Next Article in Journal
Correction: Kędzierska, H., et al. Decreased Expression of SRSF2 Splicing Factor Inhibits Apoptotic Pathways in Renal Cancer. Int. J. Mol. Sci. 2016, 17, 1598
Next Article in Special Issue
Essential Oils as Antiviral Agents, Potential of Essential Oils to Treat SARS-CoV-2 Infection: An In-Silico Investigation
Previous Article in Journal
Chitosan Modified Zeolite Molecular Sieve Particles as a Filter for Ammonium Nitrogen Removal from Water
Previous Article in Special Issue
Less Cytotoxic Protoflavones as Antiviral Agents: Protoapigenone 1′-O-isopropyl ether Shows Improved Selectivity Against the Epstein–Barr Virus Lytic Cycle
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

ThymicPeptides Reverse Immune Exhaustion in Patients with Reactivated Human Alphaherpesvirus1 Infections

1
Department of Otolaryngology and Laryngeal Oncology, Medical University of Lublin, 20-954 Lublin, Poland
2
Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland
3
Department of Immunology, St. John’s Cancer Centre, Lublin 20-090, Poland
4
Department of Pediatric Endocrinology and Diabetology, Medical University, 20-093 Lublin, Poland
5
Department of Pharmaceutical Microbiology, Medical University, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(7), 2379; https://doi.org/10.3390/ijms21072379
Submission received: 22 February 2020 / Revised: 11 March 2020 / Accepted: 29 March 2020 / Published: 30 March 2020
(This article belongs to the Special Issue Natural Products against Viral Infections II)

Abstract

:
Recurrent infection with human alphaherpesvirus 1 (HHV-1) may be associated with immune exhaustion that impairs virus elimination. Thymic peptides enhance immune function and thus could overcome immune exhaustion. In this study, we investigated whether reactivation of herpes infections was associated with immune exhaustion. Moreover, we examined the impact of treatment with thymostimulin on the expression of programmed cell death protein 1 (PD-1) and its ligand (PD-L1) on T and B lymphocytes in patients suffering from recurrent HHV-1 reactivation. We also assessed the effector function of peripheral blood mononuclear cells (PBMCs) after stimulation with thymic peptides. We enrolled 50 women with reactivated HHV-1 infections and healthy volunteers. We measured the expression of various activation and exhaustion markers on the surface of PBMCs using flow cytometry. In ex vivo experiments, we measured the secretion of inflammatory cytokines by PBMCs cultured with thymostimulin. Compared with controls, patients with reactivated HHV-1 infections had increased percentages of CD3+ co-expressing CD25, an activation marker (p < 0.001). Moreover, these patients had increased percentages of CD4+ and CD8+ cells co-expressing the inhibitory markers PD-1 and PD-L1. In cultures of PBMCs from the patients, thymostimulin increased the secretion of interferon gamma (p < 0.001) and interleukin (IL)-2 (p = 0.023), but not IL-4 or IL-10.Two-month thymostimulin therapy resulted in no reactivation of HHV-1 infection during this period and the reduction of PD-1 and PD-L1 expression on the surface of T and B lymphocytes (p < 0.001). In conclusion, reactivation of herpes infection is associated with immune exhaustion, which could be reversed by treatment with thymic peptides.

1. Introduction

Human alphaherpesvirus 1 (HHV-1) is a common pathogen; nearly 80% of people aged 22–30 years have anti-HHV-1 antibodies, and the incidence increases with age [1,2]. Mucocutaneous lesions are the most common symptom of HHV-1 infections [3]. After the first infection, HHV-1 persists in neurons, and the infection can recur with or without symptoms [4,5]. Immunocompetent people combat HHV-1 infections without antiviral medications, whereas those with impaired immunity can have severe infections despite treatment [6].
Viral infections affect the immune function. T cells launch antiviral responses during acute infections, whereas chronic infection can cause T-cell exhaustion [7]. Exhausted T cells display a reduced production of antiviral cytokines, decreased cytotoxicity, and an impaired immediate response to previously encountered viral antigens [7].
Antigen stimulation activates lymphocytes, but it also increases the expression of programmed cell death protein 1 (PD-1), an inhibitory immune protein. The binding of PD-1 to programmed death ligand 1 (PD-L1) regulates the response to antigens [8]; it reduces the immune response and enhances tolerance to foreign antigens, increasing the risk of chronic infections [9].
Different thymic peptides enhance immune function [10]. Thymosin alpha and prothymosin alpha are among the best-studied thymic peptides [11]. Thymosin inhibits the replication of HHV-1 [12], and it increases the major histocompatibility complex(MHC)-mediated presentation of viral antigens on the cell surface for enhanced recognition of infected cells [13,14]. Moreover, thymosin alpha stimulates the production of interferon gamma (IFN-γ), interleukin (IL)-7, and IL-15, thereby increasing the number of T helper cells (Th), cytotoxic T cells, and natural killer (NK) cells [13,15].
The aim of this study was to measure the percentages of CD4+ and CD8+ cells co-expressing PD-1 and PD-L1 in patients with reactivated HHV-1 infections and to investigate the ability of immune cells to produce cytokines after stimulation with thymic peptides. We hypothesized that reactivation of herpes infections would be associated with the appearance of lymphocyte exhaustion markers on lymphocytes and that thymic peptides would stimulate antiviral responses in peripheral blood mononuclear cells (PBMCs).

2. Results

2.1. Participants

We enrolled 50 women with reactivated HHV-1 infections (age range: 19–66 years; mean age ± standard deviation (SD): 41.21 ± 12.42 years). Twenty volunteers served as a control group (age range: 18–69 years; mean age ± SD:42.87 ± 15.81 years). Table 1 presents the characteristics of the patients.

2.2. Frequencies of Basic Lymphocyte Subsets in Patients with Reactivated HHV-1 Infections and Controls

We assessed the frequencies of basic lymphocyte subsets in patients with reactivated HHV-1 infection and the control group. Patients exhibited significantly lower percentages ofCD19+ B cells than controls (p = 0.002, Table 2). Moreover, in the group of patients with reactivated HHV-1 infection, we found higher frequencies of CD4+CD25+highFOXP3+ T regulatory (Treg) cells and NK cells (p < 0.001, p < 0.025, respectively, Table 2).

