A Combination of Polybacterial MV140 and Candida albicans V132 as a Potential Novel Trained Immunity-Based Vaccine for Genitourinary Tract Infections

Leticia Martin-Cruz, Carmen Sevilla-Ortega, Cristina Benito-Villalvilla, Carmen M Diez-Rivero, Silvia Sanchez-Ramón, José Luis Subiza, Oscar Palomares, Leticia Martin-Cruz, Carmen Sevilla-Ortega, Cristina Benito-Villalvilla, Carmen M Diez-Rivero, Silvia Sanchez-Ramón, José Luis Subiza, Oscar Palomares

Abstract

Recurrent urinary tract infections (RUTIs) and recurrent vulvovaginal candidiasis (RVVCs) represent major healthcare problems with high socio-economic impact worldwide. Antibiotic and antifungal prophylaxis remain the gold standard treatments for RUTIs and RVVCs, contributing to the massive rise of antimicrobial resistance, microbiota alterations and co-infections. Therefore, the development of novel vaccine strategies for these infections are sorely needed. The sublingual heat-inactivated polyvalent bacterial vaccine MV140 shows clinical efficacy for the prevention of RUTIs and promotes Th1/Th17 and IL-10 immune responses. V132 is a sublingual preparation of heat-inactivated Candida albicans developed against RVVCs. A vaccine formulation combining both MV140 and V132 might well represent a suitable approach for concomitant genitourinary tract infections (GUTIs), but detailed mechanistic preclinical studies are still needed. Herein, we showed that the combination of MV140 and V132 imprints human dendritic cells (DCs) with the capacity to polarize potent IFN-γ- and IL-17A-producing T cells and FOXP3+ regulatory T (Treg) cells. MV140/V132 activates mitogen-activated protein kinases (MAPK)-, nuclear factor-κB (NF-κB)- and mammalian target of rapamycin (mTOR)-mediated signaling pathways in human DCs. MV140/V132 also promotes metabolic and epigenetic reprogramming in human DCs, which are key molecular mechanisms involved in the induction of innate trained immunity. Splenocytes from mice sublingually immunized with MV140/V132 display enhanced proliferative responses of CD4+ T cells not only upon in vitro stimulation with the related antigens contained in the vaccine formulation but also upon stimulation with phytohaemagglutinin. Additionally, in vivo sublingual immunization with MV140/V132 induces the generation of IgG and IgA antibodies against all the components contained in the vaccine formulation. We uncover immunological mechanisms underlying the potential mode of action of a combination of MV140 and V132 as a novel promising trained immunity-based vaccine (TIbV) for GUTIs.

Keywords: candida albicans V132; dendritic cells; polybacterial preparation MV140; recurrent urinary tract infections (RUTIs); recurrent vulvovaginal candidiasis (RVVCs); trained immunity-based vaccines (TIbVs).

Conflict of interest statement

OP has received fee for lectures or participation in Advisory Boards from Allergy Therapeutics, Amgen, AstraZeneca, Diater, GSK, Inmunotek SL, Novartis, Sanofi Genzyme, Stallergenes and Regeneron. OP has received research grants from Inmunotek SL and Novartis SL. JS is the founder and CEO of Inmunotek SL. The remaining 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. The reviewer SI declared a shared affiliation, with no collaboration, with several of the authors, LC, CO, CV and OP, to the handling editor at the time of review.

Copyright © 2021 Martin-Cruz, Sevilla-Ortega, Benito-Villalvilla, Diez‐Rivero, Sanchez-Ramón, Subiza and Palomares.

