Loading

Review Article Open Access
Volume 4 | Issue 1 | DOI: https://doi.org/10.33696/immunology.4.127

Towards a Better Understanding of Staphylococcus aureus Skin Infections-The Interactions with Dendritic Cells

  • 1Laboratoire Microorganismes: Génome et Environnement, CNRS UMR 6023, Université Clermont-Auvergne, Clermont- Ferrand, France
  • 2Laboratoire Microbes Intestin Inflammation et Susceptibilité de l’Hôte, Inserm U1071, INRA USC 2018, Université Clermont-Auvergne, Clermont-Ferrand, France
+ Affiliations - Affiliations

*Corresponding Author

 Pascale Gueirard, pascale.gueirard@uca.fr

Received Date: October 30, 2021

Accepted Date: January 19, 2022

Abstract

Staphylococcus aureus (S. aureus) is the leading cause of skin and soft tissue infections in humans. Additionally, local infections further lead to dissemination and colonization of secondary infections sites including the lungs, heart valves and even medical prostheses. It is well known that this bacterial species is capable of altering host immune responses and that long-term protection against S. aureus is not completely effective. Antigen presenting cells represent key players in the implementation of these responses. Among these, dendritic cells (DCs) represent a wide variety of cell subsets that are heterogeneously distributed throughout the skin. As sentinels of the epithelial barrier, they are often the first cells in contact with pathogens and play a crucial role in the activation of specific T cell responses. In its planktonic form, the interactions between S. aureus and various DC cell subsets have been extensively studied in in vitro and ex vivo models. However,the tendency of bacteria to transition towards the biofilm lifestyle in the host suggest a necessity to study these interactions under in vivo conditions. Of note, mouse skin infection models provide a cheap and easy support to study the longitudinal responses of DCs through the use of fluorescence and intravital imaging. The development of models capable of comparing S. aureus planktonic and biofilm in vivo DC responses could prove essential in explaining the chronic nature of biofilms or the absence of an effective protective response. In this review, we highlight the different DC subsets found in the skin and their roles during S. aureus skin infections. We also address how this bacterium is capable of subverting these functions and review the known literature of existing mouse skin infection models that have and could potentially aid in the study of S. aureus planktonic or biofilm immune responses.

Keywords

Dendritic cells, Staphylococcus aureus, Skin, Immunity, Biofilm, In vivo models

 

Introduction

In healthy individuals, Staphylococcus aureus (S. aureus) can be found commensally in sebaceous sites in the skin microbiome [1]. Dysbiosis in the skin flora, abnormal skin barrier functions and immune abnormalities, such as in patients suffering from immunodeficiency or immunosuppressive treatments, predispose to the development of staphylococcal skin infections [2-4]. Indeed, these cocci are often isolated from patients suffering from chronic inflammatory skin diseases such as atopic dermatitis (AD) and recurring furunculosis, diseases that can be further exacerbated by the presence of S. aureus biofilms [3,4]. However, little is known about how this bacterial lifestyle interacts with the immune system in favor of chronicity [1,5].

The skin represents a specialized niche of immunity, as keratinocytes and Langerhans cells (LC) also participate in immune responses by the recognition of pathogen associated molecular patterns (PAMP), the antigen presentation and the secretion of antimicrobial peptides and cytokines [3,4]. The latter participate in the recruitment of professional phagocytes such as monocytes (MO), macrophages (MΦ) and polymorphonuclear neutrophils (PMN). These effector cells are implicated in the clearance of bacterial infections via phagocytosis and the formation of neutrophil or macrophage extracellular traps [6-8]. In addition, dendritic cells (DC) play the role of antigen presenting cells (APC) where they induce adaptive immune responses, that can be recalled in subsequent infections [9].