2.3. Frequencies of CD25+, PD-1+, and PD-L1+ T and B Lymphocytes in Patients with Reactivated HHV-1 Infections and Controls

Patients with reactivated HHV-1 infections had a significantly higher percentage of CD3+CD25+ cells than did the controls (p < 0.001, Table 3). Moreover, the percentages of CD4+ cells and CD8+ cells co-expressing PD-1 were higher in patients with reactivated HHV-1 infections than in the control group (p < 0.001 for both comparisons). The percentages of CD4+ cells and CD8+ cells co-expressing PD-L1 were increased in patients with recurrent HHV-1 infections compared with the control group (p < 0.001 for both comparisons, Table 3).

2.4. Frequencies of PD-1+ and PD-L1+ B and T Lymphocytes in Patients with Reactivated HHV-1 Infections before and after 2 Months of Thymostimulin Therapy

Thymostimulin (TFX) is an approved medication for patients with impaired immune function. However, the impact of this substance on immune exhaustion, i.e., expression of PD-1/PD-L1 antigens, has yet to be established. In order to address this issue, we assessed the changes in the frequencies of PD-1+ and PD-L1+ T and B lymphocytes. All 50 patients were treated for 2 months with TFX in the following scheme: subcutaneous injection of one ampoule (10 mg) daily for 30 days, followed, over the next 30 days, by subcutaneous injection of one ampoule two times per week. During this period, all patients were free from HHV-1 infection reactivation, and we noticed a significant reduction in the percentages of CD19+PD-1+ B lymphocytes, CD4+PD-1+ T lymphocytes, CD8+PD-1+ T lymphocytes, CD4+PD-L1+ T lymphocytes, and CD8+PD-L1+ T lymphocytes (p < 0.001 for all comparisons, Table 4).

2.5. Stimulation of PBMCs with Thymic Peptides in Patients with Reactivated HHV-1 Infections

The concentrations of IFN-γ and IL-2 increased significantly in PBMC cultures supplemented with thymic peptides at the concentration of 200 µg/mL compared with concentrations in cultures without these peptides (p < 0.001 and p = 0.023, respectively; Table 3). The addition of thymic peptides to PBMC cultures did not significantly change the concentrations of IL-4 and IL-10 (Figure 1).

3. Discussion

Exposure of lymphocytes to viral antigens induces markers of activation on cell surfaces. Such activation markers are classified as early (CD69 and CD71) or late (CD25 and HLA-DR) markers depending on the time of stimulation [16]. T cells play an essential role in antiviral responses [17]. Jeon et al. showed that CD8+ T cells close to sensory neurons latently infected with HHV-1 expressed markers of immune activation, which indicated that lymphocytes come into contact with low levels of viral antigens during latency [18].Our study found that the percentage of CD3+ and CD4+ T cells co-expressing the CD25 activation antigen was higher in patients with reactivated herpes infections than in the control group. However, peripheral blood cells may not have reflected well the population of tissue-resident immune cells that come into contact with HHV-1 antigens.
Viral infections can strongly activate lymphocytes, which may lead to functional cell exhaustion [19]. Although virus-specific lymphocytes display activation markers, they are unable to kill infected cells or release antiviral cytokines [20]. The inability of cells to carry out effector functions may be one of the mechanisms underlying the silencing of antiviral responses, which impairs virus elimination [7,20,21]. The earliest sign of immune exhaustion is a decrease in immune cells’ capacity to secrete IFN-γ and IL-2 [8,22,23]. Yang et al. showed that the Us3 protein, which is expressed by HHV-1, inhibits T-cell activation and other immune responses [24].
Chronic antigen stimulation during viral reactivation may be associated with higher percentages of CD8+ T cells co-expressing PD-1. Khan et al. showed that exhaustion of CD8+ T cells can lead to recurrent symptomatic HHV-1 infections [21]. In addition, Mott et al. found higher levels of PD-1 transcripts during HHV-1 latency in neurons [25]. PD-1 expression inactivates CD8+ T cells in sensory neurons and impairs virus elimination [25]. Likewise, we found that the percentages of CD8+ and CD4+ cells co-expressing PD-1 were significantly higher in patients with reactivated HHV-1 infections than in the control group.
The interaction between lymphocytes and antigen-presenting cells (APCs) regulates immune responses. Co-stimulatory molecules determine whether T cells are activated or anergized [26,27]. PD-L1 transmits inhibitory signals to T cells that decrease their expansion, cytotoxicity, and cytokine production [28]. Blocking PD-L1 expression in APCs enhances lymphocyte proliferation and cytokine production by CD4+ T cells [29]. Importantly, an increased expression of PD-L1 in APCs was observed after HHV-1 infection [8]. In our study, the percentages of CD8+ and CD4+ cells co-expressing PD-L1 were significantly higher in patients with reactivated HHV-1 infections than in the control group. The simultaneously increased expression of PD-1 and PDL-1 suggests that there is a negative feedback control loop in the PD-1/PD-L1 axis during reactivation of herpes infections.
Sharpe et al. found an increased expression of PD-1 in CD4+ T cells in an animal model of keratitis [30]. In that model, blocking the PD-L1/PD-1 interaction improved both the proliferation and effector functions of HHV-1-specific CD4+ T cells [30]. Antoine et al. reported that CD4+ T cells specific to cytomegalovirus expressed activation markers during primary infection with the virus, but they had a decreased ability to produce IFN-γ, tumor necrosis factor alpha (TNF-α), and IL-2 [31].In our study, reactivation of herpes infection was associated with the presence of activated immune cells that, at the same time, expressed exhaustion markers. These findings indicate that the mechanisms that impair the immune response are similar for all herpes viruses, including HHV-1 and cytomegalovirus.
Immune exhaustion in chronic viral infections is most likely caused by alterations in many cell signaling pathways other than the upregulation of the PD-1/PD-L1 pathway. For example, infection with HIV or the hepatitis C virus increases the expression of a number of negative co-stimulatory molecules, including lymphocyte activation gene-3 (LAG-3), CD160, cytolytic T-lymphocyte antigen-4 (CTLA-4), T-cell immunoglobulin mucin-containing domain-3 (TIM-3), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) [32,33]. Moreover, immune exhaustion might be related to mitochondrial dysfunction in immune effector cells [34]. Importantly, chronic viral infections might exhaust innate cells such as mucosal-cell-associated invariant T cells [35,36].
Matteucci et al. reported an enhanced antiviral response after the administration of thymosin alpha 1, which improves recognition of viral antigens and regulates cytokine gene expression [13,37,38]. In a study on patients with hepatitis B, Jiang et al. showed that treatment with thymosine alpha increased helper T cells’ production of IFN-γ, IL-2, IL-4, and TNF-α [39]. Decman et al. found that the addition of IFN-γ to cell cultures of neurons infected with HHV-1 blocked viral reactivation [40]. In our study, the thymic peptides contained in thymostimulin increased PBMCs’ production of IFN-γ and IL-2. This finding suggests that thymic peptides could increase the production of proinflammatory cytokines in patients with reactivated HHV-1 infections.
Our proof-of-concept study had limitations. The effect of thymostimulin was tested in vitro only, and the mechanism underlying thymostimulin-induced IFN-γ and IL-2 hypersecretion remains unknown. For example, it could involve foreign-antigen stimulation (thymostimulin is produced from bovine thymus extract). Experiments with recombinant human thymic peptides could help to test this possibility. Future studies should investigate the effect of thymostimulinon other diseases and healthy people. Moreover, further experiments should investigate whether reactivation of herpes infection is associated with impaired immune responses in general or with the antigens of HHV-1 only. Such experiments could determine whether thymostimulin improves antigen-specific immune responses or the immune response in general. Finally, we used CD25 as an activation marker of CD4+ cells; however, this approach is limited because a subset of these cells (~20%) are regulatory cells co-expressing FOXP3.