Figures

Figure 1
Figure 1
MV140/V132 induces hmoDCs maturation and production of pro-inflammatory cytokines and IL-10. (A) Cytokine production after stimulation of hmoDCs with control excipients (containing all excipients except the bacteria and yeast), V132, MV140, and MV140/V132 for 18 h quantified in cell-free supernatants by ELISA. (B) Cytokine levels of V132-treated hmoDCs plus cytokine levels of MV140-treated hmoDCs compared with cytokine levels of MV140/V132-stimulated hmoDCs. (C) Percentage of CD83- and HLA-DR-positive cells after stimulation with control excipients, V132, MV140 and MV140/V132 for 18 h. (D) Flow cytometry representative dot plots for the expression of surface markers CD83 and HLA-DR after stimulation of hmoDCs with the indicated stimulus. (E) Mean Fluorescence Intensity (MFI) values for CD83 and HLA-DR. Results are mean ± s.e.m. of 6-8 (A), 6 (B), and 7-8 (C, E) independent experiments. Paired Student t test, *P < 0.05, **P < 0.01, and ***P < 0.001.
Figure 2
Figure 2
MV140/V132-activated hmoDCs induce T cell proliferation and Th1, Th17, and IL-10 producing T cells. (A) Representative dot plots of proliferating CFSE-labeled allogeneic naive CD4+ T cells after 3 days of co-culture with control excipients-, V132-, MV140-, and MV140/V132-stimulated hmoDCs and the graph with the frequency of proliferating cells (right side). (B) Cytokines produced by allogeneic naive CD4+ T cells primed by hmoDCs in the presence of the indicated stimulus after 3 days quantified in cell-free supernatants by ELISA. (C) Levels of IL-17A and IL-10 for V132- plus MV140-stimulated co-cultures compared to MV140/V132-stimulated co-cultures. Results are mean ± s.e.m. of 5 (A), and 8 to 12 (B, C) independent experiments. Paired Student t test, *P < 0.05, **P < 0.01, and ***P < 0.001.
Figure 3
Figure 3
MV140/V132-activated hmoDCs generate IFN-γ-, IL-17A-, and IL-10-producing T cell production as well as FOXP3+ Treg cells. (A) Percentage of CD3+CD4+ T cells producing IFN-γ, IL-10 and IL-17A generated after 3 days of co-culture with control excipients-, V132-, MV140-, and MV140/V132-stimulated hmoDCs as determined by intracellular staining and flow cytometry analysis. Representative dot plots are shown for the intracellular staining after flow cytometry analysis. (B) Percentage of induced CD4+CD25highCD127-FOXP3+ Treg cells gated on CD4+ T cells after 3 days of co-culture with allogeneic control excipients-, V132-, MV140-, and MV140/V132-activated hmoDCs. Results are mean ± s.e.m. of 7-8 (A), and 5 (B) independent experiments. Paired Student t test, *P < 0.05, and **P < 0.01.
Figure 4
Figure 4
Activation of MAPKs, NF-κB, and mTOR signaling pathways in hmoDCs stimulated with control excipients, V132, MV140, or MV140/V132. (A, B) Western blot analysis of protein extracts from hmoDCs stimulated for 30 min in the indicated conditions. Quantification of the reactive phosphorylated bands by scanning densitometry. β-actin was used as a loading control. One representative example is shown. Results are mean ± s.e.m. of four to seven independent experiments. Paired Student t test, *P < 0.05, and **P < 0.01.
Figure 5
Figure 5
MV140/V132 induces metabolic and epigenetic reprogramming in hmoDCs. (A) Glucose consumption by hmoDCs treated with control excipients, V132, MV140 and MV140/V142 after 18 h and calculated metabolic rate. (B) Increase in lactate production in cell-free supernatants from MV140/V132-stimulated hmoDCs relative to MV140-activated hmoDCs. (C) Relative changes in fluorescence intensity of stimulated hmoDCs stained with MitoTracker Red. (D) Mitochondrial mass determination as the ratio of mitochondrial (mt) and nuclear (n) DNA as expressed by mt COXII/n GAPDH. (E) Messenger RNA expression levels of HIF-1α gene in hmoDCs stimulated for 18 h. Arbitrary units (A.u.) are 2−ΔCT values multiplied by 104, with ΔCT defined as the difference between the cycle threshold value for the gene of interest and EF1α. (F) Cytokine production by V132-, MV140-, and MV140/V132-activated hmoDCs in the presence of 5′-methylthioadenosine (MTA) or pargyline as inhibitors of histone methyltransferases and demethylases, respectively. Results are mean ± s.e.m. of 6 (A), 4 (B), 7 (C), 4 (D), 5 (E), and 4-7 (F) independent experiments. Paired Student t test, *P < 0.05, **P < 0.01, and ***P < 0.01.
Figure 6
Figure 6
Sublingual immunization of BALB/c mice with MV140/V132 enhances splenic T cell responses and induces specific antibodies against all the components included in the formulation. (A) Scheme of the sublingual immunization protocol and analysis of induced systemic responses. (B) Proliferation of CFSE-labeled CD4+ T cells from splenocytes isolated from mice immunized sublingually with MV140/V132 or control excipients after in vitro stimulation with V132, MV140, V140/V132, phytohaemagglutinin (PHA), or control (containing all excipients except the bacteria and yeast). (C) Serum IgG and IgA antibodies specific for (C)albicans, K pneumoniae, E coli, E faecalis and P. vulgaris from mice immunized with MV140/V132 or with control excipients. (D) Fold change of specific IgG and IgA antibodies generated in mice immunized with MV140/V132 relative to control mice. (E) Fold change of specific IgG and IgA antibodies for K pneumoniae, E coli, E faecalis and P. vulgaris generated in mice immunized with MV140/V132 or MV140 alone relative to control mice. (F) Fold change of specific IgG and IgA antibodies for C albicans generated in mice immunized with MV140/V132 or V132 alone relative to control mice. Results are mean ± s.e.m. of 7-8 (B) and 5-6 (C–F) individual mice per condition of one single experiment. Unpaired t test, *P < 0.05, **P < 0.01, and ***P < 0.001.