The adaptive cellular responses to S. aureus skin infections have mainly implicated T helper (Th)17 cells. Via the secretion of interleukin (IL)-17A and IL-17F, these cells enhance the barrier function and antimicrobial properties of epithelial cells, while also increasing recruitment of PMNs [9]. In a murine immunization model, Th1/Th17 populations were upregulated in skin lesions of immunized mice, accompanied by an increase in PMN recruitment, resulting in a decrease of skin lesion area and bacterial burden in comparison to naïve mice [10]. Supporting this concept, IL- 17A from γδ T cells has also been described in the control of S. aureus cutaneous infections [11,12]. Similarly, in patients with Hyper IgE Syndrome or HIV, characterized by impaired Th17 formation, an increased susceptibility to S. aureus skin infections is observed. However, the beneficial actions of these cells are not observed in the case of AD patients colonized by AD specific S. aureus strains where a Th2 immune environment is induced in lesional skin [3]. Outside of AD, a protective role of Th2 responses have been demonstrated in animal models [9,13].

Clearly, the mechanisms regulating T cell activation in the context of S. aureus skin infections remains unclear. Further, the presence of biofilms adds a second level of complexity to the subject. Although the impact of S. aureus biofilms in relation to skin infections is not well documented, the chronic nature of both entities suggests an impaired adaptive immune response incapable of mediating bacterial clearance. At the center of T cell coordination, DCs play a critical role as APCs that could potentially contribute to these ineffective responses.

In this review, we discuss the different DC populations in the skin and their responses towards S. aureus in a cutaneous setting. We address the mechanisms employed by S. aureus to counter DC functions and lastly, we review the known literature on the specific interactions between biofilms and DCs in the skin.

 

Dendritic Cells Subsets in the Skin

Skin is composed of two anatomical distinct layers: the epidermis and the dermis. The outer epidermis is a highly stratified-epithelium mainly composed of keratinocytes, and separated from the underlying dermis by the basement membrane. The dermis is made of various stromal cells that include fibroblasts, and houses blood and lymph vessels, sweat glands and hair follicles. Skin DCs are divided into LCs in the epidermis, and dermal DCs (dDCs) in the dermis that are closely related to conventional or classical DCs (cDCs) subsets present in lymphoid tissues. They represent heterogeneous populations of highly specialized phagocytic cells. As professional APCs, their main function is to transport cutaneous antigens to skin draining lymph nodes (dLN) and to present them to naïve CD4+ and CD8+ T cells to initiate adaptive immune responses. They also interact with skin resident and infiltrated cells at steady state or under inflammation to modulate both innate and adaptive immune responses.

Dendritic Cells at Steady State

At steady state, the only resident APCs in the epidermis are LCs, which represent 3-5% of epidermal cells, with approximately 700 LCs/mm2 [14]. They are related to MΦs and have unique properties. Indeed, they self-renew in the epidermis and originate, in mouse, from yolk sac- or fetal liverderived hematopoietic precursors rather than bone marrow Hematopoietic Stem Cells [15]. Compared with dermal cDCs, LCs express lower major histocompatibility complex class II (MHC-II) levels, intermediate CD11c levels and very high levels of the C-type lectin Langerin (CD207) [16,17]. In humans, LCs are further subdivided into LC1 and LC2 subsets that differ in abundance and also transcription profile [18]. As sentinel cells, they continuously sense the external environment and maintain tolerance to skin commensals by establishing connections with the surrounding epithelium via protruding dendrites [19,20]. When skin inflammation occurs, they take up antigen and transport it to the dLN to initiate T cell responses. Their migratory capacity to lymph nodes T cell zone is similar to that of cDCs [20,21].

Dermal DCs originate from blood-borne precursors known as pre-cDCs. In the skin, 3 distinct subsets of migratory dDCs are found: XCR1+ dDCs (or cDC1), CD11b+ dDCs (or cDC2) and double negative dDCs (For reviews see Malissen 2014 [23]; Kashem 2017 [15]; Collin 2018 [21]). CD11b- XCR1+ dDCs represent a minor population of DCs in human and mice that express high levels of CD207. They include CD103+ and CD103- cells. They are able to quickly migrate to the deep T cell area of the LN. CD11b+ dDCs are the most abundant sub-population of DCs in the dermis at steady state. They express the CD11b and CD1c markers respectively in mice and in humans, and other numerous markers both in human and mice as CX3CR1+, SIRP α+, CCR2+ and CD11c+. Classically, the presence of CD14 is used to distinguish MOs and MΦs from cDCs, although a novel CD1c+ CD14+ DC subset that remain phenotypically distinct from MOs, has been identified in the human dermis [22]. After migration, dDCs are directed into the peripheral paracortex of the LN. Double negative dDCs represent a small population of dDCs exclusively present in the mouse dermis. Their phenotype is XCR1 - CD207 - CD11blo [15,21,23].