4. Materials and Methods

4.1. Patients with Reactivated HHV-1 Infections and the Control Group

Participants were patients who were referred to the Department of Clinical Immunology and Immunotherapy due to recurrent oral herpes. In practice, nearly all such patients are women, who seek medical help for oral herpes much more often than do men. We decided to include only women in order to have a more homogenous sample. All patients presented with reactivated HHV-1 infection at the time of the study. The control group included women scheduled for elective septoplasty due to nasal septum deviation or external nose deformity; we included women with no history of herpes virus infection. All controls were seronegative for HHV-1 and HHV-2. The exclusion criteria for both patients and controls were: treatment with an immunomodulating or hormonal medication, an infection within the 6 month period prior to enrolment, a confirmed allergy, an autoimmune disease, a history of blood transfusion, tuberculosis, cancer, and any condition impairing immunity. Peripheral blood samples were taken before treatment from 50 women with reactivated HHV-1 infections. Twenty volunteers served as a control group. The study was approved by the Medical Ethical Committee of the Medical University in Lublin (approval no., KE-0254/263/2014, 29 September 2014), and informed consent was obtained from each participant. The trial with TFX was open, single-arm, and uncontrolled.

4.2. Material

Peripheral blood was collected in EDTA-coated tubes (10 mL) (Sarstedt, Nümbrecht, Germany) for immunophenotyping and isolation of PBMCs. Peripheral blood was also collected in tubes (5 mL) coated with a clotting activator to obtain serum for the measurement of anti-HHV-1 and HHV-2 antibodies.

4.3. Immunophenotyping

For immunophenotyping, flow cytometry was performed as described previously [41,42]. The cells were phenotypically characterized by incubation (20 min in the dark at room temperature) with a combination of fluorescein isothiocyanate (FITC), phycoerythrin (PE), and CyChrome-labeled monoclonal antibodies. Percentages of PD-1-positive and PD-L1-positive T and B lymphocytes were measured using a combination of the following monoclonal antibodies: CD45FITC/CD14 PE, CyChrome Mouse Anti-Human CD3, FITC Mouse Anti-Human CD19, FITC Mouse Anti-Human CD4, FITC Mouse Anti-Human CD8, PE Mouse Anti-Human CD279 (PD-1), and PE Mouse Anti-Human CD274 (PD-L1). Figure S1 shows CD4+ and CD8+ PD-1-positive (A) and CD4+ and CD8+ PD-L1-positive (B) T cells. The percentage of CD4+CD25+highFOXP3+ Tregs in the CD4+ T lymphocyte subpopulation was determined using the Human Treg Flow kit (FOXP3 Alexa Fluor 488/CD4 PE-Cy5/CD25 PE, BioLegend, San Diego, CA, USA). To identify the activated peripheral blood cells, we used the anti-CD25 PE-Cy5 mouse monoclonal antibody. Percentages of NK and natural killer T-like (NKT-like) cells were measured with flow cytometry using anti-CD3/FITC, CD16CD56/PE, and CD45/PerCPmonoclonal antibodies (BD Biosciences, San Jose, CA, USA), which allowed for the simultaneous assessment of CD3+T lymphocytes and NK (CD16+CD56+) cells. During analysis, the CD3+CD16+CD56+ (NKT-like cell) population was also determined. Monoclonal antibodies were purchased from BDBiosciences, USA. The “cleanness”of the lymphocyte gateway was evaluated by examining the distribution of cells in the coordinates of CD45CD14 (BDBiosciences, USA). The percentage of positive cells was measured froma cut-offset using an isotype-matched non-specific control antibody. Three-color immunofluorescence analyses were performed using a FACSCalibur flow cytometer (BD Biosciences, USA) equipped with a 488 nm argon laser. A minimum of 10,000 events were acquired and analyzed using CellQuest Software (BD Biosciences, USA). The percentages of cells expressing surface markers were measured. Background fluorescence was determined. The samples were gated on forwardscatter versus side scatter to exclude debris and cell aggregates.

