Hypothetical Immunological and Immunogenetic Model of Heterogenous Effects of BCG Vaccination in SARS-CoV-2 Infections: BCG-induced Trained and Heterologous Immunity

Authors

  • Dženan Kovačić International Burch University, Faculty of Engineering and Natural Sciences, Department of Genetics and Bioengineering, Sarajevo, Bosnia and Herzegovina; Sharklab ADRIA: Center for marine and frewater biology, Triq il-Kappella Tal-Lunzjata, In-Naxxar, Malta https://orcid.org/0000-0003-3218-5073
  • Andrej A. Gajić Shark Tales, Funded by the National Geographic Society, Washington, D.C., USA; Sharklab ADRIA: Center for marine and frewater biology, Triq il-Kappella Tal-Lunzjata, In-Naxxar, Malta https://orcid.org/0000-0001-5138-9368
  • Dado Latinović International Burch University, Faculty of Engineering and Natural Sciences, Department of Genetics and Bioengineering, Sarajevo, Bosnia and Herzegovina
  • Adna Softić International Burch University, Faculty of Engineering and Natural Sciences, Department of Genetics and Bioengineering, Sarajevo, Bosnia and Herzegovina https://orcid.org/0000-0002-4405-2916

DOI:

https://doi.org/10.20883/medical.e551

Keywords:

immunogenetics, immunology, trained immunity, heterologous immunity, BCG trained immunity, TLR signalling, COVID-19 and BCG

Abstract

Though SARS-CoV-2 infections are yet to be completely characterised in a host-pathogen interaction context, some of the mechanisms governing the interaction between the novel betacoronavirus and the human host, have been brought to light in satisfactory detail. Among the emerging evidence, postulates regarding potential benefits of innate immune memory and heterologous immunity have been put under discussion. Innate immune memory entails epigenetic reprogramming of innate immune cells caused by vaccination or infections, whereas heterologous immunity denotes cross-reactivity of T cells with unrelated epitopes and bystander CD8+ activation. Familiarization of the host immune system with a certain pathogen, educates monocytes, macrophages and other innate cells into phenotypes competent for combating unrelated pathogens. Indeed, the resolution at which non-specific innate immune memory occurs, is predominant at the level of enhanced cytokine secretion as a result of epigenetic alterations. One vaccine whose non-specific effects have been documented and harnessed in treating infections, cancer and autoimmunity, is the Bacillus Calmette–Guérin (BCG) vaccine currently used for immunization against pulmonary tuberculosis (TB). The BCG vaccine induces a diverse cytokine secretion profile in immunized subjects, which in turn may stimulate epigenetic changes mediated by immunoreceptor signalling. Herein, we provide a concise summarization of previous findings regarding the effects of the BCG vaccine on innate immune memory and heterologous immunity, supplemented with clinical evidence of the non-specific effects of this vaccine on non-mycobacterial infections, cancer and autoimmunity. This interpretative synthesis aims at providing a plausible immunological and immunogenetic model by which BCG vaccination may, in fact, be beneficial for the current efforts in combating COVID-19.

Downloads

Download data is not yet available.

References

LH. Hogan, DO. Co, J. Karman, E. Heninger, M. Suresh, and M. Sandor, “Virally activated CD8 T cells home to Mycobacterium bovis BCG-induced granulomas but enhance antimycobacterial protection only in immunodeficient mice,” Infection and Immunity, vol. 75, no. 3, pp. 1154–1166, Mar. 2007, doi: 10.1128/IAI.00943-06.

G. Lertmemongkolchai, G. Cai, CA. Hunter, and GJ. Bancroft, “Bystander Activation of CD8 + T Cells Contributes to the Rapid Production of IFN-γ in Response to Bacterial Pathogens,” The Journal of Immunology, vol. 166, no. 2, pp. 1097–1105, Jan. 2001, doi: 10.4049/jimmunol.166.2.1097.

B. Gilbertson, S. Germano, P. Steele, S. Turner, BF. Barbara, and C. Cheers, “Bystander activation of CD8+ T lymphocytes during experimental mycobacterial infection,” Infection and Immunity, vol. 72, no. 12, pp. 6884–6891, Dec. 2004, doi: 10.1128/IAI.72.12.6884-6891.2004.

J. Kim et al., “Innate-like Cytotoxic Function of Bystander-Activated CD8 + T Cells Is Associated with Liver Injury in Acute Hepatitis A,” Immunity, vol. 48, no. 1, pp. 161-173.e5, Jan. 2018, doi: 10.1016/j.immuni.2017.11.025.

DJ. Perkins, MC. Patel, JCG. Blanco, and SN. Vogel, “Epigenetic mechanisms governing innate inflammatory responses,” Journal of Interferon and Cytokine Research, vol. 36, no. 7, pp. 454–461, Jul. 2016, doi: 10.1089/jir.2016.0003.

B. Gourbal, S. Pinaud, GJM. Beckers, JWM. van der Meer, U. Conrath, and MG. Netea, “Innate immune memory: An evolutionary perspective,” Immunological Reviews, vol. 283, no. 1, pp. 21–40, May 2018, doi: 10.1111/imr.12647.

E. Töpfer, D. Boraschi, and P. Italiani, “Innate Immune Memory: The Latest Frontier of Adjuvanticity,” Journal of Immunology Research, vol. 2015, 2015, doi: 10.1155/2015/478408.

MG. Netea and R. van Crevel, “BCG-induced protection: Effects on innate immune memory,” Seminars in Immunology, vol. 26, no. 6, pp. 512–517, Dec. 2014, doi: 10.1016/j.smim.2014.09.006.

JD. van Belleghem and PL. Bollyky, “Macrophages and innate immune memory against Staphylococcus skin infections,” Proceedings of the National Academy of Sciences of the United States of America, vol. 115, no. 47, pp. 11865–11867, Nov. 2018, doi: 10.1073/pnas.1816935115.

D. Boraschi and P. Italiani, “Innate immune memory: Time for adopting a correct terminology,” Frontiers in Immunology, vol. 9, no. APR, p. 799, Apr. 2018, doi: 10.3389/fimmu.2018.00799.

VA.CM. Koeken, AJ. Verrall, MG. Netea, PC. Hill, and R. van Crevel, “Trained innate immunity and resistance to Mycobacterium tuberculosis infection,” Clinical Microbiology and Infection, vol. 25, no. 12. Elsevier BV., pp. 1468–1472, Dec. 01, 2019. doi: 10.1016/j.cmi.2019.02.015.

B. Pulendran and R. Ahmed, “Translating innate immunity into immunological memory: Implications for vaccine development,” Cell, vol. 124, no. 4, pp. 849–863, Feb. 2006, doi: 10.1016/j.cell.2006.02.019.

BL. Brady, NC. Steinel, and CH. Bassing, “Antigen Receptor Allelic Exclusion: An Update and Reappraisal,” The Journal of Immunology, vol. 185, no. 7, pp. 3801–3808, Oct. 2010, doi: 10.4049/jimmunol.1001158.

R. Levin-Klein and Y. Bergman, “Epigenetic regulation of monoallelic rearrangement (allelic exclusion) of antigen receptor genes,” Frontiers in Immunology, vol. 5, no. DEC, p. 625, Dec. 2014, doi: 10.3389/fimmu.2014.00625.