Finally, LY6Chi MOs migrate from the blood circulation in a CC-chemokine receptor 2 (CCR2)-dependent manner to generate dermal monocyte-derived DCs (mo-DCs) that are detected in the skin at steady state. Their transcriptional program is similar to that of CD11b+ dDCs, but their capacity to migrate and activate CD4+ and CD8+ T lymphocytes is lower.

 

Dendritic Cells during Inflammation

Inflammatory DCs appear transiently when inflammation occurs. Contrary to LCs and dDCs which are present at steady state and enriched in inflammatory conditions, inflammatory DCs disappear when skin returns to a steady state, once infection is cured. They represent a highly heterogeneous populations of MHC-II+ CD11c+ cells, with no unique surface markers. Their origin is currently not well established, with a predominant hypothesis as MO-derived cells (For reviews see Kashem 2017 [15]; Collin 2018 [21]). During skin inflammation, they are recruited in a CCR2-dependent manner, and play a major role at the site of inflammation [24].

Plasmacytoid DCs (pDCs) are interferon alpha (IFNα- producing population of cells, with a similar origin as cDCs. They are detected in the blood circulation and in lymphoid tissues at steady state [15]. In humans, pDCs located in tonsillar crypts and oro-nasopharyngeal epithelium can be exposed to extracellular bacteria such as S. aureus [25]. During inflammation, they are also present in the skin, playing distinct roles in inflammatory skin diseases [15,26]. In mice, they express membrane markers shared with members of the B cell family, including B220 and Ly6C. In humans, pDCs are CD11clo, CD11b-, MHC-IIlow, CD123/IL- 3R+, CD303 (BDCA- 2)+ and CD304 (BDCA-4)+ [7,15]. When activated, they secrete high concentrations of IFNα, tumor necrosis factor alpha (TNFα and IL-6 and upregulate the CD86 membrane marker [25,27]. Additionally, Chen et al. (2020) have identified a transient pDC-like subset, that infiltrate human skin wounds, expressing CD11c+, HLA-DR+, CD123+ and CD1a+ [28].

 

Dendritic Cell Activation after S. aureus Recognition in the Skin

In the skin, cutaneous DCs can recognize S. aureus antigens through their toll like receptors (TLR). The most documented pattern recognition receptor (PRR) in this regard is TLR2, found on the plasma membrane, which can sense a diverse range of PAMPs, namely bacterial peptidoglycan (PGN), acetylated lipoproteins and lipoteichoic acid, the latter involving heterodimerization with TLR6 along with CD36 [29,30]. CD207 and macrophage galactose?type lectin (CD301) expressed in LCs and dDCs respectively have also been shown to interact with specific carbohydrate motifs located on S. aureus wall teichoic acids (WTA) [31,32]. The intracellular TLRs 8 and 9 recognize ssDNA and dsDNA respectively, but have not been described in human LCs [33].

The binding of PRRs to their PAMPs leads to the internalization of bacteria by phagocytosis, inducing DC maturation, characterized by the expression of MHC and costimulatory molecules, and their migration towards skin dLNs, where they present antigens to naïve T cells. The latter has also been shown to occur directly in the skin, following infection, where DC-T cell clusters were observed at the perivascular area in the dermis of various animal models [15,34]. Recently, the expression of activity-regulated cytoskeleton associated protein/activity-regulated gene 3.1 (Arc/Arg3.1) in a proportion of mice DCs and LCs was shown to enhance migratory capacity and CD4+ and CD8+ T cell activation in dLNs [35]. The differentiation of naïve T cells into the different effector T cell populations is influenced by the secretion of cytokines in addition to T cell receptor and co-stimulatory molecules activation. Indeed, activation of a particular PRR influences the downstream secretion of cytokines and chemokines, thus influencing the fate of the inflammatory environment [30]. Mouse LCs expressing human langerin secreted CXCL1 (KC), IL-6 and IL-17 in response to WTA β-GlcNAc while PGN, an agonist of TLR2, induced the secretion of IL-6, IL-10 and IL-8 but not IL-12 or IL-1β in LCs isolated from human skin [32,33]. Thus, LCs have been implicated in the differentiation of Th17 cells and also the accumulation of IL-17 producing γδ T cells in the skin [36,37]. In contrast, cDC1s have been shown to drive Th1 polarization via the cytokine IL-12 [15,34]. Activated EpCAM+CD59+Ly- 6D+ cDC1s also mediated PMN recruitment through the secretion of vascular endothelial growth factor α following intradermal challenge with S. aureus [38].