4.4. Cytokine Secretion in Cultures of PBMCs Stimulated with Thymic Peptides

We measured the concentrations of cytokines secreted by cultured PBMCs from 15 patients with reactivated HHV-1 infections. PBMCs were obtained from peripheral blood by Ficoll-Hypaque density gradient centrifugation (2500 r/min for 20 min at room temperature). After collection, PBMCs were washed twice in phosphate-buffered saline. The viability of isolated cells was checked by trypan blue staining, and samples with a viability of<90% were excluded. PBMCs were stimulated with thymostimulin (Finepharm, Jelenia Góra, Poland), which is a bovine thymic extract containing proteins and polypeptides with a mass of 4000–12,000 Da.In Poland, thymostimulin is an approved medication for patients with an impaired immune function; however, the impact of this substance on immune exhaustion, i.e., expression of PD-1/PD-L1 antigens, has yet to be established.Cell cultures contained 106 PBMCS/mL of medium. Two concentrations of thymostimulin were used: 100 µg/mL and 200 µg/mL. The cultures were performed in complete medium (RPMI-1640 supplemented with 10% fetal bovine serum (FBS), 200mM L-Gln(PAA Laboratories, Cölbe, Germany ), penicillin (100 IU/mL), streptomycin (50ug/mL), and neomycin (100ug/mL, Sigma Aldrich, Hamburg, Germany)). Cultures without thymic peptides were also performed. After 24 h at 37°C in 5% CO2, the cell cultures were collected. Cytokine concentrations in supernatants from cell cultures were then measured via enzyme-linked immunosorbent assays (ELISA).

4.5. Measurement of the Concentrations of IFN-γ, IL-2, IL-4, IL-10, anti-HHV-1 IgM Antibodies, anti-HHV-1 IgG Antibodies, anti-HHV-2 IgM Antibodies, and anti-HHV-2 IgG Antibodies

The concentrations of IFN-γ, IL-2, IL-4, and IL-10 were measured using commercially available ELISA kits (Human IFN-γ Quantikine (sensitivity, <8 pg/mL), Human IL-2 Quantikine (sensitivity, <7 pg/mL), Human IL-4 Quantikine (sensitivity, 0.03–0.22 pg/mL), and Human IL-10 Quantikine (sensitivity, 0.03–0.17 pg/mL), all purchased from RQD Systems, Minneapolis, MN, USA). The concentrations of anti-HSV1 IgM, anti-HSV1 IgG, anti-HHV-2IgM, and anti-HHV-2IgGantibodies were measured usingELISAs (IBL International, Hamburg, Germany). The cut-off value for anti-HHV-1IgM and anti-HHV-1IgGantibodies was >12 U/mL.The cut-off value for anti-HHV-2IgM and anti-HHV-2IgG antibodies was >11 U/mL. The concentrations of antibodies and cytokines were calculated based on standard curves. All ELISAs were performed according to the manufacturers’ instructions.

4.6. Statistical Analysis

The normality of the distribution of continuous variables was verified with the Shapiro–Wilk test. The groups were compared with the Mann–Whitney test or the Wilcoxon test. p < 0.05 was considered to be statistically significant. Statistica 12 software (StatSoft, Tulsa, OK, USA) was used for all analyses.

5. Conclusions

Reactivation of herpes infections is associated with an increased proportion of anergic immune cells, which impairs the immune response to the pathogen. The stimulation of PBMCs with thymic peptides increases the immune response, particularly the Th1-dependent response, which confirms the antiviral effects of thymic peptides. This approach may be useful to overcome anergy in patients with reactivated HHV-1 infections.

Smentary Materials

Smentary Materials can be found at https://www.mdpi.com/1422-0067/21/7/2379/s1.

Author Contributions

Conceptualization, all authors; Methodology, all authors; Software, not applicable; Validation, not applicable; Formal Analysis, all authors; Investigation, all authors; Resources, all authors; Data Curation, all authors; Writing—Original Draft Preparation, A.H. and E.G.; Writing—Review & Editing, all authors; Visualization, all authors; Supervision, all authors; Project Administration, all authors; Funding Acquisition, all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Polish National Science Center (grant no UMO-2016/21/B/NZ6/02279) and the Medical University of Lublin (grant no DS460).

Acknowledgments

The authors would like to thank Proper Medical Writing Sp. z o.o. for correcting the language in the final version of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

APCantigen-presenting cells
CTLA-4cytolytic T-lymphocyte antigen-4
ELISAenzyme-linked immunosorbent assay
HHV-1human alphaherpesvirus 1
IFN-γinterferon gamma
ILinterleukin
LAG-3lymphocyte activation gene-3
NKnatural killer cell
NKT-likenatural killer T-like cell
PBMC peripheral blood mononuclear cells
PD-1Programmed cell death protein 1
PD-L1programmed death ligand 1
SDstandard deviation
TFXthymostimulinum
ThT helper cells
TIM-3T-cell immunoglobulin mucin-containing domain-3
TNF-αtumor necrosis factor alpha
TRAILtumor necrosis-factor-related apoptosis-inducing ligand