References

    1. Klein RD, Hultgren SJ. Urinary tract infections: microbial pathogenesis, host-pathogen interactions and new treatment strategies. Nat Rev Microbiol (2020) 18(4):211–26. 10.1038/s41579-020-0324-0
    1. Medina M, Castillo-Pino E. An introduction to the epidemiology and burden of urinary tract infections. Ther Adv Urol (2019) 11:1756287219832172. 10.1177/1756287219832172
    1. Flores-Mireles AL, Walker JN, Caparon M, Hultgren SJ. Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat Rev Microbiol (2015) 13(5):269–84. 10.1038/nrmicro3432
    1. Foxman B. Urinary tract infection syndromes: occurrence, recurrence, bacteriology, risk factors, and disease burden. Infect Dis Clin North Am (2014) 28(1):1–13. 10.1016/j.idc.2013.09.003
    1. Dason S, Dason JT, Kapoor A. Guidelines for the diagnosis and management of recurrent urinary tract infection in women. Can Urol Assoc J (2011) 5(5):316–22. 10.5489/cuaj.11214
    1. Laxminarayan R, Van Boeckel T, Frost I, Kariuki S, Khan EA, Limmathurotsakul D, et al. The Lancet Infectious Diseases Commission on antimicrobial resistance: 6 years later. Lancet Infect Dis (2020) 20(4):e51–60. 10.1016/S1473-3099(20)30003-7
    1. Macklaim JM, Clemente JC, Knight R, Gloor GB, Reid G. Changes in vaginal microbiota following antimicrobial and probiotic therapy. Microb Ecol Health Dis (2015) 26:27799. 10.3402/mehd.v26.27799
    1. Goncalves B, Ferreira C, Alves CT, Henriques M, Azeredo J, Silva S. Vulvovaginal candidiasis: Epidemiology, microbiology and risk factors. Crit Rev Microbiol (2016) 42(6):905–27. 10.3109/1040841X.2015.1091805
    1. Shukla A, Sobel JD. Vulvovaginitis Caused by Candida Species Following Antibiotic Exposure. Curr Infect Dis Rep (2019) 21(11):44. 10.1007/s11908-019-0700-y
    1. Xu J, Schwartz K, Bartoces M, Monsur J, Severson RK, Sobel JD. Effect of antibiotics on vulvovaginal candidiasis: a MetroNet study. J Am Board Fam Med (2008) 21(4):261–8. 10.3122/jabfm.2008.04.070169
    1. McCool L, Mai H, Essmann M, Larsen B. Tetracycline effects on Candida albicans virulence factors. Infect Dis Obstet Gynecol (2008) 2008:493508. 10.1155/2008/493508
    1. Wilton L, Kollarova M, Heeley E, Shakir S. Relative risk of vaginal candidiasis after use of antibiotics compared with antidepressants in women: postmarketing surveillance data in England. Drug Saf (2003) 26(8):589–97. 10.2165/00002018-200326080-00005
    1. Sobel JD. Vulvovaginal candidosis. Lancet (2007) 369(9577):1961–71. 10.1016/S0140-6736(07)60917-9
    1. Sobel JD. Recurrent vulvovaginal candidiasis. Am J Obstet Gynecol (2016) 214(1):15–21. 10.1016/j.ajog.2015.06.067
    1. Guzel AB, Ilkit M, Akar T, Burgut R, Demir SC. Evaluation of risk factors in patients with vulvovaginal candidiasis and the value of chromID Candida agar versus CHROMagar Candida for recovery and presumptive identification of vaginal yeast species. Med Mycol (2011) 49(1):16–25. 10.3109/13693786.2010.497972
