#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Extracellular vesicles in human semen modulate antigen-presenting cell function and decrease downstream antiviral T cell responses


Autoři: Lucia Vojtech aff001;  Mengying Zhang aff002;  Veronica Davé aff003;  Claire Levy aff001;  Sean M. Hughes aff001;  Ruofan Wang aff001;  Fernanda Calienes aff001;  Martin Prlic aff003;  Elizabeth Nance aff002;  Florian Hladik aff001
Působiště autorů: Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America aff001;  Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington, United States of America aff002;  Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America aff003;  Department of Global Health, University of Washington, Seattle, Washington, United states of America aff004;  Department of Immunology, University of Washington, Seattle, Washington, United States of America aff005;  Department of Chemical Engineering, University of Washington, Seattle, Washington, United States of America aff006;  Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, United States of America aff007
Vyšlo v časopise: PLoS ONE 14(10)
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pone.0223901

Souhrn

Human semen contains trillions of extracellular vesicles (SEV) similar in size to sexually transmitted viruses and loaded with potentially bioactive miRNAs, proteins and lipids. SEV were shown to inhibit HIV and Zika virus infectivity, but whether SEV are able also to affect subsequent immune responses is unknown. We found that SEV efficiently bound to and entered antigen-presenting cells (APC) and thus we set out to further dissect the impact of SEV on APC function and the impact on downstream T cell responses. In an APC–T cell co-culture system, SEV exposure to APC alone markedly reduced antigen-specific cytokine production, degranulation and cytotoxicity by antigen-specific memory CD8+ T cells. In contrast, inhibition of CD4+ T cell responses required both APC and T cell exposure to SEV. Surprisingly, SEV did not alter MHC or co-stimulatory receptor expression on APCs, but caused APCs to upregulate indoleamine 2,3 deoxygenase, an enzyme known to indirectly inhibit T cells. Thus, SEV reduce the ability of APCs to activate T cells. We propose here that these immune-inhibitory properties of SEV may be intended to prevent immune responses against semen-derived antigens, but can be hi-jacked by genitally acquired viral infections to compromise adaptive cellular immunity.

Klíčová slova:

Antigen-presenting cells – Cell staining – Cytokines – Cytotoxic T cells – Flow cytometry – Immune response – Semen – T cells


Zdroje

1. Sharkey DJ, Tremellen KP, Jasper MJ, Gemzell-Danielsson K, Robertson SA. Seminal fluid induces leukocyte recruitment and cytokine and chemokine mRNA expression in the human cervix after coitus. J Immunol. 2012;188(5):2445–54. Epub 2012/01/25. doi: 10.4049/jimmunol.1102736 22271649.

2. Remes Lenicov F, Varese A, Merlotti A, Geffner J, Ceballos A. Prostaglandins in semen compromise the immune response against sexually transmitted pathogens. Med Hypotheses. 2014;83(2):208–10. Epub 2014/05/20. doi: 10.1016/j.mehy.2014.04.028 24837236.

3. Doncel GF, Joseph T, Thurman AR. Role of semen in HIV-1 transmission: inhibitor or facilitator? Am J Reprod Immunol. 2011;65(3):292–301. doi: 10.1111/j.1600-0897.2010.00931.x 21087339

4. Rametse CL, Olivier AJ, Masson L, Barnabas S, McKinnon LR, Ngcapu S, et al. Role of semen in altering the balance between inflammation and tolerance in the female genital tract: does it contribute to HIV risk? Viral Immunol. 2014;27(5):200–6. Epub 2014/05/14. doi: 10.1089/vim.2013.0136 24821528.

5. Introini A, Bostrom S, Bradley F, Gibbs A, Glaessgen A, Tjernlund A, et al. Seminal plasma induces inflammation and enhances HIV-1 replication in human cervical tissue explants. PLoS Pathog. 2017;13(5):e1006402. Epub 2017/05/26. doi: 10.1371/journal.ppat.1006402 28542587; PubMed Central PMCID: PMC5453613.