References

  1. Denes, C.E.; Miranda-Saksena, M.; Cunningham, A.L.; Diefenbach, R.J. Cytoskeletons in the Closet—Subversion in Alphaherpesvirus Infections. Viruses 2018, 10, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Schulte, J.M.; Bellamy, A.R.; Hook, E.W.; Bernstein, D.I.; Levin, M.J.; Leone, P.A.; Sokol-Anderson, M.L.; Ewell, M.G.; Wolff, P.A.; Heineman, T.C.; et al. HSV-1 and HSV-2 Seroprevalence in the United States among Asymptomatic Women Unaware of Any Herpes Simplex Virus Infection (Herpevac Trial for Women). South. Med. J. 2014, 107, 79–84. [Google Scholar] [CrossRef] [PubMed]
  3. Li, W.; Wang, X.-H.; Luo, Z.; Liu, L.-F.; Yan, C.; Yan, C.-Y.; Chen, G.-D.; Gao, H.; Duan, W.-J.; Kurihara, H.; et al. Traditional Chinese Medicine as a Potential Source for HSV-1 Therapy by Acting on Virus or the Susceptibility of Host. Int. J. Mol. Sci. 2018, 19, 3266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Roy, M.; Lebeau, L.; Chessa, C.; Damour, A.; Ladram, A.; Oury, B.; Boutolleau, D.; Bodet, C.; Lévêque, N. Comparison of Anti-Viral Activity of Frog Skin Anti-Microbial Peptides Temporin-Sha and [K3]SHa to LL-37 and Temporin-Tb against Herpes Simplex Virus Type 1. Viruses 2019, 11, 77. [Google Scholar] [CrossRef] [Green Version]
  5. Sharifi-Rad, J.; Iriti, M.; Setzer, W.N.; Sharifi-Rad, M.; Roointan, A.; Salehi, B. Antiviral activity of Veronica persica Poir. on herpes virus infection. Cell. Mol. Biol. 2018, 64, 11–17. [Google Scholar] [CrossRef] [Green Version]
  6. Egan, K.P.; Wu, S.; Wigdahl, B.; Jennings, S.R. Immunological control of herpes simplex virus infections. J. Neurovirol. 2013, 19, 328–345. [Google Scholar] [CrossRef] [Green Version]
  7. Blackburn, S.D.; Crawford, A.; Shin, H.; Polley, A.; Freeman, G.J.; Wherry, E.J. Tissue-Specific Differences in PD-1 and PD-L1 Expression during Chronic Viral Infection: Implications for CD8 T-Cell Exhaustion. J. Virol. 2010, 84, 2078–2089. [Google Scholar] [CrossRef] [Green Version]
  8. Griffin, J.; Moulton, M.; Elmezayen, R.; Moorman, J. Negative Immunomodulators–Blunting Immunostimulation and Facilitating Infection. In Immune Response Activation; InTech: London, UK, 2014. [Google Scholar]
  9. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [Green Version]
  10. Goldstein, A.L. History of the Discovery of the Thymosins. Ann. N. Y. Acad. Sci. 2007, 1112, 1–13. [Google Scholar] [CrossRef]
  11. Tuthill, C.; Rios, I.; McBeath, R. Thymosin alpha 1: Past clinical experience and future promise. Ann. N. Y. Acad. Sci. 2010, 1194, 130–135. [Google Scholar] [CrossRef]
  12. Mamedov, M.K.; Safarova, S.M.; Dadasheva, A.E. Impact of thymosin-alpha1 on reproduction of herpes simplex virus in cell system and on course of acute experimental infection in mice. Georgian Med. News 2012, 71–75. [Google Scholar]
  13. Li, J.; Liu, C.H.; Wang, F.S. Thymosin alpha 1: Biological activities, applications and genetic engineering production. Peptides 2010, 31, 2151–2158. [Google Scholar] [CrossRef] [PubMed]
  14. Matteucci, C.; Minutolo, A.; Sinibaldi-Vallebona, P.; Palamara, A.T.; Rasi, G.; Mastino, A.; Garaci, E. Transcription profile of human lymphocytes following in vitro treatment with thymosin alpha-1. Ann. N. Y. Acad. Sci. 2010, 1194, 6–19. [Google Scholar] [CrossRef] [PubMed]
  15. Robert, S.; King, C.W.T. Thymosin Apha 1–A Peptide Immune Modulator with a Broad Range of Clinical Applications. Clin. Exp. Pharmacol. 2013, 03, 133. [Google Scholar]
  16. Reddy, M.; Eirikis, E.; Davis, C.; Davis, H.M.; Prabhakar, U. Comparative analysis of lymphocyte activation marker expression and cytokine secretion profile in stimulated human peripheral blood mononuclear cell cultures: An in vitro model to monitor cellular immune function. J. Immunol. Methods 2004, 293, 127–142. [Google Scholar] [CrossRef]
  17. Macleod, B.L.; Bedoui, S.; Hor, J.L.; Mueller, S.N.; Russell, T.A.; Hollett, N.A.; Heath, W.R.; Tscharke, D.C.; Brooks, A.G.; Gebhardt, T. Distinct APC Subtypes Drive Spatially Segregated CD4+ and CD8+ T-Cell Effector Activity during Skin Infection with HSV-1. PLoS Pathog. 2014, 10, e1004303. [Google Scholar] [CrossRef] [Green Version]
  18. Jeon, S.; St Leger, A.J.; Cherpes, T.L.; Sheridan, B.S.; Hendricks, R.L. PD-L1/B7-H1 regulates the survival but not the function of CD8+ T cells in herpes simplex virus type 1 latently infected trigeminal ganglia. J. Immunol. 2013, 190, 6277–6286. [Google Scholar] [CrossRef] [Green Version]
  19. Kahan, S.M.; Wherry, E.J.; Zajac, A.J. T cell exhaustion during persistent viral infections. Virology 2015, 479–480, 180–193. [Google Scholar] [CrossRef]
  20. Miranda-Saksena, M.; Denes, C.E.; Diefenbach, R.J.; Cunningham, A.L. Infection and Transport of Herpes Simplex Virus Type 1 in Neurons: Role of the Cytoskeleton. Viruses 2018, 10, 92. [Google Scholar] [CrossRef] [Green Version]
  21. Khan, A.A.; Srivastava, R.; Chentoufi, A.A.; Geertsema, R.; Thai, N.T.U.; Dasgupta, G.; Osorio, N.; Kalantari, M.; Nesburn, A.B.; Wechsler, S.L.; et al. Therapeutic Immunization with a Mixture of Herpes Simplex Virus 1 Glycoprotein D-Derived “Asymptomatic” Human CD8+ T-Cell Epitopes Decreases Spontaneous Ocular Shedding in Latently Infected HLA Transgenic Rabbits: Association with Low Frequency of Local PD-1+ TIM-3+ CD8+ Exhausted T Cells. J. Virol. 2015, 89, 6619–6632. [Google Scholar]
  22. Crawford, A.; Wherry, E.J. The diversity of costimulatory and inhibitory receptor pathways and the regulation of antiviral T cell responses. Curr. Opin. Immunol. 2009, 21, 179–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Torti, N.; Oxenius, A. T cell memory in the context of persistent herpes viral infections. Viruses 2012, 4, 1116–1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Yang, Y.; Wu, S.; Wang, Y.; Pan, S.; Lan, B.; Liu, Y.; Zhang, L.; Leng, Q.; Chen, D.; Zhang, C.; et al. The Us3 Protein of Herpes Simplex Virus 1 Inhibits T Cell Signaling by Confining Linker for Activation of T Cells (LAT) Activation via TRAF6 Protein. J. Biol. Chem. 2015, 290, 15670–15678. [Google Scholar] [CrossRef] [Green Version]