    1. Denning DW, Kneale M, Sobel JD, Rautemaa-Richardson R. Global burden of recurrent vulvovaginal candidiasis: a systematic review. Lancet Infect Dis (2018) 18(11):e339–e47. 10.1016/S1473-3099(18)30103-8
    1. Yano J, Sobel JD, Nyirjesy P, Sobel R, Williams VL, Yu Q, et al. Current patient perspectives of vulvovaginal candidiasis: incidence, symptoms, management and post-treatment outcomes. BMC Womens Health (2019) 19(1):48. 10.1186/s12905-019-0748-8
    1. Crouss T, Sobel JD, Smith K, Nyirjesy P. Long-Term Outcomes of Women With Recurrent Vulvovaginal Candidiasis After a Course of Maintenance Antifungal Therapy. J Low Genit Tract Dis (2018) 22(4):382–6. 10.1097/LGT.0000000000000413
    1. Marchaim D, Lemanek L, Bheemreddy S, Kaye KS, Sobel JD. Fluconazole-resistant Candida albicans vulvovaginitis. Obstet Gynecol (2012) 120(6):1407–14. 10.1097/AOG.0b013e31827307b2
    1. Wagenlehner F, Wullt B, Ballarini S, Zingg D, Naber KG. Social and economic burden of recurrent urinary tract infections and quality of life: a patient web-based study (GESPRIT). Expert Rev Pharmacoecon Outcomes Res (2018) 18(1):107–17. 10.1080/14737167.2017.1359543
    1. Fukazawa EI, Witkin SS, Robial R, Vinagre JG, Baracat EC, Linhares IM. Influence of recurrent vulvovaginal candidiasis on quality of life issues. Arch Gynecol Obstet (2019) 300(3):647–50. 10.1007/s00404-019-05228-3
    1. Loubet P, Ranfaing J, Dinh A, Dunyach-Remy C, Bernard L, Bruyere F, et al. Alternative Therapeutic Options to Antibiotics for the Treatment of Urinary Tract Infections. Front Microbiol (2020) 11:1509. 10.3389/fmicb.2020.01509
    1. Sanchez-Ramon S, Conejero L, Netea MG, Sancho D, Palomares O, Subiza JL. Trained Immunity-Based Vaccines: A New Paradigm for the Development of Broad-Spectrum Anti-infectious Formulations. Front Immunol (2018) 9:2936. 10.3389/fimmu.2018.02936
    1. Tejera-Alhambra M, Palomares O, Perez de Diego R, Diaz-Lezcano I, Sanchez-Ramon S. New Biological Insights in the Immunomodulatory Effects of Mucosal Polybacterial Vaccines in Clinical Practice. Curr Pharm Des (2016) 22(41):6283–93. 10.2174/1381612822666160829143129
    1. Tso GHW, Reales-Calderon JA, Pavelka N. The Elusive Anti-Candida Vaccine: Lessons From the Past and Opportunities for the Future. Front Immunol (2018) 9:897. 10.3389/fimmu.2018.00897
    1. Benito-Villalvilla C, Cirauqui C, Diez-Rivero CM, Casanovas M, Subiza JL, Palomares O. MV140, a sublingual polyvalent bacterial preparation to treat recurrent urinary tract infections, licenses human dendritic cells for generating Th1, Th17, and IL-10 responses via Syk and MyD88. Mucosal Immunol (2017) 10(4):924–35. 10.1038/mi.2016.112
    1. Cirauqui C, Benito-Villalvilla C, Sanchez-Ramon S, Sirvent S, Diez-Rivero CM, Conejero L, et al. Human dendritic cells activated with MV130 induce Th1, Th17 and IL-10 responses via RIPK2 and MyD88 signalling pathways. Eur J Immunol (2018) 48(1):180–93. 10.1002/eji.201747024