How Staphylococcus aureus Planktonic Bacteria Manipulate the Dendritic Cells

S. aureus has developed numerous strategies to avoid host immune attacks and modulate the establishment of adaptive immune responses (For a review, see Darisipudi et al., 2018 [7]; Wu et al., 2014 [25]). First, the bacteria produce numerous bicomponent pore-forming leukocidins as LukAB and LukED that target the CD11b and CCR5 receptors respectively, and lyse PMNs, MOs and MΦs, but also DCs, even after efficient phagocytosis [39-43].

Inside phagocytes, the bacteria are also able to survive and multiply. Several virulence factors are involved, as phenolsoluble modulin α (PSM α) peptides to counteract phagolysosomal acidification, the O-Acetyltransferase (OatA) to resist to lysozyme degradation and the Catalase (KatA), the Alkyl Hydroperoxide Reductase (AhpC) and the Staphylococcal Peroxidase Inhibitor (SPIN) to resist oxidative stress [44-47]. Even if DCs lack myeloperoxidase, the target of the SPIN protein, their functional capacity to prime T cells in the lymphoid tissues is impaired by the PMNderived protein [7]. PSM peptides have additional effects on DC activity, as PSM3 produced by community-associated methicillin-resistant S. aureus have the capacity to induce tolerogenic DCs upon TLR2 ligand stimulation of regulatory T cells via the activation of the p38-CREB pathway [48,49].

The bacteria cell wall is another key component of DC attenuated functional properties. In the skin, WTA producing S. aureus strains induce LC activation and maturation [50]. The lipoteichoic acid thereafter downregulates DC activation and antigen presenting activity via a TLR2 dependent signal pathway [51].

Another immune evasion strategy developed by S. aureus bacteria is DC modulation of cytokine production, by secreting several virulence factors as Esx factors, PSMs or alpha-toxin. In vitro, the two Esx proteins, EsxA and EsxB secreted by the type VII-like secretion system, reduce the production of Th1/Th17 pro-inflammatory cytokines by infected DCs [52]. PSMs are other key modulators of DC cytokines production [48,49,53]. In particular, they inhibit the production of IL- 32, IL-6 and IL-8 by epithelial cells, and IL-32 is involved in DC maturation [53]. In AD, toxins as the alpha-toxin first contribute to the induction of a Th1 like cytokine response [54]. Bacteria further skew the immune response toward Th2, with LCs producing high amounts of IL-2 and lower amounts of IFNγ, therefore contributing to bacterial persistence in the skin, and to chronicization of the inflammatory skin disorder [3,55,56].

Finally, S. aureus induces an overstimulation of the host immune system by producing numerous pyrogenic superantigens [57]. They do not affect any parameters of DC function but lead to an overstimulation of T cell lymphocytes [58].

 

Dendritic Cells and Staphylococcus aureus Chronic Skin Infections- In vivo Infection Models

A contributing factor to the pathogenicity of S. aureus is its capacity to transition towards the biofilm lifestyle. The latter are mono- or poly-species species microbial aggregates embedded in an extracellular matrix that can be found attached to a variety of surfaces, conferring protection to environmental dangers [59]. In humans, S. aureus can attach to either host proteins such as collagen and fibronectin or to inert surfaces namely, implanted medical devices in order to form biofilms [60]. In non-inflammatory conditions, S. aureus of the human microbiota have been shown to form biofilms [5]. In pathological conditions, S. aureus have been linked to the severity of certain inflammatory skin diseases namely AD [1]. Moreover, S. aureus clinical isolates from AD patients have been shown to be strong producers of biofilms [61,62]. The capacity of biofilms to survive the treatment of antimicrobials such as antibiotics and host anti-microbial peptides, while also resisting host immune attacks makes them strong contributors to the chronic nature of skin diseases [63,64]. Thus, the study of the in vivo interactions between S. aureus biofilms and specific actors of the adaptive immune system is essential in the understanding of S. aureus chronic infections. Review of the current literature has shown a lack of in vivo information on how DCs respond to biofilms and how this differs to planktonic bacteria [7,65].