  25. Mott, K.R.; Bresee, C.J.; Allen, S.J.; BenMohamed, L.; Wechsler, S.L.; Ghiasi, H. Level of herpes simplex virus type 1 latency correlates with severity of corneal scarring and exhaustion of CD8+ T cells in trigeminal ganglia of latently infected mice. J. Virol. 2009, 83, 2246–2254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Zhang, Y.; Chung, Y.; Bishop, C.; Daugherty, B.; Chute, H.; Holst, P.; Kurahara, C.; Lott, F.; Sun, N.; Welcher, A.A.; et al. Regulation of T cell activation and tolerance by PDL2. Proc. Natl. Acad. Sci. USA 2006, 103, 11695–11700. [Google Scholar] [CrossRef] [Green Version]
  27. Latchman, Y.E.; Liang, S.C.; Wu, Y.; Chernova, T.; Sobel, R.A.; Klemm, M.; Kuchroo, V.K.; Freeman, G.J.; Sharpe, A.H. PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. Proc. Natl. Acad. Sci. USA 2004, 101, 10691–10696. [Google Scholar] [CrossRef] [Green Version]
  28. Gianchecchi, E.; Delfino, D.V.; Fierabracci, A. Recent insights into the role of the PD-1/PD-L1 pathway in immunological tolerance and autoimmunity. Autoimmun. Rev. 2013, 12, 1091–1100. [Google Scholar] [CrossRef]
  29. Brown, J.A.; Dorfman, D.M.; Ma, F.-R.; Sullivan, E.L.; Munoz, O.; Wood, C.R.; Greenfield, E.A.; Freeman, G.J. Blockade of Programmed Death-1 Ligands on Dendritic Cells Enhances T Cell Activation and Cytokine Production. J. Immunol. 2003, 170, 1257–1266. [Google Scholar] [CrossRef]
  30. Sharpe, A.H.; Wherry, E.J.; Ahmed, R.; Freeman, G.J. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat. Immunol. 2007, 8, 239–245. [Google Scholar] [CrossRef]
  31. Antoine, P.; Olislagers, V.; Huygens, A.; Lecomte, S.; Liesnard, C.; Donner, C.; Marchant, A. Functional Exhaustion of CD4+ T Lymphocytes during Primary Cytomegalovirus Infection. J. Immunol. 2012, 189, 2665–2672. [Google Scholar] [CrossRef] [Green Version]
  32. Shankar, E.M.; Che, K.F.; Messmer, D.; Lifson, J.D.; Larsson, M. Expression of a Broad Array of Negative Costimulatory Molecules and Blimp-1 in T Cells following Priming by HIV-1 Pulsed Dendritic Cells. Mol. Med. 2011, 17, 229–240. [Google Scholar] [CrossRef] [PubMed]
  33. Barathan, M.; Mohamed, R.; Vadivelu, J.; Chang, L.Y.; Vignesh, R.; Krishnan, J.; Sigamani, P.; Saeidi, A.; Ram, M.R.; Velu, V.; et al. CD8+ T cells of chronic HCV-infected patients express multiple negative immune checkpoints following stimulation with HCV peptides. Cell. Immunol. 2017, 313, 1–9. [Google Scholar] [CrossRef] [PubMed]
  34. Saeidi, A.; Zandi, K.; Cheok, Y.Y.; Saeidi, H.; Wong, W.F.; Lee, C.Y.Q.; Cheong, H.C.; Yong, Y.K.; Larsson, M.; Shankar, E.M. T-Cell Exhaustion in Chronic Infections: Reversing the State of Exhaustion and Reinvigorating Optimal Protective Immune Responses. Front. Immunol. 2018, 9, 2569. [Google Scholar] [CrossRef] [PubMed]
  35. Saeidi, A.; Tien Tien, V.L.; Al-Batran, R.; Al-Darraji, H.A.; Tan, H.Y.; Yong, Y.K.; Ponnampalavanar, S.; Barathan, M.; Rukumani, D.V.; Ansari, A.W.; et al. Attrition of TCR Vα7.2+ CD161++ MAIT Cells in HIV-Tuberculosis Co-Infection Is Associated with Elevated Levels of PD-1 Expression. PLoS ONE 2015, 10, e0124659. [Google Scholar] [CrossRef] [PubMed]
  36. Yong, Y.K.; Saeidi, A.; Tan, H.Y.; Rosmawati, M.; Enström, P.F.; Al Batran, R.; Vasuki, V.; Chattopadhyay, I.; Murugesan, A.; Vignesh, R.; et al. Hyper-Expression of PD-1 Is Associated with the Levels of Exhausted and Dysfunctional Phenotypes of Circulating CD161++TCR iVα7.2+ Mucosal-Associated Invariant T Cells in Chronic Hepatitis B Virus Infection. Front. Immunol. 2018, 9, 472. [Google Scholar] [CrossRef] [Green Version]
  37. Matteucci, C.; Minutolo, A.; Pollicita, M.; Balestrieri, E.; Grelli, S.; D’Ettorre, G.; Vullo, V.; Bucci, I.; Luchini, A.; Aquaro, S.; et al. Thymosin α1 potentiates the release by CD8+ cells of soluble factors able to inhibit HIV-1 and human T lymphotropic virus 1 infection in vitro. Expert Opin. Biol. Ther. 2015, 15, 83–100. [Google Scholar] [CrossRef]
  38. Pierluigi, B.; D’Angelo, C.; Fallarino, F.; Moretti, S.; Zelante, T.; Bozza, S.; De Luca, A.; Bistoni, F.; Garaci, E.; Romani, L. Thymosin α1: The regulator of regulators? Ann. N. Y. Acad. Sci. 2010, 1194, 1–5. [Google Scholar] [CrossRef]
  39. Jiang, Y.-F.; Ma, Z.-H.; Zhao, P.-W.; Pan, Y.; Liu, Y.-Y.; Feng, J.-Y.; Niu, J.-Q. Effect of Thymosin-α 1 on T-helper 1 Cell and T-helper 2 Cell Cytokine Synthesis in Patients with Hepatitis B Virus e Antigen-positive Chronic Hepatitis B. J. Int. Med. Res. 2010, 38, 2053–2062. [Google Scholar] [CrossRef]
  40. Decman, V.; Kinchington, P.R.; Harvey, S.A.K.; Hendricks, R.L. Gamma Interferon Can Block Herpes Simplex Virus Type 1 Reactivation from Latency, Even in the Presence of Late Gene Expression. J. Virol. 2005, 79, 10339–10347. [Google Scholar] [CrossRef] [Green Version]
  41. Pellegatta, S.; Di Ianni, N.; Pessina, S.; Paterra, R.; Anghileri, E.; Eoli, M.; Finocchiaro, G. ABCC3 Expressed by CD56dim CD16+ NK Cells Predicts Response in Glioblastoma Patients Treated with Combined Chemotherapy and Dendritic Cell Immunotherapy. Int. J. Mol. Sci. 2019, 20, 5886. [Google Scholar] [CrossRef] [Green Version]
  42. Korona-Glowniak, I.; Grywalska, E.; Grzegorczyk, A.; Roliński, J.; Glowniak, A.; Malm, A. Bacterial Colonization in Patients with Chronic Lymphocytic Leukemia and Factors Associated with Infections and Colonization. J. Clin. Med. 2019, 8, 861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Secretion of IFN-γ (A), IL-2 (B), IL-4 (C), and IL-10 (D) by peripheral blood mononuclear cells from patients (n = 50) with reactivated HHV-1 infections. Boxes represent the interquartile range and error bars represent the range of observations. p-values are shown for significant differences. TFX: thymostimulin.