Y. Bergman and H. Cedar, “A stepwise epigenetic process controls immunoglobulin allelic exclusion,” Nature Reviews Immunology, vol. 4, no. 10, pp. 753–761, Oct. 2004, doi: 10.1038/nri1458.

P. Borst, “Antigenic variation and allelic exclusion,” Cell, vol. 109, no. 1, pp. 5–8, Apr. 2002, doi: 10.1016/S0092-8674(02)00711-0.

KW. Wucherpfennig et al., “Polyspecificity of T cell and B cell receptor recognition,” Seminars in Immunology, vol. 19, no. 4, pp. 216–224, Aug. 2007, doi: 10.1016/j.smim.2007.02.012.

M. Malissen, J. Trucy, E. Jouvin-Marche, PA. Cazenave, R. Scollay, and B. Malissen, “Regulation of TCR α and β gene allelic exclusion during T-cell development,” Immunology Today, vol. 13, no. 8, pp. 315–322, 1992, doi: 10.1016/0167-5699(92)90044-8.

AK. Sewell, “Why must T cells be cross-reactive?,” Nature Reviews Immunology, vol. 12, no. 9, pp. 669–677, Sep. 2012, doi: 10.1038/nri3279.

I. Santecchia et al., “Innate immune memory through TLR2 and NOD2 contributes to the control of Leptospira interrogans infection,” PLOS Pathogens, vol. 15, no. 5, p. e1007811, May 2019, doi: 10.1371/journal.ppat.1007811.

C. Yan and DD. Boyd, “Histone H3 Acetylation and H3 K4 Methylation Define Distinct Chromatin Regions Permissive for Transgene Expression,” Molecular and Cellular Biology, vol. 26, no. 17, pp. 6357–6371, Sep. 2006, doi: 10.1128/mcb.00311-06.

VR. Ramirez-Carrozzi et al., “Selective and antagonistic functions of SWI/SNF and Mi-2β nucleosome remodeling complexes during an inflammatory response,” Genes and Development, vol. 20, no. 3, pp. 282–296, Feb. 2006, doi: 10.1101/gad.1383206.

VR. Ramirez-Carrozzi et al., “A Unifying Model for the Selective Regulation of Inducible Transcription by CpG Islands and Nucleosome Remodeling,” Cell, vol. 138, no. 1, pp. 114–128, Jul. 2009, doi: 10.1016/j.cell.2009.04.020.

DC. Hargreaves, T. Horng, and R. Medzhitov, “Control of Inducible Gene Expression by Signal-Dependent Transcriptional Elongation,” Cell, vol. 138, no. 1, pp. 129–145, Jul. 2009, doi: 10.1016/j.cell.2009.05.047.

EL. Lousberg, CK. Fraser, MG. Tovey, KR. Diener, and JD. Hayball, “Type I Interferons Mediate the Innate Cytokine Response to Recombinant Fowlpox Virus but Not the Induction of Plasmacytoid Dendritic Cell-Dependent Adaptive Immunity,” Journal of Virology, vol. 84, no. 13, pp. 6549–6563, Jul. 2010, doi: 10.1128/jvi.02618-09.

R. Kamada et al., “Interferon stimulation creates chromatin marks and establishes transcriptional memory,” Proceedings of the National Academy of Sciences of the United States of America, vol. 115, no. 39, pp. E9162–E9171, Sep. 2018, doi: 10.1073/pnas.1720930115.

CM. Leopold Wager et al., “IFN-γ immune priming of macrophages in vivo induces prolonged STAT1 binding and protection against Cryptococcus neoformans,” PLoS Pathogens, vol. 14, no. 10, Oct. 2018, doi: 10.1371/journal.ppat.1007358.

MK. Lalor et al., “BCG vaccination induces different cytokine profiles following infant BCG vaccination in the UK and Malawi,” Journal of Infectious Diseases, vol. 204, no. 7, pp. 1075–1085, Oct. 2011, doi: 10.1093/infdis/jir515.

SJ.CFM. Moorlag, RJW. Arts, R. van Crevel, and MG. Netea, “Non-specific effects of BCG vaccine on viral infections,” Clinical Microbiology and Infection, vol. 25, no. 12. Elsevier BV., pp. 1473–1478, Dec. 01, 2019. doi: 10.1016/j.cmi.2019.04.020.

IA. Clark, AC. Allison, and FE. Cox, “Protection of mice against Babesia, and Plasmodium with BCG,” Nature, vol. 259, no. 5541, pp. 309–311, 1976, doi: 10.1038/259309a0.

LAJ. O’Neill and MG. Netea, “BCG-induced trained immunity: can it offer protection against COVID-19?,” Nature Reviews Immunology, vol. 20, no. 6. Nature Research, pp. 335–337, Jun. 01, 2020. doi: 10.1038/s41577-020-0337-y.

SM. Taghioff, BR. Slavin, T. Holton, and D. Singh, “Examining the potential benefits of the influenza vaccine against SARS-CoV-2: A retrospective cohort analysis of 74,754 patients,” PLOS ONE, vol. 16, no. 8, p. e0255541, Aug. 2021, doi: 10.1371/JOURNAL.PONE.0255541.

A. Conlon, C. Ashur, L. Washer, KA. Eagle, and MA. Hofmann Bowman, “Impact of the influenza vaccine on COVID-19 infection rates and severity,” American Journal of Infection Control, vol. 49, no. 6, pp. 694–700, Jun. 2021, doi: 10.1016/J.AJIC.2021.02.012.

K. Huang, SW. Lin, WH. Sheng, and CC. Wang, “Influenza vaccination and the risk of COVID-19 infection and severe illness in older adults in the United States,” Scientific Reports 2021 11:1, vol. 11, no. 1, pp. 1–6, May 2021, doi: 10.1038/s41598-021-90068-y.

FK. Föhse et al., “The BNT162b2 mRNA vaccine against SARS-CoV-2 reprograms both adaptive and innate immune responses,” medRxiv, p. 2021.05.03.21256520, May 2021, doi: 10.1101/2021.05.03.21256520.

A. Miller, MJ. Reandelar, K. Fasciglione, V. Roumenova, Y. Li, and GH. Otazu, “Correlation between universal BCG vaccination policy and reduced morbidity and mortality for COVID-19: an epidemiological study”, doi: 10.1101/2020.03.24.20042937.

S. Perlman and AA. Dandekar, “Immunopathogenesis of coronavirus infections: Implications for SARS,” Nature Reviews Immunology, vol. 5, no. 12. pp. 917–927, Dec. 01, 2005. doi: 10.1038/nri1732.

I. Glowacka, S. Bertram, and S. Pöhlmann, “Cellular Entry of the SARS Coronavirus: Implications for Transmission, Pathogenicity and Antiviral Strategies,” Molecular Biology of the Sars-coronavirus, pp. 3–22, Jul. 2009, doi: 10.1007/978-3-642-03683-5_1.

J. He, H. Tao, Y. Yan, S.-Y. Huang, and Y. Xiao, “Molecular Mechanism of Evolution and Human Infection with SARS-CoV-2,” Viruses, vol. 12, no. 4, p. 428, Apr. 2020, doi: 10.3390/v12040428.