Animal models, namely rodents, provide the complex interactions between host cells that are absent in in vitro conditions, while allowing the study of a variety of infections in different contexts, such as those present at the different depths of the skin, in wounds and also in the presence of implanted materiel [65-67]. Epicutaneous colonization models have demonstrated a beneficial effect of the oral administration of Lactococcus lactis on the maturation of pDCs in the skin dLN [68]. The langerin receptor can be used in inducible ablation models when expressed with the diphtheria toxin receptor (Langerin-diphtheria toxin receptor knock-in mice) to study the functional roles of LCs and cDC1s during inflammation [38]. To selectively deplete a subset of DCs, transcription factors specific to certain subsets can also be targeted, such as Batf3 in the case of cDC1s [38]. The development of humanized rodent models has led to the use of xenografts containing tissue resident LCs and T cells, while the use of transgenic mice constitutively expressing human langerin on LCs have shown an increase in inflammatory markers following the recognition of WTA [32,69]. The implication of downstream signaling pathways for both T cell polarization and innate immune activation, can further be studied in DC specific MyD88 KO mice [70]. Models where bacteria are introduced into the subcutaneous level of skin tissue have been essential in studying the effectors of innate immune memory. Indeed, after an initial subcutaneous inoculation of S. aureus in the flanks of mice, an increase in Langerin+ DC infiltration was observed in the abscess of mice during a subsequent infection compared to mice that never received the initial dose of bacteria [71,72]. Seeing as S. aureus preferentially transition towards the multicellular lifestyle when under harmful conditions in the host, it is likely that bacteria in colonized tissue contain a mixture of planktonic or biofilm bacteria or an intermediate between the two phenotypes. However, this parameter is rarely verified in in vivo models and thus, the specificity of the observed responses to either lifestyle is not clear.

In vivo biofilm infection models involve more invasive colonization of the skin tissue at the intradermal or the subcutaneous level of the skin. Biofilms are introduced either directly by inoculation into tissues [73-75] or by inserting abiotic supports, such as tissue cages, catheters or medical sutures, through incisions or pre-formed air pouches [76-78]. Abiotic supports are either sterile upon insertion and subsequently inoculated with bacteria to allow biofilm formation or are already pre-colonized by biofilms [79-81]. The common entry of S. aureus through wounds can be mimicked by mouse wound infection models which involve disruption of the skin barrier through skin incisions, external wounds or by scalding followed by bacterial inoculation [82-85]. However, the use of less invasive techniques has facilitated the monitoring of the evolution of infections; for example, using bioluminescent S. aureus strains to follow the bacterial charge at the infection site over time and to study their dispersion towards secondary sites in the same animal [78,82,83,86-88]. Intravital imaging of transgenic reporter mice, such as the LysM-EGFP or CX3CR1-EGFP mouse lines, allows insight into the interactions of immune cell populations, namely PMNs, MOs and DCs, with in vivo biofilms [75,89,90]. To visualize cDC populations specifically, CD11c-YFP reporter mice have been used in both bacterial and non-infectious models [91-93]. The application of multiphoton imaging in in vivo models has also improved our understanding of the interactions between DCs and T cells in the lymph node and could prove useful in highlighting differential adaptive immune responses between planktonic and biofilm S. aureus [94-97].

 

Conclusion

Biofilm specific immune evasion strategies, coupled with the rise in multidrug resistant S. aureus strains, have made it essential to develop alternative therapeutic strategies. Namely, anti-infectious immunotherapy has proven to be effective at controlling S. aureus implant associated biofilm infections in mice. For example, local administration of pre-activated inflammatory MΦs at biofilm infection sites or MO metabolic reprogramming by oligomycin containing nanoparticles, both amplify pro-inflammatory responses and promote biofilm clearance [98,99]. In the case of cancer immunotherapy, the manipulation of DCs is becoming a promising avenue of investigation [100]. The chronic nature of both biofilm infections and tumors may suggest common deficiencies in DC functionality and initiation of adaptive immunity. Further research is required to determine if DC manipulation can be applied as an alternative or adjuvant therapy to antibiotics treatments against biofilms.