6. Rametse CL, Adefuye AO, Olivier AJ, Curry L, Gamieldien H, Burgers WA, et al. Inflammatory Cytokine Profiles of Semen Influence Cytokine Responses of Cervicovaginal Epithelial Cells. Front Immunol. 2018;9:2721. Epub 2018/12/21. doi: 10.3389/fimmu.2018.02721 30568652; PubMed Central PMCID: PMC6290331.

7. Robertson SA, Guerin LR, Bromfield JJ, Branson KM, Ahlström AC, Care AS. Seminal fluid drives expansion of the CD4+CD25+ T regulatory cell pool and induces tolerance to paternal alloantigens in mice. Biol Reprod. 2009;80(5):1036–45. doi: 10.1095/biolreprod.108.074658 19164169

8. Kim BJ, Choi YM, Rah SY, Park DR, Park SA, Chung YJ, et al. Seminal CD38 is a pivotal regulator for fetomaternal tolerance. Proc Natl Acad Sci U S A. 2015;112(5):1559–64. Epub 2015/01/17. doi: 10.1073/pnas.1413493112 25591581; PubMed Central PMCID: PMC4321276.

9. Remes Lenicov F, Rodriguez Rodrigues C, Sabatte J, Cabrini M, Jancic C, Ostrowski M, et al. Semen promotes the differentiation of tolerogenic dendritic cells. J Immunol. 2012;189(10):4777–86. Epub 2012/10/16. doi: 10.4049/jimmunol.1202089 23066152.

10. Sharkey DJ, Macpherson AM, Tremellen KP, Mottershead DG, Gilchrist RB, Robertson SA. TGF-beta mediates proinflammatory seminal fluid signaling in human cervical epithelial cells. J Immunol. 2012;189(2):1024–35. doi: 10.4049/jimmunol.1200005 22706080.

11. Vojtech L, Woo S, Hughes S, Levy C, Ballweber L, Sauteraud RP, et al. Exosomes in human semen carry a distinctive repertoire of small non-coding RNAs with potential regulatory functions. Nucleic Acids Res. 2014;42(11):7290–304. Epub 2014/05/20. doi: 10.1093/nar/gku347 24838567; PubMed Central PMCID: PMC4066774.

12. Arienti G, Carlini E, Saccardi C, Palmerini CA. Interactions between prostasomes and leukocytes. Biochim Biophys Acta. 1998;1425(1):36–40. doi: 10.1016/s0304-4165(98)00047-6 9813232.

13. Kelly RW, Holland P, Skibinski G, Harrison C, McMillan L, Hargreave T, et al. Extracellular organelles (prostasomes) are immunosuppressive components of human semen. Clin Exp Immunol. 1991;86(3):550–6. doi: 10.1111/j.1365-2249.1991.tb02968.x 1747961.

14. Ronquist G, Brody I. The prostasome: its secretion and function in man. Biochim Biophys Acta. 1985;822(2):203–18. Epub 1985/09/09. doi: 10.1016/0304-4157(85)90008-5 2992593.

15. Yanez-Mo M, Siljander PR, Andreu Z, Zavec AB, Borras FE, Buzas EI, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066. Epub 2015/05/17. doi: 10.3402/jev.v4.27066 25979354; PubMed Central PMCID: PMC4433489.

16. Kitamura M, Namiki M, Matsumiya K, Tanaka K, Matsumoto M, Hara T, et al. Membrane cofactor protein (CD46) in seminal plasma is a prostasome-bound form with complement regulatory activity and measles virus neutralizing activity. Immunology. 1995;84(4):626–32. 7790037

17. Tarazona R, Delgado E, Guarnizo MC, Roncero RG, Morgado S, Sánchez-Correa B, et al. Human prostasomes express CD48 and interfere with NK cell function. Immunobiology. 2011;216(1–2):41–6. doi: 10.1016/j.imbio.2010.03.002 20382443

18. Ia Rooney, Atkinson JP, Krul ES, Schonfeld G, Polakoski K, Saffitz JE, et al. Physiologic relevance of the membrane attack complex inhibitory protein CD59 in human seminal plasma: CD59 is present on extracellular organelles (prostasomes), binds cell membranes, and inhibits complement-mediated lysis. The Journal of experimental medicine. 1993;177(5):1409–20. doi: 10.1084/jem.177.5.1409 7683035

19. Selva KJ, Juno JA, Worley MJ, Chung AW, Tachedjian G, Kent SJ, et al. Effect of seminal plasma on functions of monocytes and granulocytes. AIDS Res Hum Retroviruses. 2019. Epub 2019/05/01. doi: 10.1089/aid.2018.0219 31037950.