VK. Shah, P. Firmal, A. Alam, D. Ganguly, and S. Chattopadhyay, “Overview of Immune Response During SARS-CoV-2 Infection: Lessons From the Past,” Frontiers in Immunology, vol. 11, p. 1949, Aug. 2020, doi: 10.3389/fimmu.2020.01949.

M. Letko, A. Marzi, and V. Munster, “Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses,” Nature Microbiology, vol. 5, no. 4, pp. 562–569, 2020, doi: 10.1038/s41564-020-0688-y.

R. Lu et al., “Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding,” www.thelancet.com, vol. 395, p. 565, 2020, doi: 10.1016/S0140-6736(20)30251-8.

L. Lin, L. Lu, W. Cao, and T. Li, “Hypothesis for potential pathogenesis of SARS-CoV-2 infection–a review of immune changes in patients with viral pneumonia,” Emerging Microbes & Infections, vol. 9, no. 1, pp. 727–732, Jan. 2020, doi: 10.1080/22221751.2020.1746199.

N. Zhu et al., “A novel coronavirus from patients with pneumonia in China, 2019,” New England Journal of Medicine, vol. 382, no. 8, pp. 727–733, Feb. 2020, doi: 10.1056/NEJMoa2001017.

D. Harmer, M. Gilbert, R. Borman, and KL. Clark, “Quantitative mRNA expression pro¢ling of ACE 2, a novel homologue of angiotensin converting enzyme.”

F. Ali, A. Kasry, and M. Amin, “The new SARS-CoV-2 strain shows a stronger binding affinity to ACE2 due to N501Y mutant,” Medicine in Drug Discovery, vol. 10, p. 100086, Jun. 2021, doi: 10.1016/J.MEDIDD.2021.100086.

J. Shang et al., “Structural basis of receptor recognition by SARS-CoV-2,” Nature, vol. 581, no. 7807, pp. 221–224, May 2020, doi: 10.1038/s41586-020-2179-y.

M. Murakami, D. Kamimura, and T. Hirano, “Pleiotropy and Specificity: Insights from the Interleukin 6 Family of Cytokines,” Immunity, vol. 50, no. 4, pp. 812–831, Apr. 2019, doi: 10.1016/j.immuni.2019.03.027.

F. Chiodo et al., “Novel ACE2-Independent Carbohydrate-Binding of SARS-CoV-2 Spike Protein to Host Lectins and Lung Microbiota,” bioRxiv, p. 2020.05.13.092478, May 2020, doi: 10.1101/2020.05.13.092478.

SA. Jeffers et al., “CD209L (L-SIGN) is a receptor for severe acute respiratory syndrome coronavirus,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 44, pp. 15748–15753, Nov. 2004, doi: 10.1073/pnas.0403812101.

R. Amraie et al., “CD209L/L-SIGN and CD209/DC-SIGN act as receptors for SARS-CoV-2 and are differentially expressed in lung and kidney epithelial and endothelial cells,” bioRxiv : the preprint server for biology, 2020, doi: 10.1101/2020.06.22.165803.

E. Prompetchara, C. Ketloy, and T. Palaga, “Immune responses in COVID-19 and potential vaccines: Lessons learned from SARS and MERS epidemic,” Asian Pacific Journal of Allergy and Immunology, 2020, doi: 10.12932/AP-200220-0772.

E. de Wit, N. van Doremalen, D. Falzarano, and VJ. Munster, “SARS and MERS: Recent insights into emerging coronaviruses,” Nature Reviews Microbiology, vol. 14, no. 8. Nature Publishing Group, pp. 523–534, Aug. 01, 2016. doi: 10.1038/nrmicro.2016.81.

T. Yoshikawa, T. Hill, K. Li, CJ. Peters, and C.-TK. Tseng, “Severe Acute Respiratory Syndrome (SARS) Coronavirus-Induced Lung Epithelial Cytokines Exacerbate SARS Pathogenesis by Modulating Intrinsic Functions of Monocyte-Derived Macrophages and Dendritic Cells,” Journal of Virology, vol. 83, no. 7, pp. 3039–3048, Apr. 2009, doi: 10.1128/jvi.01792-08.

J. Zhao, J. Zhao, K. Legge, and S. Perlman, “Age-related increases in PGD 2 expression impair respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice,” Journal of Clinical Investigation, vol. 121, no. 12, pp. 4921–4930, Dec. 2011, doi: 10.1172/JCI59777.

JS. Turner et al., “SARS-CoV-2 infection induces long-lived bone marrow plasma cells in humans,” Nature 2021 595:7867, vol. 595, no. 7867, pp. 421–425, May 2021, doi: 10.1038/s41586-021-03647-4.

Z. Wang et al., “Naturally enhanced neutralizing breadth against SARS-CoV-2 one year after infection,” Nature 2021 595:7867, vol. 595, no. 7867, pp. 426–431, Jun. 2021, doi: 10.1038/s41586-021-03696-9.

A. Sette and S. Crotty, “Adaptive immunity to SARS-CoV-2 and COVID-19,” Cell, vol. 184, no. 4, p. 861, Feb. 2021, doi: 10.1016/J.CELL.2021.01.007.

K. -Y Yuen et al., “Coronavirus Disease 2019 (COVID-19) Re-infection by a Phylogenetically Distinct Severe Acute Respiratory Syndrome Coronavirus 2 Strain Confirmed by Whole Genome Sequencing,” Clinical Infectious Diseases, vol. 73, no. 9, pp. e2946–e2951, Nov. 2021, doi: 10.1093/CID/CIAA1275.

RL. Tillett et al., “Genomic evidence for reinfection with SARS-CoV-2: a case study,” The Lancet Infectious Diseases, vol. 21, no. 1, pp. 52–58, Jan. 2021, doi: 10.1016/S1473-3099(20)30764-7.

R. Medzhitov and CA. Janeway, “Innate immunity: Impact on the adaptive immune response,” Current Opinion in Immunology, vol. 9, no. 1, pp. 4–9, Feb. 1997, doi: 10.1016/S0952-7915(97)80152-5.

L. Mohamed Khosroshahi, M. Rokni, T. Mokhtari, and F. Noorbakhsh, “Immunology, immunopathogenesis and immunotherapeutics of COVID-19; an overview,” International Immunopharmacology, vol. 93, p. 107364, Apr. 2021, doi: 10.1016/J.INTIMP.2020.107364.

WJ. Liu et al., “T-cell immunity of SARS-CoV: Implications for vaccine development against MERS-CoV,” Antiviral Research, vol. 137. Elsevier BV., pp. 82–92, Jan. 01, 2017. doi: 10.1016/j.antiviral.2016.11.006.

L. Cheng et al., “Dynamic landscape mapping of humoral immunity to SARS-CoV-2 identifies non-structural protein antibodies associated with the survival of critical COVID-19 patients,” Signal Transduction and Targeted Therapy 2021 6:1, vol. 6, no. 1, pp. 1–14, Aug. 2021, doi: 10.1038/s41392-021-00718-w.

P. Zhou et al., “A pneumonia outbreak associated with a new coronavirus of probable bat origin,” Nature, vol. 579, no. 7798, pp. 270–273, Mar. 2020, doi: 10.1038/s41586-020-2012-7.

J. Zhao et al., “Antibody responses to SARS-CoV-2 in patients of novel coronavirus disease 2019 Brief Title: Antibody responses in COVID-19 patients,” 2020, doi: 10.1101/2020.03.02.20030189.