 

Conflicts of Interest

The authors declare no conflict of interest.

Funding Statement

This work was supported by funds from the Auvergne- Rhône-Alpes (AURA) region (Pack Ambition Recherche 2017-IMMUNOFILM-Staph project) obtained by Pascale Gueirard.

 

Acknowledgments

We wish to thank Jérôme Josse (Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Lyon, France) for the helpful insight and discussions.

References

0.

1. Di Domenico EG, Cavallo I, Capitanio B, Ascenzioni F, Pimpinelli F, Morrone A, et al. Staphylococcus aureus and the Cutaneous Microbiota Biofilms in the Pathogenesis of Atopic Dermatitis. Microorganisms. 2019 Aug 29;7(9):301.

2. Ricciardi BF, Muthukrishnan G, Masters E, Ninomiya M, Lee CC, Schwarz EM. Staphylococcus aureus Evasion of Host Immunity in the Setting of Prosthetic Joint Infection: Biofilm and Beyond. Current Reviews in Musculoskeletal Medicine. 2018 Sep;11(3):389–400.

3. Iwamoto K, Moriwaki M, Miyake R, Hide M. Staphylococcus aureus in atopic dermatitis: Strain- specific cell wall proteins and skin immunity. Allergology International. 2019 Jul 1;68(3):309–15.

4. Nowicka D, Grywalska E. Staphylococcus aureus and Host Immunity in Recurrent Furunculosis. DRM. 2019;235(4):295–305.

5. Brandwein M, Steinberg D, Meshner S. Microbial biofilms and the human skin microbiome. Npj Biofilms Microbiomes. 2016 Nov 23;2(1):1–6.

6. Vor L de, Rooijakkers SHM, Strijp JAG van. Staphylococci evade the innate immune response by disarming neutrophils and forming biofilms. FEBS Letters. 2020;594(16):2556–69.

7. Darisipudi MN, Nordengrün M, Bröker BM, Péton V. Messing with the Sentinels—The Interaction of Staphylococcus aureus with Dendritic Cells. Microorganisms. 2018 Aug 15;6(3):87.

8. Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol. 2021 Jan 19;11:620339.

9. Karauzum H, Datta SK. Adaptive immunity against Staphylococcus aureus. Curr Top Microbiol Immunol. 2017;409:419–39.

10. Peng Z, Cao D-Y, Wu H-Y, Saito S. Immunization with a Bacterial Lipoprotein Establishes an Immuno-Protective Response with Upregulation of Effector CD4+ T Cells and Neutrophils Against Methicillin-Resistant Staphylococcus aureus Infection. Pathogens. 2020 Feb 20;9(2):138.

11. Cho JS, Pietras EM, Garcia NC, Ramos RI, Farzam DM, Monroe HR, et al. IL-17 is essential for host defense against cutaneous Staphylococcus aureus infection in mice. J Clin Invest. 2010 24 May 3;120(5):1762–73.

12. Dillen CA, Pinsker BL, Marusina AI, Merleev AA, Farber ON, Liu H, et al. Clonally expanded ?d T cells protect against Staphylococcus aureus skin reinfection. J Clin Invest. 2018 Mar 1;128(3):1026–42.

13. González JF, Hahn MM, Gunn JS. Chronic biofilm-based infections: skewing of the immune response. Pathog Dis. 2018 Mar 28;76(3):fty023.

14. Merad M, Ginhoux F, Collin M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol. 2008 Dec;8(12):935–47.

15. Kashem SW, Haniffa M, Kaplan DH. Antigen-Presenting Cells in the Skin. Annual Review of Immunology. 2017;35(1):469–99.

16. Nestle FO, Turka LA, Nickoloff BJ. Characterization of dermal dendritic cells in psoriasis. Autostimulation of T lymphocytes and induction of Th1 type cytokines. J Clin Invest. 1994 Jul 1;94(1):202–9.

17. Merad M, Sathe P, Helft J, Miller J, Mortha A. The Dendritic Cell Lineage: Ontogeny and Function of Dendritic Cells and Their Subsets in the Steady State and the Inflamed Setting. Annu Rev Immunol. 2013;31:10.1146/annurev-immunol-020711–74950.