20. Muller M, Sales KJ, Katz AA, Jabbour HN. Seminal plasma promotes the expression of tumorigenic and angiogenic genes in cervical adenocarcinoma cells via the E-series prostanoid 4 receptor. Endocrinology. 2006;147(7):3356–65. Epub 2006/04/01. doi: 10.1210/en.2005-1429 16574793.

21. Zhao X, Deak E, Soderberg K, Linehan M, Spezzano D, Zhu J, et al. Vaginal submucosal dendritic cells, but not Langerhans cells, induce protective Th1 responses to herpes simplex virus-2. J Exp Med. 2003;197(2):153–62. doi: 10.1084/jem.20021109 12538655.

22. Welch JL, Kaddour H, Schlievert PM, Stapleton JT, Okeoma CM. Semen Exosomes Promote Transcriptional Silencing of HIV-1 by Disrupting NF-kappaB/Sp1/Tat Circuitry. J Virol. 2018;92(21). Epub 2018/08/17. doi: 10.1128/jvi.00731-18 30111566; PubMed Central PMCID: PMC6189507.

23. Lamparski HG, Metha-Damani A, Yao JY, Patel S, Hsu DH, Ruegg C, et al. Production and characterization of clinical grade exosomes derived from dendritic cells. J Immunol Methods. 2002;270(2):211–26. doi: 10.1016/s0022-1759(02)00330-7 12379326.

24. Ballweber L, Robinson B, Kreger A, Fialkow M, Lentz G, McElrath MJ, et al. Vaginal Langerhans cells nonproductively transporting HIV-1 mediate infection of T cells. J Virol. 2011;85(24):13443–7. Epub 2011/10/07. JVI.05615-11 [pii] doi: 10.1128/JVI.05615-11 21976645; PubMed Central PMCID: PMC3233146.

25. Horton H, Thomas EP, Stucky JA, Frank I, Moodie Z, Huang Y, et al. Optimization and validation of an 8-color intracellular cytokine staining (ICS) assay to quantify antigen-specific T cells induced by vaccination. J Immunol Methods. 2007;323(1):39–54. Epub 2007/04/25. S0022-1759(07)00089-0 [pii] doi: 10.1016/j.jim.2007.03.002 17451739; PubMed Central PMCID: PMC2683732.

26. McElrath MJ, De Rosa SC, Moodie Z, Dubey S, Kierstead L, Janes H, et al. HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case-cohort analysis. Lancet. 2008;372(9653):1894–905. doi: 10.1016/S0140-6736(08)61592-5 19012957.

27. O'Neill DW, Bhardwaj N. Differentiation of peripheral blood monocytes into dendritic cells. Curr Protoc Immunol. 2005;Chapter 22:Unit 22F.4. Epub 2008/04/25. doi: 10.1002/0471142735.im22f04s67 18432951.

28. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3(6):1101–8. Epub 2008/06/13. 18546601.

29. Betts MR, Brenchley JM, Price DA, De Rosa SC, Douek DC, Roederer M, et al. Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. J Immunol Methods. 2003;281(1–2):65–78. Epub 2003/10/29. S0022175903002655 [pii]. doi: 10.1016/s0022-1759(03)00265-5 14580882.

30. Pendergrass PB, Belovicz MW, Reeves CA. Surface area of the human vagina as measured from vinyl polysiloxane casts. Gynecol Obstet Invest. 2003;55(2):110–3. doi: 10.1159/000070184 12771458.

31. Barnhart KT, Izquierdo A, Pretorius ES, Shera DM, Shabbout M, Shaunik A. Baseline dimensions of the human vagina. Human Reproduction. 2006;21(6):1618–22. doi: 10.1093/humrep/del022 16478763

32. Evers H, Birngruber CG, Ramsthaler F, Muller U, Bruck S, Verhoff MA. [Differentiation of epithelial cell types by cell diameter]. Arch Kriminol. 2011;228(1–2):11–9. Epub 2011/08/20. 21850882.