B. Lou et al., “Serology characteristics of SARS-CoV-2 infection after exposure and post-symptom onset,” European Respiratory Journal, vol. 56, no. 2, Aug. 2020, doi: 10.1183/13993003.00763-2020.

R. Krajewski, J. Gołębiowska, S. Makuch, G. Mazur, and S. Agrawal, “Update on serologic testing in COVID–19,” Clinica Chimica Acta, vol. 510, pp. 746–750, Nov. 2020, doi: 10.1016/J.CCA.2020.09.015.

J. Zhao et al., “Antibody responses to SARS-CoV-2 in patients of novel coronavirus disease 2019,” Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America, vol. 71, no. 16, pp. 2027–2034, Oct. 2020, doi: 10.1093/CID/CIAA344.

W. Liu et al., “Two‐Year Prospective Study of the Humoral Immune Response of Patients with Severe Acute Respiratory Syndrome,” The Journal of Infectious Diseases, vol. 193, no. 6, pp. 792–795, Mar. 2006, doi: 10.1086/500469.

CK. Li et al., “T Cell Responses to Whole SARS Coronavirus in Humans,” The Journal of Immunology, vol. 181, no. 8, pp. 5490–5500, Oct. 2008, doi: 10.4049/jimmunol.181.8.5490.

H. Fehrenbach, “Alveolar epithelial type II cell: Defender of the alveolus revisited,” Respiratory Research, vol. 2, no. 1. pp. 33–46, Jan. 15, 2001. doi: 10.1186/rr36.

WE. Wei, Z. Li, CJ. Chiew, SE. Yong, MP. Toh, and VJ. Lee, “Presymptomatic Transmission of SARS-CoV-2 — Singapore, January 23–March 16, 2020,” MMWR. Morbidity and Mortality Weekly Report, vol. 69, no. 14, Apr. 2020, doi: 10.15585/mmwr.mm6914e1.

World Health Organization, “Situation Report-73 HIGHLIGHTS”, doi: 10.3201/eid2606.200239.

JB. Aguilar and JB. Gutierrez, “Investigating the Impact of Asymptomatic Carriers on COVID-19 Transmission,” medRxiv, p. 2020.03.18.20037994, Mar. 2020, doi: 10.1101/2020.03.18.20037994.

SG. Devaraj et al., “Regulation of IRF-3-dependent innate immunity by the papain-like protease domain of the severe acute respiratory syndrome coronavirus,” Journal of Biological Chemistry, vol. 282, no. 44, pp. 32208–32221, Nov. 2007, doi: 10.1074/jbc.M704870200.

M. Frieman, K. Ratia, RE. Johnston, AD. Mesecar, and RS. Baric, “Severe Acute Respiratory Syndrome Coronavirus Papain-Like Protease Ubiquitin-Like Domain and Catalytic Domain Regulate Antagonism of IRF3 and NF-κB Signaling,” Journal of Virology, vol. 83, no. 13, pp. 6689–6705, Jul. 2009, doi: 10.1128/jvi.02220-08.

IM. Verma, JK. Stevenson, EM. Schwarz, D. van Antwerp, and S. Miyamoto, “Rel/NF-κB/IκB family: Intimate tales of association and dissociation,” Genes and Development, vol. 9, no. 22. Cold Spring Harbor Laboratory Press, pp. 2723–2735, Nov. 15, 1995. doi: 10.1101/gad.9.22.2723.

D. Wang et al., “Clinical Characteristics of 138 Hospitalized Patients with 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China,” JAMA - Journal of the American Medical Association, Mar. 2020, doi: 10.1001/jama.2020.1585.

F. Chiappelli, “CoViD-19 Immunopathology & Immunotherapy,” Bioinformation, vol. 16, no. 3, pp. 219–222, Mar. 2020, doi: 10.6026/97320630016219.

C. Qin et al., “Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China,” Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, vol. 71, no. 15, pp. 762–768, Jul. 2020, doi: 10.1093/cid/ciaa248.

L. Roncati, V. Nasillo, B. Lusenti, and G. Riva, “Signals of Th2 immune response from COVID-19 patients requiring intensive care,” Annals of Hematology, vol. 99, no. 6, pp. 1419–1420, Jun. 2020, doi: 10.1007/s00277-020-04066-7.

E. Lozano, M. Dominguez-Villar, V. Kuchroo, and DA. Hafler, “The TIGIT/CD226 Axis Regulates Human T Cell Function,” The Journal of Immunology, vol. 188, no. 8, pp. 3869–3875, Apr. 2012, doi: 10.4049/jimmunol.1103627.

JL. Riley, “PD-1 signaling in primary T cells,” Immunological Reviews, vol. 229, no. 1, pp. 114–125, May 2009, doi: 10.1111/j.1600-065X.2009.00767.x.

M. Das, C. Zhu, and VK. Kuchroo, “Tim-3 and its role in regulating anti-tumor immunity,” Immunological Reviews, vol. 276, no. 1, pp. 97–111, Mar. 2017, doi: 10.1111/imr.12520.

N. Joller et al., “Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses,” Immunity, vol. 40, no. 4, pp. 569–581, Apr. 2014, doi: 10.1016/j.immuni.2014.02.012.

SD. Levin et al., “Vstm3 is a member of the CD28 family and an important modulator of T-cell function,” European Journal of Immunology, vol. 41, no. 4, pp. 902–915, Apr. 2011, doi: 10.1002/eji.201041136.

N. Joller et al., “Cutting Edge: TIGIT Has T Cell-Intrinsic Inhibitory Functions,” The Journal of Immunology, vol. 186, no. 3, pp. 1338–1342, Feb. 2011, doi: 10.4049/jimmunol.1003081.

KS. Boles et al., “A novel molecular interaction for the adhesion of follicular CD4 T cells to follicular DC,” European Journal of Immunology, vol. 39, no. 3, pp. 695–703, 2009, doi: 10.1002/eji.200839116.

X. Yu et al., “The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells,” Nature Immunology, vol. 10, no. 1, pp. 48–57, 2009, doi: 10.1038/ni.1674.

R. Tanner, B. Villarreal-Ramos, HM. Vordermeier, and H. McShane, “The humoral immune response to BCG vaccination,” Frontiers in Immunology, vol. 10, no. JUN. Frontiers Media SA., 2019. doi: 10.3389/fimmu.2019.01317.

A. Salem, A. Nofal, and D. Hosny, “Treatment of common and plane warts in children with topical viable bacillus calmette-guerin,” Pediatric Dermatology, vol. 30, no. 1, pp. 60–63, Jan. 2013, doi: 10.1111/j.1525-1470.2012.01848.x.

HM. Dockrell and SG. Smith, “What have we learnt about BCG vaccination in the last 20 years?,” Frontiers in Immunology, vol. 8, no. SEP. Frontiers Media SA., p. 1134, Sep. 13, 2017. doi: 10.3389/fimmu.2017.01134.

A. Roy et al., “Effect of BCG vaccination against Mycobacterium tuberculosis infection in children: systematic review and meta-analysis,” BMJ, vol. 349, Aug. 2014, doi: 10.1136/BMJ.G4643.

