Skip to main content

EDITORIAL article

Front. Immunol., 19 January 2024
Sec. Molecular Innate Immunity
This article is part of the Research Topic The Role of Toll-like Receptors and their Related Signaling Pathways in Viral Infection and Inflammation View all 14 articles

Editorial: The role of toll-like receptors and their related signaling pathways in viral infection and inflammation

  • 1Institute for Immunology, Eberhard Karls University Tuebingen, Tuebingen, Germany
  • 2Department of R&D, Syntacoll GmbH, Saal an der Donau, Germany

The Research Topic addresses the role of Toll-like receptors (TLRs) and their associated signaling pathways in viral infections and inflammation. The focus is on how these receptors and pathways contribute to protecting the body against viruses and their involvement in associated inflammatory processes. The present Research Topic encompasses 13 studies shedding light on the role of TLRs in viral infections and inflammation from various perspectives.

The impact of TLRs on viral infections has been elucidated in three studies, effectively illustrating the dual nature of TLR activation in viral infection – both necessary for immune protection and contributing to virus-triggered immunopathology (13). Miles et al. demonstrated that TLR7 supports chronic respiratory disease after RSV infection in mice. This study unveils a previously unknown molecular mechanism of lower respiratory tract pathogenesis by RSV, emphasizing the potential of TLR7 modulation to confine RSV pathology to the upper respiratory tract. This restriction prevents its spread to the lower respiratory tract and results in a significant reduction in clinical symptoms.

Theiler’s Virus, known for its ability to infect the central nervous system of mice and trigger an inflammatory reaction, is commonly utilized as a model in research to investigate the mechanisms of virus infections and the course of inflammatory reactions in the brain (46). In this study, Kim reviewed the role of TLRs in the mouse model of Theiler’s Virus infection, concluding that the state of TLR activation in the host plays a crucial role in the initial virus replication and persistence.

The use of estrogens in feminizing hormone therapy (FHT) can elevate inflammatory reactions and the risk of cardiovascular mortality in transgender women (TW), who are at an increased risk for both human immunodeficiency virus (HIV) and cardiovascular diseases (CVD) (79). Kettelhut et al. demonstrate that in vitro data suggest estrogen exposure can enhance the activation of the innate immune system in FHT. The authors discuss the complex interactions of FHT, HIV, and CVD in TW, emphasizing the need for further research to fully understand these interactions and determine optimal FHT regimens or complementary treatments aiming to reduce excessive immune activation.

Lipopolysaccharide (LPS), a TLR4 agonist, is used as a stimulus in many in vitro and in vivo models (10, 11). O’Neill et al. investigated the impact of dexamethasone on the expression of antimicrobial mediators in LPS-activated primary macrophages, demonstrating the inhibition of both the expression and function of interferon-β. Interferon-β plays a crucial role in the early antiviral immune response.

In a study with healthy volunteers using an imiquimod (IMQ) model, Assil et al. showed that orally administered prednisolone suppresses skin inflammation induced by IMQ, a topical agent that triggers local inflammation through TLR7.

Resiquimod, an imidazoquinoline compound with antiviral and antitumor activity, functions as a TLR7/TLR8 agonist (12, 13). In their work, Keppler et al. point out that clinical use is limited to topical application. Systemic applications of TLR ligands like Resiquimod have failed due to side effects restricting dose and efficacy. Therefore, the authors developed TLR7/8-agonistic imidazoquinolines designed to distribute via endosomes using a macrolide carrier. The substances were designed to distribute to cellular compartments where the target receptor and a specific combination of signaling molecules relevant for IFNα release are present.

The immune reaction to biomaterial implants, known as the foreign body reaction, poses a significant challenge in biomedical engineering, as it can lead to chronic inflammatory reactions to the implanted material (14). McKiel et al. demonstrate that damage-associated molecular patterns (DAMPs) and other intracellular proteins easily adsorb to biomaterial surfaces in competition with plasma proteins. Adsorbed DAMPs in adherent macrophages trigger an inflammatory reaction mediated by the MyD88-dependent TLR2 signaling pathway.