    1. Guevara-Hoyer K, Saz-Leal P, Diez-Rivero CM, Ochoa-Grullon J, Fernandez-Arquero M, Perez de Diego R, et al. Trained Immunity Based-Vaccines as a Prophylactic Strategy in Common Variable Immunodeficiency. A Proof of Concept Study. Biomedicines (2020) 8(7):203. 10.3390/biomedicines8070203
    1. Netea MG, Quintin J, van der Meer JW. Trained immunity: a memory for innate host defense. Cell Host Microbe (2011) 9(5):355–61. 10.1016/j.chom.2011.04.006
    1. Quintin J, Saeed S, Martens JHA, Giamarellos-Bourboulis EJ, Ifrim DC, Logie C, et al. Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe (2012) 12(2):223–32. 10.1016/j.chom.2012.06.006
    1. Cheng SC, Quintin J, Cramer RA, Shepardson KM, Saeed S, Kumar V, et al. mTOR- and HIF-1alpha-mediated aerobic glycolysis as metabolic basis for trained immunity. Science (2014) 345(6204):1250684. 10.1126/science.1250684
    1. Saeed S, Quintin J, Kerstens HH, Rao NA, Aghajanirefah A, Matarese F, et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science (2014) 345(6204):1251086. 10.1126/science.1251086
    1. Lamichhane A, Azegamia T, Kiyonoa H. The mucosal immune system for vaccine development. Vaccine (2014) 32(49):6711–23. 10.1016/j.vaccine.2014.08.089
    1. Palomares O, Akdis M, Martin-Fontecha M, Akdis CA. Mechanisms of immune regulation in allergic diseases: the role of regulatory T and B cells. Immunol Rev (2017) 278(1):219–36. 10.1111/imr.12555
    1. Wculek SK, Khouili SC, Priego E, Heras-Murillo I, Sancho D. Metabolic Control of Dendritic Cell Functions: Digesting Information. Front Immunol (2019) 10:775. 10.3389/fimmu.2019.00775
    1. O’Neill LA, Kishton RJ. Rathmell J. A guide to immunometabolism for immunologists. Nat Rev Immunol (2016) 16(9):553–65. 10.1038/nri.2016.70
    1. Benito-Villalvilla C, Soria I, Perez-Diego M, Fernandez-Caldas E, Subiza JL, Palomares O. Alum impairs tolerogenic properties induced by allergoid-mannan conjugates inhibiting mTOR and metabolic reprogramming in human DCs. Allergy (2020) 75(3):648–59. 10.1111/all.14036
    1. Nickel JC, Saz-Leal P, Doiron RC. Could sublingual vaccination be a viable option for the prevention of recurrent urinary tract infection in Canada? A systematic review of the current literature and plans for the future. Can Urol Assoc J (2020) 14(8):281–7. 10.5489/cuaj.6690
    1. Lorenzo-Gomez MF, Padilla-Fernandez B, Garcia-Criado FJ, Miron-Canelo JA, Gil-Vicente A, Nieto-Huertos A, et al. Evaluation of a therapeutic vaccine for the prevention of recurrent urinary tract infections versus prophylactic treatment with antibiotics. Int Urogynecol J (2013) 24(1):127–34. 10.1007/s00192-012-1853-5