LK. Schrager, RC. Harris, and J. Vekemans, “Research and development of new tuberculosis vaccines: a review,” F1000Research, vol. 7, 2018, doi: 10.12688/F1000RESEARCH.16521.2.

JM. Achkar, J. Chan, and A. Casadevall, “B cells and antibodies in the defense against Mycobacterium tuberculosis infection,” Immunological Reviews, vol. 264, no. 1, pp. 167–181, Mar. 2015, doi: 10.1111/imr.12276.

C. Zufferey, S. Germano, B. Dutta, N. Ritz, and N. Curtis, “The Contribution of Non-Conventional T Cells and NK Cells in the Mycobacterial-Specific IFNγ Response in Bacille Calmette-Guérin (BCG)-Immunized Infants,” PLoS ONE, vol. 8, no. 10, pp. 819–835, Oct. 2013, doi: 10.1371/journal.pone.0077334.

RM. Brown et al., “Lipoarabinomannan-Reactive Human Secretory Immunoglobulin A Responses Induced by Mucosal Bacille Calmette-Guérin Vaccination,” The Journal of Infectious Diseases, vol. 187, no. 3, pp. 513–517, Feb. 2003, doi: 10.1086/368096.

R. Monteiro-Maia, MB. Ortigão-de-Sampaio, RT. Pinho, and LRR. Castello-Branco, “Modulation of humoral immune response to oral BCG vaccination by Mycobacterium bovis BCG Moreau Rio de Janeiro (RDJ) in healthy adults,” Journal of Immune Based Therapies and Vaccines, vol. 4, no. 1, pp. 1–6, Sep. 2006, doi: 10.1186/1476-8518-4-4/FIGURES/4.

I. v. Lyadova, HM. Vordermeier, EB. Eruslanov, S. v. Khaidukov, AS. Apt, and RG. Hewinson, “Intranasal BCG vaccination protects BALB/c mice against virulent Mycobacterium bovis and accelerates production of IFN-gamma in their lungs,” Clinical and Experimental Immunology, vol. 126, no. 2, pp. 274–279, Nov. 2001, doi: 10.1046/j.1365-2249.2001.01667.x.

G. Falero-Diaz, S. Challacombe, D. Banerjee, G. Douce, A. Boyd, and J. Ivanyi, “Intranasal vaccination of mice against infection with Mycobacterium tuberculosis.,” Vaccine, vol. 18, no. 28, pp. 3223–9, Aug. 2000, doi: 10.1016/s0264-410x(00)00134-1.

SR. Rosenthal, JT. Mcenery, and N. Raisys, “Aerogenic BCG vaccination against tuberculosis in animal and human subjects,” Journal of Asthma, vol. 5, no. 4, pp. 309–323, 1968, doi: 10.3109/02770906809100348.

PA. Darrah et al., “Prevention of tuberculosis in macaques after intravenous BCG immunization,” Nature, vol. 577, no. 7788, pp. 95–102, Jan. 2020, doi: 10.1038/s41586-019-1817-8.

S. Mehra et al., “Transcriptional reprogramming in nonhuman primate (Rhesus Macaque) tuberculosis granulomas,” PLoS ONE, vol. 5, no. 8, p. e12266, Aug. 2010, doi: 10.1371/journal.pone.0012266.

J. Liu, V. Tran, AS. Leung, DC. Alexander, and B. Zhu, “BCG vaccines: Their mechanisms of attenuation and impact on safety and protective efficacy,” Human Vaccines, vol. 5, no. 2. Taylor & Francis, pp. 70–78, 2009. doi: 10.4161/hv.5.2.7210.

S. Luca and T. Mihaescu, “History of BCG Vaccine,” Iasi, 2013.

HM. Dockrell and SG. Smith, “What have we learnt about BCG vaccination in the last 20 years?,” Frontiers in Immunology, vol. 8, no. SEP. Frontiers Media SA., p. 1134, Sep. 13, 2017. doi: 10.3389/fimmu.2017.01134.

PEM. Fine et al., “Environmental mycobacteria in nothern Malawi: Implications for the epidemiology of tuberculosis and leprosy,” Epidemiology and Infection, vol. 126, no. 3, pp. 379–387, 2001, doi: 10.1017/S0950268801005532.

RE. Weir et al., “The influence of previous exposure to environmental mycobacteria on the interferon-gamma response to bacille Calmette?Gurin vaccination in southern England and northern Malawi,” Clinical and Experimental Immunology, vol. 146, no. 3, pp. 390–399, Dec. 2006, doi: 10.1111/j.1365-2249.2006.03222.x.

L. Brandt et al., “Failure of the Mycobacterium bovis BCG vaccine: Some species of environmental mycobacteria block multiplication of BCG and induction of protective immunity to tuberculosis,” Infection and Immunity, vol. 70, no. 2, pp. 672–678, Feb. 2002, doi: 10.1128/IAI.70.2.672-678.2002.

VL. Petricevich et al., “A single strain of Mycobacterium bovis bacillus Calmette-Guérin (BCG) grown in two different media evokes distinct humoral immune responses in mice,” Brazilian Journal of Medical and Biological Research, vol. 34, no. 1, pp. 81–92, 2001, doi: 10.1590/S0100-879X2001000100010.

RJW. Arts et al., “Long-term in vitro and in vivo effects of γ-irradiated BCG on innate and adaptive immunity,” Journal of Leukocyte Biology, vol. 98, no. 6, pp. 995–1001, Dec. 2015, doi: 10.1189/jlb.4ma0215-059r.

J. Kleinnijenhuis et al., “Bacille Calmette-Guérin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 43, pp. 17537–17542, Oct. 2012, doi: 10.1073/pnas.1202870109.

AM. Minassian, I. Satti, ID. Poulton, J. Meyer, AVS. Hill, and H. McShane, “A human challenge model for Mycobacterium tuberculosis using Mycobacterium bovis bacille Calmette-Guérin,” Journal of Infectious Diseases, vol. 205, no. 7, pp. 1035–1042, Apr. 2012, doi: 10.1093/infdis/jis012.

R. Tanner, B. Villarreal-Ramos, HM. Vordermeier, and H. McShane, “The humoral immune response to BCG vaccination,” Frontiers in Immunology, vol. 10, no. JUN, p. 1317, 2019, doi: 10.3389/fimmu.2019.01317.

P. Ravn, H. Boesen, BK. Pedersen, and P. Andersen, “Human T cell responses induced by vaccination with Mycobacterium bovis bacillus Calmette-Guérin.,” The Journal of Immunology, vol. 158, no. 4, 1997.

P. Andersen and SHE. Kaufmann, “Novel vaccination strategies against tuberculosis,” Cold Spring Harbor perspectives in medicine, vol. 4, no. 6, 2014, doi: 10.1101/CSHPERSPECT.A018523.

SHE. Kaufmann, “Tuberculosis vaccines: time to think about the next generation,” Seminars in immunology, vol. 25, no. 2, pp. 172–181, Apr. 2013, doi: 10.1016/J.SMIM.2013.04.006.

I. Sebina et al., “Long-lived memory B-cell responses following BCG vaccination,” PloS one, vol. 7, no. 12, Dec. 2012, doi: 10.1371/JOURNAL.PONE.0051381.