The TLR-2 signaling pathway is also involved in the inflammatory reaction triggered by sperm in the uterus during fertilization, but the precise molecular mechanism remains unknown. According to ligand specificity, TLR2 forms a heterodimerization with TLR1 or TLR6, respectively, as the initial step to mediate intracellular signaling and induce a specific type of immune response. Mansouri et al. aimed to identify the active TLR2 heterodimer (TLR2/1 or TLR2/6) involved in the immunological interaction between sperm and the uterus in cattle. Overall, the results show that sperm utilize the heterodimerization of TLR2/1, but not TLR2/6, to trigger a weak physiological inflammatory reaction in the bovine uterus. This could be the mechanism to remove excess dead sperm remaining in the uterus without tissue damage and create an ideal uterine environment for the uptake and implantation of the early embryo.

In the present Research Topic, there are three publications with a direct clinical relevance. In a case report by Hirsiger et al., the occurrence of agranulocytosis within a few days after the first dose of an mRNA-1273 vaccine against COVID-19 in a previously healthy older adult is described. The patient was diagnosed with large granular lymphocytic T-cell leukemia (T-LGL) of STAT3 wild type. The mRNA-1273 vaccine activated TLR-3, leading to TLR-mediated IL-6/STAT3 pathway activation. The authors changed the vaccination strategy and used a vector vaccine, and neutropenia did not recur. Tian et al. investigated the association between TLRs and the activation and differentiation of T-cells in Takayasu arteritis (TAK). They analyzed the mRNA frequency of 29 target genes in peripheral blood mononuclear cells (PBMCs) from 27 TAK patients and 10 healthy controls. They found increased mRNA levels of TLR2 and TLR4 in TAK patients. Furthermore, this study revealed a novel connection between TLRs and T-cells in the pathogenesis of autoimmune diseases. Another study by Uebelhoer et al. examines how malnutrition and acute illnesses affect immune responses in young children in low- and middle-income countries and which aspects of immunity are relevant in this particularly vulnerable population. Overall, the results show exaggerated innate immune responses to pathogen-associated molecules, especially TLR-4 and TLR-7/8, in acutely ill young children, persisting during recovery. Exaggerated innate immune responses to TLR ligands can contribute to chronic systemic inflammation and dysregulated responses to subsequent infection challenges.

Their ability to stimulate immunity makes TLR attractive targets for expanding numerous immunotherapeutic approaches to combat cancer (15). These immunotherapeutic strategies include the use of TLR ligands/agonists as monotherapy or in combined therapeutic approaches (16). In a review, Chakraborty et al. attempt to provide a comprehensive discussion of significant TLR agonists and their application, as well as the challenges in integrating them into cancer immunotherapy approaches, with a particular emphasis on the use of TLR agonists as functional adjuvants for cancer vaccines. They present the translational potential of an autologous cancer vaccine, as well as the immune-inducing potential of TLR agonists as a potential immunotherapy in various types of cancer.

In another study, Jin et al. show that the unique immune system of the Atlantic cod offers an unprecedented opportunity to explore the evolutionary history of pattern recognition receptor (PRR)-based signaling in the immune defense of vertebrates. The absence of major histocompatibility complex class II antigen presentation and several pathogen recognition receptors in the Atlantic cod has not impaired the immune response.

Overall, this Research Topic effectively illustrates the dual role of Toll-like receptors (TLRs) and their associated signaling pathways in viral infections, as well as in a broad variety of inflammatory processes. It highlights the two facets of TLR activation: one contributing to immune protection and the other to immunopathology.

Author contributions

OP: Writing – original draft, Writing – review & editing. RK: Writing – original draft, Writing – review & editing.

Conflict of interest

Author RK was employed by the company Syntacoll GmbH.