Figure 1. Secretion of IFN-γ (A), IL-2 (B), IL-4 (C), and IL-10 (D) by peripheral blood mononuclear cells from patients (n = 50) with reactivated HHV-1 infections. Boxes represent the interquartile range and error bars represent the range of observations. p-values are shown for significant differences. TFX: thymostimulin.
Ijms 21 02379 g001
Table 1. Characteristics of patients with reactivated human alphaherpesvirus 1 (HHV-1) infections and controls.
Table 1. Characteristics of patients with reactivated human alphaherpesvirus 1 (HHV-1) infections and controls.
VariableStatisticsPatients (n = 50)Controls (n = 20)
Age (years)median (min–max)40.05 (19–66)44 (18–69)
mean ± SD41.21 ± 12.4242.87 ± 15.81
anti-HHV-1IgM, n (%)Yes8 (16%)0
No42 (84%)20 (100%)
anti-HHV-1 IgM, U/mLmedian (min–max)27.72 (12.11–62.7)-
mean ± SD27.45 ± 18.33-
anti-HHV-1IgG, n (%)Yes50 (100%)0
No020 (100%)
anti-HHV-1 IgG, U/mLmedian (min–max)70.62 (22.41–1701.82)-
mean ± SD167.09 ± 301.18-
anti-HHV-2IgM, n (%)Yes00
No50 (100%)20 (100%)
anti- HHV-2 IgG, n (%)Yes00
No50 (100%)20 (100%)
Table 2. Subpopulation of lymphocytes in patients with reactivated HHV-1 infections before and after thymostimulin therapy (TFX),and the control group.
Table 2. Subpopulation of lymphocytes in patients with reactivated HHV-1 infections before and after thymostimulin therapy (TFX),and the control group.
VariablePatients with Reactivated HHV-1 Infections (n = 50)Control Group
(n = 20)
p (Patients before TFX vs. Controls)p (Patients before vs. after TFX)
before TFX after TFX
CD3+
T lymphocytes (%)
median (min–max)69.62
(62.55–74.97)
73.66
(62.23-83.19)
71.94
(61.61–74.53)
NS0.001
mean ± SD69.31 ± 3.3573.31 ± 4.2970.44 ± 3.53
CD19+ B lymphocytes
(%)
median (min–max)10.44
(6.12–16.96)
15.31
(0.29–19.46)
13.62
(8.69–16.88)
0.002<0.001
mean ± SD10.60 ± 3.2213.95 ± 4.1213.33 ± 2.46
CD4+CD3+ T lymphocytes
(%)
median (min–max)39.16
(25.19–59.88)
40.25
(23.07–55.45)
38.34
(33.40–57.43)
NSNS
mean ± SD39.92 ± 6.6238.76 ± 9.3641.06 ± 6.36
CD8+CD3+ T lymphocytes
(%)
median (min–max)29.42
(11.17–49.29)
28.81
(9.01–52.22)
27.71
(12.78–37.16)
NSNS
mean ± SD28.62 ± 7.0731.04 ± 11.3528.48 ± 6.10
CD4+CD3+/CD8+CD3+ ratiomedian (min–max)1.29
(0.51–4.86)
1.49
(0.50–6.15)
1.34
(0.92–4.49)
NSNS
mean ± SD1.58 ± 0.841.57 ± 1.041.58 ± 0.79
NK cells
(%)
median (min–max)17.77
(10.16–27.72)
10.19
(3.43–18.56)
14.26
(9.16–24.08)
0.025<0.001
mean ± SD17.51 ± 3.8510.30 ± 3.9615.09 ± 4.42
NKT-like cells (%)median (min–max)4.19
(0.84–16.06)
4.31
(0.91–17.83)
3.29
(1.86–4.91)
0.048NS
mean ± SD5.21 ± 3.355.43 ± 3.333.30 ± 1.02
T regulatory cells (CD4+CD25+
FOXP3) (%)
median (min–max)6.61
(3.13–16.85)
6.63
(3.11–13.66)
4.26
(3.23–7.45)
<0.0010.020
mean ± SD8.32 ± 3.827.10 ± 2.724.45 ± 1.06
SD: standard deviation; NS: not significant; TFX: thymostimulin therapy.
Table 3. Expression of the activation antigen CD25 and the immunosuppressive antigens PD-1 and PD-L1 on T and B lymphocytes in patients with reactivated HHV-1 infections before thymostimulin therapy (TFX) and the control group.
Table 3. Expression of the activation antigen CD25 and the immunosuppressive antigens PD-1 and PD-L1 on T and B lymphocytes in patients with reactivated HHV-1 infections before thymostimulin therapy (TFX) and the control group.
VariablePatients with Reactivated HHV-1 Infections before TFX
(n = 50)
Control Group (n = 20)p
CD3+CD25+ T lymphocytesmedian (min–max)46.71(26.99–68.01)37.06(18.65–47.13)<0.001
mean ± SD47.17 ± 10.1535.23 ± 8.33
CD19+CD25+ B lymphocytesmedian (min–max)36.00(16.05–58.87)18.64(10.88–27.50)<0.001
mean ± SD36.59 ± 11.1018.29 ± 4.16
CD4+CD25+ T lymphocytesmedian (min–max)46.12(28.49–75.50)35.79(30.64–47.50)<0.001
mean ± SD49.01 ± 10.5036.11 ± 3.97
CD19+PD-1+ B lymphocytesmedian (min–max)6.35(1.91–25.47)2.94(0.5–5.45)<0.001
mean ± SD8.44 ± 5.993.05 ± 1.36
CD4+PD-1+ T lymphocytesmedian (min–max)21.24(8.17–78.51)8.73(6.19–11.34)<0.001
mean ± SD25.03 ± 12.988.37 ± 1.46
CD8+PD-1+ T lymphocytesmedian (min–max)17.73(8.40–40.73)7.23(5.01–10.11)<0.001
mean ± SD19.42 ± 7.347.32 ± 1.62
CD19+PD-L1+B lymphocytesmedian (min–max)3.34(0.71–23.95)2.39(0.12–4.70)0.044
mean ± SD5.48 ± 5.232.33 ± 1.24
CD4+PD-L1+
T lymphocytes
median (min–max)14.67(6.78–63.81)8.37(2.58–13.31)<0.001
mean ± SD19.21 ± 12.787.99 ± 2.37
CD8+PD-L1+ T lymphocytesmedian (min–max)11.43(2.35–33.67)3.34(0.71–23.95)<0.001
mean ± SD12.39 ± 6.803.36 ± 1.24
SD: standard deviation; NS: not significant; TFX: thymostimulin therapy.
Table 4. Expression of the immunosuppressive antigens PD-1 and PD-L1 in T and B lymphocytes in patients with reactivated HHV-1 infections before and after 2 months of thymostimulin therapy (TFX).
Table 4. Expression of the immunosuppressive antigens PD-1 and PD-L1 in T and B lymphocytes in patients with reactivated HHV-1 infections before and after 2 months of thymostimulin therapy (TFX).
VariableBefore TFX (n = 50)After TFX (n = 50)p
CD19+PD-1+
B lymphocytes
median (min–max)6.35(1.91–25.47)3.42 (0.83–14.36)<0.001
mean ± SD8.44 ± 5.994.60 ± 3.37
CD4+PD-1+ T lymphocytesmedian (min–max)21.24(8.17–78.51)8.1 (1.14–27.96)<0.001
mean ± SD25.03 ± 12.9811.13 ± 7.9
CD8+PD-1+ T lymphocytesmedian (min–max)17.73(8.40–40.73)8.81(2.18–27.8)<0.001
mean ± SD19.42 ± 7.348.96 ± 5.14
CD19+PD-L1+
B lymphocytes
median (min–max)3.34(0.71–23.95)4.42(0.61–9.86)NS
mean ± SD5.48 ± 5.234.57 ± 2.60
CD4+PD-L1+ T lymphocytesmedian (min–max)14.67(6.78–63.81)7.67(1.45–23.99)<0.001
mean ± SD19.21 ± 12.788.23 ± 4.98
CD8+PD-L1+ T lymphocytesmedian (min–max)11.43(2.35–33.67)3.44(0.82–17.35)<0.001
mean ± SD12.39 ± 6.804.97 ± 3.55
SD: standard deviation; NS: not significant; TFX: thymostimulin therapy.