C. Covián et al., “BCG-Induced Cross-Protection and Development of Trained Immunity: Implication for Vaccine Design,” Frontiers in Immunology, vol. 10, p. 2806, Nov. 2019, doi: 10.3389/fimmu.2019.02806.

RA. Murray et al., “Bacillus Calmette Guerin vaccination of human newborns induces a specific, functional CD8+ T cell response,” Journal of immunology (Baltimore, Md. : 1950), vol. 177, no. 8, pp. 5647–5651, Oct. 2006, doi: 10.4049/JIMMUNOL.177.8.5647.

WA. Hanekom, “The immune response to BCG vaccination of newborns,” Annals of the New York Academy of Sciences, vol. 1062, pp. 69–78, 2005, doi: 10.1196/ANNALS.1358.010.

AP. Soares et al., “Bacillus Calmette-Guérin vaccination of human newborns induces T cells with complex cytokine and phenotypic profiles,” Journal of immunology (Baltimore, Md. : 1950), vol. 180, no. 5, pp. 3569–3577, Mar. 2008, doi: 10.4049/JIMMUNOL.180.5.3569.

AP. Soares et al., “Longitudinal changes in CD4(+) T-cell memory responses induced by BCG vaccination of newborns,” The Journal of infectious diseases, vol. 207, no. 7, pp. 1084–1094, Apr. 2013, doi: 10.1093/INFDIS/JIS941.

H. Su, B. Peng, Z. Zhang, Z. Liu, and Z. Zhang, “The Mycobacterium tuberculosis glycoprotein Rv1016c protein inhibits dendritic cell maturation, and impairs Th1 /Th17 responses during mycobacteria infection,” Molecular immunology, vol. 109, pp. 58–70, May 2019, doi: 10.1016/J.MOLIMM.2019.02.021.

RM. Steinman and H. Hemmi, “Dendritic cells: Translating innate to adaptive immunity,” Current Topics in Microbiology and Immunology, vol. 311, pp. 17–58, 2006, doi: 10.1007/3-540-32636-7_2.

CR. Hole et al., “Induction of memory-like dendritic cell responses in vivo,” Nature Communications, vol. 10, no. 1, Dec. 2019, doi: 10.1038/s41467-019-10486-5.

KM. Henkels, K. Frondorf, ME. Gonzalez-Mejia, AL. Doseff, and J. Gomez-Cambronero, “IL-8-induced neutrophil chemotaxis is mediated by Janus kinase 3 (JAK3),” FEBS Letters, vol. 585, no. 1, pp. 159–166, Jan. 2011, doi: 10.1016/j.febslet.2010.11.031.

Y. Zhang et al., “Enhanced interleukin-8 release and gene expression in macrophages after exposure to Mycobacterium tuberculosis and its components,” Journal of Clinical Investigation, vol. 95, no. 2, pp. 586–592, 1995, doi: 10.1172/JCI117702.

T. Chen et al., “Association of Human Antibodies to Arabinomannan With Enhanced Mycobacterial Opsonophagocytosis and Intracellular Growth Reduction,” The Journal of infectious diseases, vol. 214, no. 2, pp. 300–310, Jul. 2016, doi: 10.1093/INFDIS/JIW141.

A. Yáñez et al., “Detection of a TLR2 agonist by hematopoietic stem and progenitor cells impacts the function of the macrophages they produce,” European Journal of Immunology, vol. 43, no. 8, pp. 2114–2125, Aug. 2013, doi: 10.1002/eji.201343403.

JW. WOUT, R. POELL, and R. FURTH, “The Role of BCG/PPD-Activated Macrophages in Resistance against Systemic Candidiasis in Mice,” Scandinavian Journal of Immunology, vol. 36, no. 5, pp. 713–720, Nov. 1992, doi: 10.1111/j.1365-3083.1992.tb03132.x.

F. Chen et al., “Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion,” Nature Immunology, vol. 15, no. 10, pp. 938–946, Jan. 2014, doi: 10.1038/ni.2984.

E. Kaufmann et al., “BCG Educates Hematopoietic Stem Cells to Generate Protective Innate Immunity against Tuberculosis,” Cell, vol. 172, no. 1–2, pp. 176-190.e19, Jan. 2018, doi: 10.1016/j.cell.2017.12.031.

A. Dolganiuc, C. Garcia, K. Kodys, and G. Szabo, “Distinct toll-like receptor expression in monocytes and T cells in chronic HCV infection,” World Journal of Gastroenterology, vol. 12, no. 8, pp. 1198–1204, Feb. 2006, doi: 10.3748/wjg.v12.i8.1198.

GF. Black et al., “BCG-induced increase in interferon-gamma response to mycobacterial antigens and efficacy of BCG vaccination in Malawi and the UK: Two randomised controlled studies,” Lancet, vol. 359, no. 9315, pp. 1393–1401, Apr. 2002, doi: 10.1016/S0140-6736(02)08353-8.

E. Kindler, V. Thiel, and F. Weber, “Interaction of SARS and MERS Coronaviruses with the Antiviral Interferon Response,” in Advances in Virus Research, vol. 96, Academic Press Inc., 2016, pp. 219–243. doi: 10.1016/bs.aivir.2016.08.006.

E. Hamano et al., “Polymorphisms of interferon-inducible genes OAS-1 and MxA associated with SARS in the Vietnamese population,” Biochemical and Biophysical Research Communications, vol. 329, no. 4, pp. 1234–1239, Apr. 2005, doi: 10.1016/j.bbrc.2005.02.101.

A. Kapoor, YH. Fan, and R. Arav-Boger, “Bacterial Muramyl Dipeptide (MDP) Restricts Human Cytomegalovirus Replication via an IFN-β-Dependent Pathway,” Scientific Reports, vol. 6, no. 1, pp. 1–15, Feb. 2016, doi: 10.1038/srep20295.

T. Fekete, G. Koncz, B. Szabo, A. Gregus, and E. Rajnavölgyi, “Interferon gamma boosts the nucleotide oligomerization domain 2-mediated signaling pathway in human dendritic cells in an X-linked inhibitor of apoptosis protein and mammalian target of rapamycin-dependent manner,” Cellular and Molecular Immunology, vol. 14, no. 4, pp. 380–391, Apr. 2017, doi: 10.1038/cmi.2015.90.

SE. Girardin et al., “Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection,” J Biol Chem, vol. 278, doi: 10.1074/jbc.c200651200.

A. Kapoor, M. Forman, and R. Arav-Boger, “Activation of Nucleotide Oligomerization Domain 2 (NOD2) by Human Cytomegalovirus Initiates Innate Immune Responses and Restricts Virus Replication,” PLoS ONE, vol. 9, no. 3, p. e92704, Mar. 2014, doi: 10.1371/journal.pone.0092704.

A. Sabbah et al., “Activation of innate immune antiviral responses by Nod2,” Nature Immunology, vol. 10, no. 10, pp. 1073–1080, 2009, doi: 10.1038/ni.1782.

T. Higashimoto, N. Chan, YK. Lee, and E. Zandi, “Regulation of IκB kinase complex by phosphorylation of γ-binding domain of IκB kinase β by polo-like kinase 1,” Journal of Biological Chemistry, vol. 283, no. 51, pp. 35354–35367, Dec. 2008, doi: 10.1074/jbc.M806258200.