The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Thompson JM, Iwasaki A. Toll-like receptors regulation of viral infection and disease. Adv Drug Delivery Rev (2008) 60:786–94. doi: 10.1016/j.addr.2007.11.003

CrossRef Full Text | Google Scholar

2. Rouse BT, Sehrawat S. Immunity and immunopathology to viruses: what decides the outcome? Nat Rev Immunol (2010) 10:514–26. doi: 10.1038/nri2802

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-like receptor signaling and its role in cell-mediated immunity. Front Immunol (2022) 13:812774. doi: 10.3389/fimmu.2022.812774

PubMed Abstract | CrossRef Full Text | Google Scholar

4. DePaula-Silva AB, Hanak TJ, Libbey JE, Fujinami RS. Theiler’s murine encephalomyelitis virus infection of SJL/J and C57BL/6J mice: Models for multiple sclerosis and epilepsy. J Neuroimmunol (2017) 308:30–42. doi: 10.1016/j.jneuroim.2017.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

5. So EY, Kim BS. Theiler’s virus infection induces TLR3-dependent upregulation of TLR2 critical for proinflammatory cytokine production. Glia (2009) 57:1216–26. doi: 10.1002/glia.20843

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Gern OL, Mulenge F, Pavlou A, Ghita L, Steffen I, Stangel M, et al. Toll-like receptors in viral encephalitis. Viruses 13 (2021) 13:2065–98. doi: 10.3390/v13102065

CrossRef Full Text | Google Scholar

7. Nguyen HB, Chavez AM, Lipner E, Hantsoo L, Kornfield SL, Davies RD, et al. Gender-affirming hormone use in transgender individuals: impact on behavioral health and cognition. Curr Psychiatry Rep (2018) 20:110. doi: 10.1007/s11920-018-0973-0

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Baral SD, Poteat T, Stromdahl S, Wirtz AL, Guadamuz TE, Beyrer C. Worldwide burden of HIV in transgender women: a systematic review and meta-analysis. Lancet Infect Dis (2013) 13:214–22. doi: 10.1016/S1473-3099(12)70315-8

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Streed CG Jr., Beach LB, Caceres BA, Dowshen NL, Moreau KL, Mukherjee M, et al. Assessing and addressing cardiovascular health in people who are transgender and gender diverse: A scientific statement from the american heart association. Circulation (2021) 144:e136–48. doi: 10.1161/CIR.0000000000001003

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Chow JC, Young DW, Golenbock DT, Christ WJ, Gusovsky F. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J Biol Chem (1999) 274:10689–92. doi: 10.1074/jbc.274.16.10689

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Ciesielska A, Matyjek M, Kwiatkowska K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell Mol Life Sci (2021) 78:1233–61. doi: 10.1007/s00018-020-03656-y

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Ma F, Zhang J, Zhang J, Zhang C. The TLR7 agonists imiquimod and gardiquimod improve DC-based immunotherapy for melanoma in mice. Cell Mol Immunol (2010) 7:381–8. doi: 10.1038/cmi.2010.30

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Miller RL, Meng TC, Tomai MA. The antiviral activity of Toll-like receptor 7 and 7/8 agonists. Drug News Perspect (2008) 21:69–87. doi: 10.1358/dnp.2008.21.2.1188193

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Anderson JM, Rodriguez A, Chang DT. Foreign body reaction to biomaterials. Semin Immunol (2008) 20:86–100. doi: 10.1016/j.smim.2007.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Urban-Wojciuk Z, Khan MM, Oyler BL, Fahraeus R, Marek-Trzonkowska N, Nita-Lazar A, et al. The role of TLRs in anti-cancer immunity and tumor rejection. Front Immunol (2019) 10:2388. doi: 10.3389/fimmu.2019.02388

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Pahlavanneshan S, Sayadmanesh A, Ebrahimiyan H, Basiri M. Toll-like receptor-based strategies for cancer immunotherapy. J Immunol Res (2021) 2021:9912188. doi: 10.1155/2021/9912188

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: TLR - toll-like receptor, viral infection, innate immunity, inflammation, signaling pathway

Citation: Planz O and Kircheis R (2024) Editorial: The role of toll-like receptors and their related signaling pathways in viral infection and inflammation. Front. Immunol. 15:1363958. doi: 10.3389/fimmu.2024.1363958

Received: 31 December 2023; Accepted: 08 January 2024;
Published: 19 January 2024.

Edited and Reviewed by:

Francesca Granucci, University of Milano-Bicocca, Italy

Copyright © 2024 Planz and Kircheis. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Oliver Planz, oliver.planz@uni-tuebingen.de; Ralf Kircheis, rkircheis@syntacoll.de

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.