33. Keller S, Konig AK, Marme F, Runz S, Wolterink S, Koensgen D, et al. Systemic presence and tumor-growth promoting effect of ovarian carcinoma released exosomes. Cancer Lett. 2009;278(1):73–81. Epub 2009/02/04. doi: 10.1016/j.canlet.2008.12.028 19188015.

34. Arienti G, Carlini E, Polci A, Cosmi EV, Palmerini CA. Fatty acid pattern of human prostasome lipid. Archives of biochemistry and biophysics. 1998;358(2):391–5. Epub 1998/10/24. doi: 10.1006/abbi.1998.0876 9784255.

35. Arvidson G, Ronquist G, Wikander G, Ojteg AC. Human prostasome membranes exhibit very high cholesterol/phospholipid ratios yielding high molecular ordering. Biochim Biophys Acta. 1989;984(2):167–73. Epub 1989/09/04. doi: 10.1016/0005-2736(89)90212-5 2548623.

36. Brouwers JF, Aalberts M, Jansen JW, van Niel G, Wauben MH, Stout TA, et al. Distinct lipid compositions of two types of human prostasomes. Proteomics. 2013;13(10–11):1660–6. Epub 2013/02/14. doi: 10.1002/pmic.201200348 23404715.

37. Hochreiter-Hufford A, Ravichandran KS. Clearing the dead: apoptotic cell sensing, recognition, engulfment, and digestion. Cold Spring Harb Perspect Biol. 2013;5(1):a008748. Epub 2013/01/04. doi: 10.1101/cshperspect.a008748 23284042; PubMed Central PMCID: PMC3579390.

38. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14(3):195–208. Epub 2014/02/26. doi: 10.1038/nri3622 24566916.

39. Kelleher RJ Jr., Balu-Iyer S, Loyall J, Sacca AJ, Shenoy GN, Peng P, et al. Extracellular Vesicles Present in Human Ovarian Tumor Microenvironments Induce a Phosphatidylserine-Dependent Arrest in the T-cell Signaling Cascade. Cancer Immunol Res. 2015;3(11):1269–78. Epub 2015/06/27. doi: 10.1158/2326-6066.CIR-15-0086 26112921; PubMed Central PMCID: PMC4636911.

40. Stites DP, Erickson RP. Suppressive effect of seminal plasma on lymphocyte activation. Nature. 1975;253(5494):727–9. doi: 10.1038/253727a0 123041.

41. Fraser JD. Clarifying the mechanism of superantigen toxicity. PLoS Biol. 2011;9(9):e1001145. Epub 2011/09/21. doi: 10.1371/journal.pbio.1001145 21931531; PubMed Central PMCID: PMC3172192.

42. Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev Immunol. 2009;27:591–619. Epub 2009/01/10. doi: 10.1146/annurev.immunol.021908.132706 19132916; PubMed Central PMCID: PMC2740335.

43. Aktas E, Kucuksezer UC, Bilgic S, Erten G, Deniz G. Relationship between CD107a expression and cytotoxic activity. Cell Immunol. 2009;254(2):149–54. Epub 2008/10/07. doi: 10.1016/j.cellimm.2008.08.007 18835598.

44. Harden JL, Egilmez NK. Indoleamine 2,3-Dioxygenase and Dendritic Cell Tolerogenicity. Immunol Invest. 2012;41(0):738–64. doi: 10.3109/08820139.2012.676122 23017144; PubMed Central PMCID: PMC3645912.

45. Corinti S, Albanesi C, la Sala A, Pastore S, Girolomoni G. Regulatory activity of autocrine IL-10 on dendritic cell functions. J Immunol. 2001;166(7):4312–8. Epub 2001/03/20. doi: 10.4049/jimmunol.166.7.4312 11254683.