Share and Cite

MDPI and ACS Style

Hymos, A.; Grywalska, E.; Klatka, J.; Klatka, M.; Korona-Głowniak, I.; Roliński, J. ThymicPeptides Reverse Immune Exhaustion in Patients with Reactivated Human Alphaherpesvirus1 Infections. Int. J. Mol. Sci. 2020, 21, 2379. https://doi.org/10.3390/ijms21072379

AMA Style

Hymos A, Grywalska E, Klatka J, Klatka M, Korona-Głowniak I, Roliński J. ThymicPeptides Reverse Immune Exhaustion in Patients with Reactivated Human Alphaherpesvirus1 Infections. International Journal of Molecular Sciences. 2020; 21(7):2379. https://doi.org/10.3390/ijms21072379

Chicago/Turabian Style

Hymos, Anna, Ewelina Grywalska, Janusz Klatka, Maria Klatka, Izabela Korona-Głowniak, and Jacek Roliński. 2020. "ThymicPeptides Reverse Immune Exhaustion in Patients with Reactivated Human Alphaherpesvirus1 Infections" International Journal of Molecular Sciences 21, no. 7: 2379. https://doi.org/10.3390/ijms21072379

APA Style

Hymos, A., Grywalska, E., Klatka, J., Klatka, M., Korona-Głowniak, I., & Roliński, J. (2020). ThymicPeptides Reverse Immune Exhaustion in Patients with Reactivated Human Alphaherpesvirus1 Infections. International Journal of Molecular Sciences, 21(7), 2379. https://doi.org/10.3390/ijms21072379

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop