Hum Immunol. 2014;75(8):887–94.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Li W, Bribriesco AC, Nava RG, et al. Lung transplant acceptance is facilitated by early events in the graft and is associated with lymphoid neogenesis. Mucosal Immunol. 2012;5(5):544–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Shalev I, Selzner N, Shyu W, Grant D, Levy G. Role of regulatory T cells in the promotion of transplant tolerance. Liver Transpl. 2012;18(7):761–70.

    Article  PubMed  Google Scholar 

  18. Issa F, Robb RJ, Wood KJ. The where and when of T cell regulation in transplantation. Trends Immunol. 2013;34(3):107–13.

    Article  CAS  PubMed  Google Scholar 

  19. Issa F, Wood KJ. Translating tolerogenic therapies to the clinic - where do we stand? Front Immunol. 2012;3:254.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mandapathil M, Lang S, Gorelik E, Whiteside TL. Isolation of functional human regulatory T cells (Treg) from the peripheral blood based on the CD39 expression. J Immunol Methods. 2009;346:55–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jiang H, Chess L. An integrated view of suppressor T cell subsets in immunoregulation. J Clin Invest. 2004;114:1198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhang H, Guo H, Lu L, et al. Sequential monitoring and stability of ex vivo-expanded autologous and nonautologous regulatory T cells following infusion in nonhuman primates. Am J Transplant. 2015;15(5):1253–66.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Segundo DS, Ruiz JC, Izquierdo M, et al. Calcineurin inhibitors, but not rapamycin, reduce percentages of CD4+CD25+FOXP3+ regulatory T cells in renal transplant recipients. Transplantation. 2006;82(4):550–7.

    Article  PubMed  Google Scholar 

  24. Cretney E, Kallies A, Nutt SL. Differentiation and function of Foxp3(+) effector regulatory T cells. Trends Immunol. 2013;34(2):74–80.

    Article  CAS  PubMed  Google Scholar 

  25. Neujahr DC. Intragraft regulatory T cells and lung transplant outcomes--we are still at square one. Clin Transpl. 2015;29(3):177–8.

    Article  Google Scholar 

  26. Meloni F, Morosini M, Solari N, et al. Peripheral CD4+ CD25+ T-reg cell expansion in lung transplant recipients is not affected by calcineurin inhibitors. Int Immunopharmacol. 2006;6(13–14):2002–10.

    Article  CAS  PubMed  Google Scholar 

  27. Meloni F, Vitulo P, Bianco AM, Paschetto E. Regulatory CD4+CD25+ T cells in the peripheral blood of lung transplant recipients: correlation with transplant outcome. Transplantation. 2004;77(5):762–6.

    Article  PubMed  Google Scholar 

  28. Nakagiri T, Warnecke G, Avsar M, et al. Lung function early after lung transplantation is correlated with the frequency of regulatory T cells. Surg Today. 2012;42(3):250–8.

    Article  PubMed  Google Scholar 

  29. Krustrup D, Iversen M, Martinussen T, Andersen CB. Time elapsed after transplantation influences the relationship between the number of regulatory T cells in lung allograft biopsies and subsequent acute rejection episodes. Transpl Immunol. 2014;31(1):42–7.

    Article  CAS  PubMed  Google Scholar 

  30. Krustrup D, Madsen CB, Iversen M, Engelholm L, Ryder LP, Andersen CB. The number of regulatory T cells in transbronchial lung allograft biopsies is related to FoxP3 mRNA levels in bronchoalveolar lavage fluid and to the degree of acute cellular rejection. Transpl Immunol. 2013;29(1–4):71–5.

    Article  CAS  PubMed  Google Scholar 

  31. Stewart S, Fishbein MC, Snell GI, et al. Revision of the1996 working formulation for the standardization of nomenclature in the diagnosis of lung rejection. J Heart Lung Transplant. 2007;26:1229–42.

    Article  PubMed  Google Scholar 

  32. Meloni F, Solari N, Miserere S, et al. Chemokine redundancy in BOS pathogenesis. A possible role also for the CC chemokines: MIP3-beta, MIP3-alpha, MDC and their specific receptors. Transpl Immunol. 2008;18:275–80.

    Article  CAS  PubMed  Google Scholar 

  33. Estenne M, Hertz MI. Bronchiolitis obliterans after human lung transplantation. Am J Respir Crit Care Med. 2002;166:440–4.

    Article  PubMed  Google Scholar 

  34. Sato M, Waddell TK, Wagnetz U, et al. Restrictive allograft syndrome (RAS): a novel form of chronic lung allograft dysfunction. J Heart Lung Transplant. 2011;30:735–42.

    Article  PubMed  Google Scholar 

  35. Verleden GM, Vos R, Vanaudenaerde B, et al. Current views on chronic rejection after lung transplantation. Transpl Int. 2015;28(10):1131–9.

  36. Del Fante C, Scudeller L, Oggionni T, et al. Long-term off-line extracorporeal Photochemotherapy in patients with chronic lung allograft rejection not responsive to conventional treatment: a 10-year single-Centre analysis. Respiration. 2015;90(2):118–28.

    Article  PubMed  Google Scholar 

  37. Lilleri D, Gerna G, Bruno F, et al. Systemic and local human cytomegalovirus-specific T-cell response in lung transplant recipients. New Microbiol. 2013;36:267–77.

    CAS  PubMed  Google Scholar 

  38. Sommer W, Knöfel AK, Madrahimov N, et al. Allogeneic CD4+CD25high T cells regulate obliterative bronchiolitis of heterotopic bronchus allografts in both porcinized and humanized mouse models. Transplantation. 2015;99(3):482–91.

  39. Shi Q, Niu Y, Cao H, et al. CD28 superagonist antibody treatment attenuated obliterative bronchiolitis in rat allo-orthotopic tracheal transplantation by preferentially expanding Foxp3-expressing regulatory T cells. Transplant Proc. 2012;44(4):1060–6.

    Article  CAS  PubMed  Google Scholar 

  40. Shi Q, Cao H, Liu J, et al. CD4+ Foxp3+ regulatory T cells induced by TGF-β, IL-2 and all-trans retinoic acid attenuate obliterative bronchiolitis in rat trachea transplantation. Int Immunopharmacol. 2011;11(11):1887–94.

    Article  CAS  PubMed  Google Scholar 

  41. Kim SH, Oh EJ, Ghee JY, et al. Clinical significance of monitoring circulating CD4+CD25+ regulatory T cells in kidney transplantation during the early posttransplant period. J Korean Med Sci. 2009;24(Suppl):S135–42.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Giaretta F, Bussolino S, Beltramo S, et al. Different regulatory and cytotoxic CD4+ T lymphocyte profiles in renal transplants with antibody-mediated chronic rejection or long-term good graft function. Transpl Immunol. 2013;28(1):48–56.

    Article  CAS  PubMed  Google Scholar 

  43. Krystufkova E, Sekerkova A, Striz I, Brabcova I, Girmanova E, Viklicky O. Regulatory T cells in kidney transplant recipients: the effect of induction immunosuppression therapy. Nephrol Dial Transplant. 2012;27(6):2576–82.

    Article  CAS  PubMed  Google Scholar 

  44. Wang XJ, Leveson-Gower D, Golab K, et al. Influence of pharmacological immunomodulatory agents on CD4(+)CD25(high)FoxP3(+) T regulatory cells in humans. Int Immunopharmacol. 2013;16(3):364–70.

    Article  CAS  PubMed  Google Scholar 

  45. Neujahr DC, Cardona AC, Ulukpo O, et al. Dynamics of human regulatory T cells in lung lavages of lung transplant recipients. Transplantation. 2009;88(4):521–7.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Bhorade SM, Chen H, Molinero L, et al. Decreased percentage of CD4+FoxP3+ cells in bronchoalveolar lavage from lung transplant recipients correlates with development of bronchiolitis obliterans syndrome. Transplantation. 2010;90(5):540–6.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Gregson AL, Hoji A, Palchevskiy V, et al. Protection against bronchiolitis obliterans syndrome is associated with allograft CCR7+ CD45RA- T regulatory cells. PLoS One. 2010;5(6):e11354.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Levitsky J, Mathew JM, Abecassis M, et al. Systemic immunoregulatory and proteogenomic effects of tacrolimus to sirolimus conversion in liver transplant recipients. Hepatology. 2013;57(1):239–48.

    Article  CAS  PubMed  Google Scholar 

  49. Libetta C, Esposito P, Gregorini M, et al. Sirolimus vs cyclosporine after induction with basiliximab does not promote regulatory T-cell expansion in de novo kidney transplantation: results from a single-center randomized trial. Transpl Immunol. 2015;33(2):117–24.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

- The authors thank Dr. Katherine O’Donohoe for careful revision of the manuscript.

- Gruppo Comunale AIDO “Luigi Stabile” Castegnato for the ​economical support.

Funding

Not applicable.

Availability of data and materials

The dataset used and analyzed during the current study available from the corresponding author on reasonable request.

Author information

Authors and Affiliations

Authors

Contributions

D.P. participated in the writing of the paper, participated in the performance of the research and in performing clinical follow-up, participated in data analysis. M.M. participated in research design, participated in the writing of the paper, participated in the performance of the research, participated in data analysis, contributed new reagents or analytic tools. S.M. participated in the performance of the research, contributed new reagents or analytic tools. A.B. participated in the performance of the research, contributed new reagents or analytic tools. L.S. participated in the writing of the paper, participated in data analysis. E.C. participated in the performance of the research, contributed new reagents or analytic tools. T.O. participated in the performance of the research and in performing clinical follow-up. G.S. participated in the performance of the research and in performing clinical follow-up. C.T. participated in data analysis. F.A. participated in performing surgical transplant and clinical follow-up. A.M.D’A. chief of surgical unit, participated in performing surgical transplant. F.M. Participated in research design, participated in the writing of the paper, participated in the performance of the research and in performing clinical follow-up, participated in data analysis. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Davide Piloni.

Ethics declarations

Ethics approval and consent to participate

The ethics review committee of the IRCCS Policlinico San Matteo of Pavia approved the research n° ICS 30.4/RF00.65.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Piloni, D., Morosini, M., Magni, S. et al. Analysis of long term CD4+CD25highCD127- T-reg cells kinetics in peripheral blood of lung transplant recipients. BMC Pulm Med 17, 102 (2017). https://doi.org/10.1186/s12890-017-0446-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12890-017-0446-y

Keywords