S. Sharma and PG. Thomas, “The two faces of heterologous immunity: protection or immunopathology,” Journal of Leukocyte Biology, vol. 95, no. 3, pp. 405–416, Mar. 2014, doi: 10.1189/jlb.0713386.

TP. Primm, CA. Lucero, and JO. Falkinham, “Health Impacts of Environmental Mycobacteria,” Clinical Microbiology Reviews, vol. 17, no. 1, pp. 98–106, Jan. 2004, doi: 10.1128/CMR.17.1.98-106.2004.

U. Syrbe, S. Jennrich, A. Schottelius, A. Richter, A. Radbruch, and A. Hamann, “Differential regulation of P-selectin ligand expression in naive versus memory CD4+ T cells: Evidence for epigenetic regulation of involved glycosyltransferase genes,” Blood, vol. 104, no. 10, pp. 3243–3248, Nov. 2004, doi: 10.1182/blood-2003-09-3047.

G. Ristori, D. Faustman, G. Matarese, S. Romano, and M. Salvetti, “Bridging the gap between vaccination with Bacille Calmette-Guérin (BCG) and immunological tolerance: the cases of type 1 diabetes and multiple sclerosis,” Current Opinion in Immunology, vol. 55, pp. 89–96, Dec. 2018, doi: 10.1016/j.coi.2018.09.016.

RJW. Arts et al., “BCG Vaccination Protects against Experimental Viral Infection in Humans through the Induction of Cytokines Associated with Trained Immunity,” Cell Host and Microbe, vol. 23, no. 1, pp. 89-100.e5, Jan. 2018, doi: 10.1016/j.chom.2017.12.010.

HS. Goodridge et al., “Harnessing the beneficial heterologous effects of vaccination,” Nature Reviews Immunology, vol. 16, no. 6, pp. 392–400, Jun. 2016, doi: 10.1038/nri.2016.43.

KS. Mathurin, GW. Martens, H. Kornfeld, and RM. Welsh, “CD4 T-Cell-Mediated Heterologous Immunity between Mycobacteria and Poxviruses,” Journal of Virology, vol. 83, no. 8, pp. 3528–3539, Apr. 2009, doi: 10.1128/jvi.02393-08.

AB. Kulkarni, HC. Morse, JR. Bennink, JW. Yewdell, and BR. Murphy, “Immunization of mice with vaccinia virus-M2 recombinant induces epitope-specific and cross-reactive Kd-restricted CD8+ cytotoxic T cells.,” Journal of Virology, vol. 67, no. 7, pp. 4086–4092, 1993, doi: 10.1128/jvi.67.7.4086-4092.1993.

K. Koichi, VE. Reyes, RE. Humphreys, and FA. Ennis, “Recognition of disparate HA and NS1 peptides by an H-2Kd-restricted, influenza specific CTL clone,” Molecular Immunology, vol. 28, no. 1–2, pp. 1–7, Jan. 1991, doi: 10.1016/0161-5890(91)90080-4.

N. Shimojo, WL. Maloy, RW. Anderson, WE. Biddison, and JE. Coligan, “Specificity of peptide binding by the HLA-A2.1 molecule.,” The Journal of Immunology, vol. 143, no. 9, 1989.

RW. Anderson, JR. Bennink, JW. Yewdell, WL. Maloy, and JE. Coligan, “Influenza basic polymerase 2 peptides are recognized by influenza nucleoprotein-specific cytotoxic T lymphocytes,” Molecular Immunology, vol. 29, no. 9, pp. 1089–1096, 1992, doi: 10.1016/0161-5890(92)90041-U.

LK. Selin, SR. Nahill, and RM. Welsh, “Cross-reactivities in memory cytotoxic t lymphocyte recognition of heterologous viruses,” Journal of Experimental Medicine, vol. 179, no. 6, pp. 1933–1943, Jun. 1994, doi: 10.1084/jem.179.6.1933.

RM. Welsh and LK. Selin, “No one is naive: The significance of heterologous T-cell immunity,” Nature Reviews Immunology, vol. 2, no. 6, pp. 417–426, 2002, doi: 10.1038/nri820.

AH. Ellebedy and R. Ahmed, “Re-Engaging Cross-Reactive Memory B Cells: The Influenza Puzzle,” Frontiers in Immunology, vol. 3, no. MAR, p. 53, Mar. 2012, doi: 10.3389/fimmu.2012.00053.

R. Levin-Klein and Y. Bergman, “Epigenetic regulation of monoallelic rearrangement (allelic exclusion) of antigen receptor genes,” Frontiers in Immunology, vol. 5, no. DEC, 2014, doi: 10.3389/fimmu.2014.00625.

M. Yamashita et al., “Bmi1 regulates memory CD4 T cell survival via repression of the Noxa gene,” Journal of Experimental Medicine, vol. 205, no. 5, pp. 1109–1120, May 2008, doi: 10.1084/jem.20072000.

JK. Northrop, RM. Thomas, AD. Wells, and H. Shen, “ Epigenetic Remodeling of the IL-2 and IFN -γ Loci in Memory CD8 T Cells Is Influenced by CD4 T Cells ,” The Journal of Immunology, vol. 177, no. 2, pp. 1062–1069, Jul. 2006, doi: 10.4049/jimmunol.177.2.1062.

EN. Kersh et al., “ Rapid Demethylation of the IFN -γ Gene Occurs in Memory but Not Naive CD8 T Cells ,” The Journal of Immunology, vol. 176, no. 7, pp. 4083–4093, Apr. 2006, doi: 10.4049/jimmunol.176.7.4083.

T. Naito and I. Taniuchi, “Roles of repressive epigenetic machinery in lineage decision of T cells,” Immunology, vol. 139, no. 2, pp. 151–157, Jun. 2013, doi: 10.1111/imm.12058.

PS. de Araujo-Souza, SCH. Hanschke, and JPB. Viola, “Epigenetic control of interferon-gamma expression in CD8 T cells,” Journal of Immunology Research, vol. 2015. Hindawi Publishing Corporation, 2015. doi: 10.1155/2015/849573.

S. Steinfelder et al., “Epigenetic modification of the human CCR6 gene is associated with stable CCR6 expression in T cells,” Blood, vol. 117, no. 10, pp. 2839–2846, Mar. 2011, doi: 10.1182/blood-2010-06-293027.

C. Schmidl, L. Hansmann, R. Andreesen, M. Edinger, P. Hoffmann, and M. Rehli, “Epigenetic reprogramming of the RORC locus during in vitro expansion is a distinctive feature of human memory but not naïve Treg,” European Journal of Immunology, vol. 41, no. 5, pp. 1491–1498, May 2011, doi: 10.1002/eji.201041067.

TA. Fehniger and MA. Caligiuri, “Interleukin 15: Biology and relevance to human disease,” Blood, vol. 97, no. 1, pp. 14–32, Jan. 2001, doi: 10.1182/blood.V97.1.14.

B. Agrawal, “Heterologous Immunity: Role in Natural and Vaccine-Induced Resistance to Infections,” Frontiers in Immunology, vol. 10, p. 2631, Nov. 2019, doi: 10.3389/fimmu.2019.02631.

M. Lipsitch, YH. Grad, A. Sette, and S. Crotty, “Cross-reactive memory T cells and herd immunity to SARS-CoV-2,” Nature Reviews Immunology, vol. 20, no. 11, pp. 709–713, Nov. 2020, doi: 10.1038/s41577-020-00460-4.