    1. Lorenzo-Gomez MF, Padilla-Fernandez B, Garcia-Cenador MB, Virseda-Rodriguez AJ, Martin-Garcia I, Sanchez-Escudero A, et al. Comparison of sublingual therapeutic vaccine with antibiotics for the prophylaxis of recurrent urinary tract infections. Front Cell Infect Microbiol (2015) 5:50. 10.3389/fcimb.2015.00050
    1. Yang B, Foley S. First experience in the UK of treating women with recurrent urinary tract infections with the bacterial vaccine Uromune((R)). BJU Int (2018) 121(2):289–92. 10.1111/bju.14067
    1. Ramirez Sevilla C, Gomez Lanza E, Manzanera JL, Martin JAR, Sanz MAB. Active immunoprophyilaxis with uromune(R) decreases the recurrence of urinary tract infections at three and six months after treatment without relevant secondary effects. BMC Infect Dis (2019) 19(1):901. 10.1186/s12879-019-4541-y
    1. Carrion-Lopez P, Martinez-Ruiz J, Libran-Garcia L, Gimenez-Bachs JM, Pastor-Navarro H, Salinas-Sanchez AS. Analysis of the Efficacy of a Sublingual Bacterial Vaccine in the Prophylaxis of Recurrent Urinary Tract Infection. Urol Int (2020) 104(3-4):293–300. 10.1159/000505162
    1. Netea MG, Brown GD, Kullberg BJ, Gow NA. An integrated model of the recognition of Candida albicans by the innate immune system. Nat Rev Microbiol (2008) 6(1):67–78. 10.1038/nrmicro1815
    1. Malinarich F, Duan K, Hamid RA, Bijin A, Lin WX, Poidinger M, et al. High mitochondrial respiration and glycolytic capacity represent a metabolic phenotype of human tolerogenic dendritic cells. J Immunol (2015) 194(11):5174–86. 10.4049/jimmunol.1303316
    1. Edwards JE, Jr., Schwartz MM, Schmidt CS, Sobel JD, Nyirjesy P, Schodel F, et al. A Fungal Immunotherapeutic Vaccine (NDV-3A) for Treatment of Recurrent Vulvovaginal Candidiasis-A Phase 2 Randomized, Double-Blind, Placebo-Controlled Trial. Clin Infect Dis (2018) 66(12):1928–36. 10.1093/cid/ciy185
    1. De Bernardis F, Amacker M, Arancia S, Sandini S, Gremion C, Zurbriggen R, et al. A virosomal vaccine against candidal vaginitis: immunogenicity, efficacy and safety profile in animal models. Vaccine (2012) 30(30):4490–8. 10.1016/j.vaccine.2012.04.069
    1. Torosantucci A, Bromuro C, Chiani P, De Bernardis F, Berti F, Galli C, et al. A novel glyco-conjugate vaccine against fungal pathogens. J Exp Med (2005) 202(5):597–606. 10.1084/jem.20050749
    1. Cardenas-Freytag L, Cheng E, Mayeux P, Domer JE, Clements JD. Effectiveness of a vaccine composed of heat-killed Candida albicans and a novel mucosal adjuvant, LT(R192G), against systemic candidiasis. Infect Immun (1999) 67(2):826–33. 10.1128/IAI.67.2.826-833.1999
    1. Akdis M, Aab A, Altunbulakli C, Azkur K, Costa RA, Crameri R, et al. Interleukins (from IL-1 to IL-38), interferons, transforming growth factor beta, and TNF-alpha: Receptors, functions, and roles in diseases. J Allergy Clin Immunol (2016) 138(4):984–1010. 10.1016/j.jaci.2016.06.033
    1. DuPage M, Bluestone JA. Harnessing the plasticity of CD4(+) T cells to treat immune-mediated disease. Nat Rev Immunol (2016) 16(3):149–63. 10.1038/nri.2015.18