46. Belladonna ML, Volpi C, Bianchi R, Vacca C, Orabona C, Pallotta MT, et al. Cutting Edge: Autocrine TGF-β Sustains Default Tolerogenesis by IDO-Competent Dendritic Cells. The Journal of Immunology. 2008;181(8):5194. doi: 10.4049/jimmunol.181.8.5194 18832670

47. Vendelova E, Ashour D, Blank P, Erhard F, Saliba AE, Kalinke U, et al. Tolerogenic Transcriptional Signatures of Steady-State and Pathogen-Induced Dendritic Cells. Front Immunol. 2018;9:333. Epub 2018/03/16. doi: 10.3389/fimmu.2018.00333 29541071; PubMed Central PMCID: PMC5835767.

48. Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells. Immunology. 2006;117(4):433–42. doi: 10.1111/j.1365-2567.2006.02321.x 16556256.

49. Madison MN, Jones PH, Okeoma CM. Exosomes in human semen restrict HIV-1 transmission by vaginal cells and block intravaginal replication of LP-BM5 murine AIDS virus complex. Virology. 2015;482:189–201. Epub 2015/04/17. doi: 10.1016/j.virol.2015.03.040 25880110; PubMed Central PMCID: PMC4461544.

50. Muller JA, Harms M, Kruger F, Gross R, Joas S, Hayn M, et al. Semen inhibits Zika virus infection of cells and tissues from the anogenital region. Nat Commun. 2018;9(1):2207. Epub 2018/06/09. doi: 10.1038/s41467-018-04442-y 29880824; PubMed Central PMCID: PMC5992203.

51. Santambrogio L, Sato AK, Fischer FR, Dorf ME, Stern LJ. Abundant empty class II MHC molecules on the surface of immature dendritic cells. Proc Natl Acad Sci U S A. 1999;96(26):15050–5. Epub 1999/12/28. doi: 10.1073/pnas.96.26.15050 10611336; PubMed Central PMCID: PMC24771.

52. York IA, Rock KL. Antigen processing and presentation by the class I major histocompatibility complex. Annu Rev Immunol. 1996;14:369–96. Epub 1996/01/01. doi: 10.1146/annurev.immunol.14.1.369 8717519.

53. Day PM, Yewdell JW, Porgador A, Germain RN, Bennink JR. Direct delivery of exogenous MHC class I molecule-binding oligopeptides to the endoplasmic reticulum of viable cells. Proc Natl Acad Sci U S A. 1997;94(15):8064–9. Epub 1997/07/22. doi: 10.1073/pnas.94.15.8064 9223315; PubMed Central PMCID: PMC21557.

54. Bevan MJ. Antigen recognition. Class discrimination in the world of immunology. Nature. 1987;325(6101):192–4. Epub 1987/01/15. doi: 10.1038/325192b0 2433584.

55. Handsfield HH, Chandler SH, Caine VA, Meyers JD, Corey L, Medeiros E, et al. Cytomegalovirus infection in sex partners: evidence for sexual transmission. J Infect Dis. 1985;151(2):344–8. Epub 1985/02/01. doi: 10.1093/infdis/151.2.344 2981937.

56. Thomas R, Macsween KF, McAulay K, Clutterbuck D, Anderson R, Reid S, et al. Evidence of shared Epstein-Barr viral isolates between sexual partners, and low level EBV in genital secretions. J Med Virol. 2006;78(9):1204–9. Epub 2006/07/19. doi: 10.1002/jmv.20682 16847960.

57. Kalinski P. Regulation of immune responses by prostaglandin E2. J Immunol. 2012;188(1):21–8. Epub 2011/12/22. doi: 10.4049/jimmunol.1101029 22187483; PubMed Central PMCID: PMC3249979.

58. Poloso NJ, Urquhart P, Nicolaou A, Wang J, Woodward DF. PGE2 differentially regulates monocyte-derived dendritic cell cytokine responses depending on receptor usage (EP2/EP4). Mol Immunol. 2013;54(3–4):284–95. Epub 2013/01/23. doi: 10.1016/j.molimm.2012.12.010 23337716.