HS. Goodridge et al., “Harnessing the beneficial heterologous effects of vaccination,” Nature Reviews Immunology, vol. 16, no. 6. Nature Publishing Group, pp. 392–400, Jun. 01, 2016. doi: 10.1038/nri.2016.43.

VC. Senterfitt and JW. Shands, “Salmonellosis in Mice Infected with Mycobacterium bovis BCG II. Resistance to Infection,” Infection and Immunity, vol. 1, no. 6, pp. 583–586, Jun. 1970.

MOC. Ota et al., “ Influence of Mycobacterium bovis Bacillus Calmette-Guérin on Antibody and Cytokine Responses to Human Neonatal Vaccination ,” The Journal of Immunology, vol. 168, no. 2, pp. 919–925, Jan. 2002, doi: 10.4049/jimmunol.168.2.919.

N. Ritz, M. Mui, A. Balloch, and N. Curtis, “Non-specific effect of Bacille Calmette-Guérin vaccine on the immune response to routine immunisations,” Vaccine, vol. 31, no. 30, pp. 3098–3103, Jun. 2013, doi: 10.1016/j.vaccine.2013.03.059.

A. Kiravu et al., “Bacille Calmette-Guérin Vaccine Strain Modulates the Ontogeny of Both Mycobacterial-Specific and Heterologous T Cell Immunity to Vaccination in Infants,” Frontiers in Immunology, vol. 10, p. 2307, Oct. 2019, doi: 10.3389/FIMMU.2019.02307/BIBTEX.

LG. Stensballe et al., “BCG Vaccination at Birth and Rate of Hospitalization for Infection Until 15 Months of Age in Danish Children: A Randomized Clinical Multicenter Trial,” Journal of the Pediatric Infectious Diseases Society, vol. 8, no. 3, pp. 213–220, Jul. 2019, doi: 10.1093/JPIDS/PIY029.

E. Datau, A. Sultana, V. Mandang, and E. Jim, “The Efficacy of Bacillus Calmette-Guérin Vaccinations for The Prevention of Acute Upper Respiratory Tract Infection in The Elderly,” 2010.

LG. Stensballe et al., “Acute lower respiratory tract infections and respiratory syncytial virus in infants in Guinea-Bissau: A beneficial effect of BCG vaccination for girls: Community based case-control study,” Vaccine, vol. 23, no. 10, pp. 1251–1257, Jan. 2005, doi: 10.1016/j.vaccine.2004.09.006.

E. Nemes et al., “Prevention of M. tuberculosis Infection with H4:IC31 Vaccine or BCG Revaccination,” New England Journal of Medicine, vol. 379, no. 2, pp. 138–149, Jul. 2018, doi: 10.1056/NEJMoa1714021.

D. Daulatabad, D. Pandhi, and A. Singal, “BCG vaccine for immunotherapy in warts: is it really safe in a tuberculosis endemic area?,” Dermatologic Therapy, vol. 29, no. 3, pp. 168–172, May 2016, doi: 10.1111/dth.12336.

I. Podder et al., “Immunotherapy in viral warts with intradermal Bacillus Calmette–Guerin vaccine versus intradermal tuberculin purified protein derivative: A double-blind, randomized controlled trial comparing effectiveness and safety in a tertiary care center in Eastern India,” Indian Journal of Dermatology, Venereology, and Leprology, vol. 83, no. 3, p. 411, May 2017, doi: 10.4103/0378-6323.193623.

A. Salem, A. Nofal, and D. Hosny, “Treatment of Common and Plane Warts in Children with Topical Viable Bacillus Calmette-Guerin,” Pediatric Dermatology, vol. 30, no. 1, pp. 60–63, Jan. 2013, doi: 10.1111/j.1525-1470.2012.01848.x.

L. Jenneke et al., “BCG Vaccination Enhances the Immunogenicity of Subsequent Influenza Vaccination in Healthy Volunteers: A Randomized, Placebo-Controlled Pilot Study,” The Journal of Infectious Diseases, vol. 2012, no. 12, pp. 1930–1938, Dec. 2015, doi: https://doi.org/10.1093/infdis/jiv332.

VAK. Rathinam and KA. Fitzgerald, “Inflammasomes and anti-viral immunity,” Journal of Clinical Immunology, vol. 30, no. 5. Springer, pp. 632–637, Sep. 28, 2010. doi: 10.1007/s10875-010-9431-4.

H. Yang and WA. Tompkins, “Nonspecific cytotoxicity of vaccinia-induced peritoneal exudates in hamsters is mediated by Thy-1.2 homologue-positive cells distinct from NK cells and macrophages.,” Journal of immunology (Baltimore, Md. : 1950), vol. 131, no. 5, pp. 2545–50, Nov. 1983, Accessed: Apr. 06, 2020. [Online]. Available: http://www.ncbi.nlm.nih.gov/pubmed/6195269

PG. Thomas et al., “The Intracellular Sensor NLRP3 Mediates Key Innate and Healing Responses to Influenza A Virus via the Regulation of Caspase-1,” Immunity, vol. 30, no. 4, pp. 566–575, Apr. 2009, doi: 10.1016/j.immuni.2009.02.006.

“IL1B interleukin 1 beta [Homo sapiens (human)] - Gene - NCBI.” https://www.ncbi.nlm.nih.gov/gene?Db=gene&Cmd=ShowDetailView&TermToSearch=3553 (accessed Apr. 06, 2020).

J. Kleinnijenhuis et al., “Long-Lasting Effects of BCG Vaccination on Both Heterologous Th1/Th17 Responses and Innate Trained Immunity,” Journal of Innate Immunity, vol. 6, no. 2, pp. 152–158, 2014, doi: 10.1159/000355628.

N. Curtis, A. Sparrow, T. A. Ghebreyesus, and M. G. Netea, “Considering BCG vaccination to reduce the impact of COVID-19,” The Lancet, vol. 0, no. 0, 2020, doi: 10.1016/S0140-6736(20)31025-4.

J. Liu, V. Tran, A. S. Leung, D. C. Alexander, and B. Zhu, “BCG vaccines: their mechanisms of attenuation and impact on safety and protective efficacy,” Human vaccines, vol. 5, no. 2, pp. 70–78, 2009, doi: 10.4161/HV.5.2.7210.

J. M. Chen, S. T. Islam, H. Ren, and J. Liu, “Differential productions of lipid virulence factors among BCG vaccine strains and implications on BCG safety,” Vaccine, vol. 25, no. 48, pp. 8114–8122, Nov. 2007, doi: 10.1016/J.VACCINE.2007.09.041.

Downloads

Published

2021-12-29

How to Cite

1.
Kovačić D, Gajić AA, Latinović D, Softić A. Hypothetical Immunological and Immunogenetic Model of Heterogenous Effects of BCG Vaccination in SARS-CoV-2 Infections: BCG-induced Trained and Heterologous Immunity. JMS [Internet]. 2021 Dec. 29 [cited 2024 Apr. 25];90(4):e551. Available from: https://jms.ump.edu.pl/index.php/JMS/article/view/551

Issue

Section

Review Papers
Received 2021-08-12
Accepted 2021-11-28
Published 2021-12-29