    1. Palomares O, Martin-Fontecha M, Lauener R, Traidl-Hoffmann C, Cavkaytar O, Akdis M, et al. Regulatory T cells and immune regulation of allergic diseases: roles of IL-10 and TGF-beta. Genes Immun (2014) 15(8):511–20. 10.1038/gene.2014.45
    1. Soerens AG, Da Costa A, Lund JM. Regulatory T cells are essential to promote proper CD4 T-cell priming upon mucosal infection. Mucosal Immunol (2016) 9(6):1395–406. 10.1038/mi.2016.19
    1. Pandiyan P, Bhaskaran N, Zou M, Schneider E, Jayaraman S, Huehn J. Microbiome Dependent Regulation of Tregs and Th17 Cells in Mucosa. Front Immunol (2019) 10:426. 10.3389/fimmu.2019.00426
    1. Boyaka PN. Inducing Mucosal IgA: A Challenge for Vaccine Adjuvants and Delivery Systems. J Immunol (2017) 199(1):9–16. 10.4049/jimmunol.1601775
    1. O’Neill LA, Pearce EJ. Immunometabolism governs dendritic cell and macrophage function. J Exp Med (2016) 213(1):15–23. 10.1084/jem.20151570
    1. Ferreira GB, Vanherwegen AS, Eelen G, Gutierrez ACF, Van Lommel L, Marchal K, et al. Vitamin D3 Induces Tolerance in Human Dendritic Cells by Activation of Intracellular Metabolic Pathways. Cell Rep (2015) 10(5):711–25. 10.1016/j.celrep.2015.01.013
    1. Fliesser M, Morton CO, Bonin M, Ebel F, Hunniger K, Kurzai O, et al. Hypoxia-inducible factor 1alpha modulates metabolic activity and cytokine release in anti-Aspergillus fumigatus immune responses initiated by human dendritic cells. Int J Med Microbiol (2015) 305(8):865–73. 10.1016/j.ijmm.2015.08.036
    1. Sukhbaatar N, Hengstschlager M, Weichhart T. mTOR-Mediated Regulation of Dendritic Cell Differentiation and Function. Trends Immunol (2016) 37(11):778–89. 10.1016/j.it.2016.08.009
    1. Ifrim DC, Quintin J, Joosten LA, Jacobs C, Jansen T, Jacobs L, et al. Trained immunity or tolerance: opposing functional programs induced in human monocytes after engagement of various pattern recognition receptors. Clin Vaccine Immunol (2014) 21(4):534–45. 10.1128/CVI.00688-13
    1. Jantsch J, Chakravortty D, Turza N, Prechtel AT, Buchholz B, Gerlach RG, et al. Hypoxia and hypoxia-inducible factor-1 alpha modulate lipopolysaccharide-induced dendritic cell activation and function. J Immunol (2008) 180(7):4697–705. 10.4049/jimmunol.180.7.4697
    1. Arts RJ, Joosten LA, Netea MG. Immunometabolic circuits in trained immunity. Semin Immunol (2016) 28(5):425–30. 10.1016/j.smim.2016.09.002
    1. Tserel L, Kolde R, Rebane A, Kisand K, Org T, Peterson H, et al. Genome-wide promoter analysis of histone modifications in human monocyte-derived antigen presenting cells. BMC Genomics (2010) 11:642. 10.1186/1471-2164-11-642
    1. Mulder WJM, Ochando J, Joosten LAB, Fayad ZA, Netea MG. Therapeutic targeting of trained immunity. Nat Rev Drug Discov (2019) 18(7):553–66. 10.1038/s41573-019-0025-4

Source: PubMed

3
Předplatit