59. Van Elssen CHMJ, Vanderlocht J, Oth T, Senden-Gijsbers BLMG, Germeraad WTV, Bos GMJ. Inflammation restraining effects of prostaglandin E2 on natural killer–dendritic cell (NK-DC) interaction are imprinted during DC maturation. Blood. 2011;118(9):2473. doi: 10.1182/blood-2010-09-307835 21715307

60. von Bergwelt-Baildon MS, Popov A, Saric T, Chemnitz J, Classen S, Stoffel MS, et al. CD25 and indoleamine 2,3-dioxygenase are up-regulated by prostaglandin E2 and expressed by tumor-associated dendritic cells in vivo: additional mechanisms of T-cell inhibition. Blood. 2006;108(1):228. doi: 10.1182/blood-2005-08-3507 16522817

61. Pallotta MT, Orabona C, Volpi C, Vacca C, Belladonna ML, Bianchi R, et al. Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells. Nat Immunol. 2011;12(9):870–8. Epub 2011/08/02. doi: 10.1038/ni.2077 21804557.

62. Kikete S, Chu X, Wang L, Bian Y. Endogenous and tumour-derived microRNAs regulate cross-presentation in dendritic cells and consequently cytotoxic T cell function. Cytotechnology. 2016. Epub 2016/05/20. doi: 10.1007/s10616-016-9975-0 27193424.

63. Haderk F, Schulz R, Iskar M, Cid LL, Worst T, Willmund KV, et al. Tumor-derived exosomes modulate PD-L1 expression in monocytes. Sci Immunol. 2017;2(13). Epub 2017/07/30. doi: 10.1126/sciimmunol.aah5509 28754746.

64. Birge RB, Boeltz S, Kumar S, Carlson J, Wanderley J, Calianese D, et al. Phosphatidylserine is a global immunosuppressive signal in efferocytosis, infectious disease, and cancer. Cell Death Differ. 2016. Epub 2016/02/27. doi: 10.1038/cdd.2016.11 26915293.

65. Moller-Tank S, Maury W. Phosphatidylserine receptors: enhancers of enveloped virus entry and infection. Virology. 2014;468–470:565–80. Epub 2014/10/04. doi: 10.1016/j.virol.2014.09.009 25277499; PubMed Central PMCID: PMC4252826.

66. Matsumoto A, Takahashi Y, Nishikawa M, Sano K, Morishita M, Charoenviriyakul C, et al. Role of Phosphatidylserine-Derived Negative Surface Charges in the Recognition and Uptake of Intravenously Injected B16BL6-Derived Exosomes by Macrophages. J Pharm Sci. 2016. Epub 2016/09/22. doi: 10.1016/j.xphs.2016.07.022 27649887.

67. Morelli AE, Larregina AT, Shufesky WJ, Sullivan ML, Stolz DB, Papworth GD, et al. Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood. 2004;104(10):3257–66. doi: 10.1182/blood-2004-03-0824 15284116.

68. Feng D, Zhao WL, Ye YY, Bai XC, Liu RQ, Chang LF, et al. Cellular internalization of exosomes occurs through phagocytosis. Traffic. 2010;11(5):675–87. Epub 2010/02/09. doi: 10.1111/j.1600-0854.2010.01041.x 20136776.

69. Allen RD, Roberts TK. The relationship between the immunosuppressive and cytotoxic effects of human seminal plasma. Am J Reprod Immunol Microbiol. 1986;11(2):59–64. Epub 1986/06/01. 3740349.

70. Kelly RW, Carr GG, Critchley HO. A cytokine switch induced by human seminal plasma: an immune modulation with implications for sexually transmitted disease. Hum Reprod. 1997;12(4):677–81. doi: 10.1093/humrep/12.4.677 9159423.

71. Kelly RW. Immunomodulators in human seminal plasma: a vital protection for spermatozoa in the presence of infection? International journal of andrology. 1999;22(1):2–12. doi: 10.1046/j.1365-2605.1999.00142.x 10068938

72. Robertson SA, Mau VJ, Hudson SN, Tremellen KP. Cytokine-leukocyte networks and the establishment of pregnancy. Am J Reprod Immunol. 1997;37(6):438–42. doi: 10.1111/j.1600-0897.1997.tb00257.x 9228299


Článek vyšel v časopise

PLOS One


2019 Číslo 10
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#