18. Liu X, Zhu R, Luo Y, Wang S, Zhao Y, Qiu Z, et al. Distinct human Langerhans cell subsets orchestrate reciprocal functions and require different developmental regulation. Immunity. 2021 Oct 12;54(10):2305-2320.e11.

19. Nishibu A, Ward BR, Jester JV, Ploegh HL, Boes M, Takashima A. Behavioral Responses of Epidermal Langerhans Cells In Situ to Local Pathological Stimuli. Journal of Investigative Dermatology. 2006 Apr 1;126(4):787–96.

20. Kubo A, Nagao K, Yokouchi M, Sasaki H, Amagai M. External antigen uptake by Langerhans cells with reorganization of epidermal tight junction barriers. J Exp Med. 2009 Dec 21;206(13):2937– 46.

21. Collin M, Bigley V. Human dendritic cell subsets: an update. Immunology. 2018 May;154(1):3–20.

22. Cytlak U, Resteu A, Pagan S, Green K, Milne P, Maisuria S, et al. Differential IRF8 Transcription Factor Requirement Defines Two Pathways of Dendritic Cell Development in Humans. Immunity. 2020 Aug 18;53(2):353-370.e8.

23. Malissen B, Tamoutounour S, Henri S. The origins and functions of dendritic cells and macrophages in the skin. Nat Rev Immunol. 2014 Jun;14(6):417–28.

24. Guilliams M, van de Laar L. A Hitchhiker’s Guide to Myeloid Cell Subsets: Practical Implementation of a Novel Mononuclear Phagocyte Classification System. Front Immunol. 2015 Aug 11;6:406.

25. Wu X, Xu F. Dendritic cells during Staphylococcus aureus infection: subsets and roles. J Transl Med. 2014 Dec 18;12:358.

26. Wollenberg A, Günther S, Moderer M, Wetzel S, Wagner M, Towarowski A, et al. Plasmacytoid Dendritic Cells: A New Cutaneous Dendritic Cell Subset with Distinct Role in Inflammatory Skin Diseases. J Invest Dermatol. 2002 Nov 1;119(5):1096–102.

27. Michea P, Vargas P, Donnadieu M-H, Rosemblatt M, Bono MR, Duménil G, et al. Epithelial control of the human pDC response to extracellular bacteria. Eur J Immunol. 2013 May;43(5):1264–73.

28. Chen Y-L, Gomes T, Hardman CS, Vieira Braga FA, Gutowska- Owsiak D, Salimi M, et al. Re- evaluation of human BDCA-2+ DC during acute sterile skin inflammation. J Exp Med. 2019 Dec 17;217(3):jem.20190811.

29. Triantafilou M, Gamper FGJ, Haston RM, Mouratis MA, Morath S, Hartung T, et al. Membrane Sorting of Toll-like Receptor (TLR)-2/6 and TLR2/1 Heterodimers at the Cell Surface Determines Heterotypic Associations with CD36 and Intracellular Targeting *. Journal of Biological Chemistry. 2006 Oct 13;281(41):31002–11.

30. Miller LS. Toll-like receptors in skin. Adv Dermatol. 2008;24:71– 87.

31. Mnich ME, van Dalen R, Gerlach D, Hendriks A, Xia G, Peschel A, et al. The C-type lectin receptor MGL senses N-acetylgalactosamine on the unique Staphylococcus aureus ST395 wall teichoic acid. Cell Microbiol. 2019 Oct;21(10):e13072.

32. van Dalen R, De La Cruz Diaz JS, Rumpret M, Fuchsberger FF, van Teijlingen NH, Hanske J, et al. Langerhans Cells Sense Staphylococcus aureus Wall Teichoic Acid through Langerin To Induce Inflammatory Responses. mBio. 2019 May 14;10(3):e00330-19.

33. Flacher V, Bouschbacher M, Verronèse E, Massacrier C, Sisirak V, Berthier-Vergnes O, et al. Human Langerhans Cells Express a Specific TLR Profile and Differentially Respond to Viruses and Gram-Positive Bacteria. The Journal of Immunology. 2006 Dec 1;177(11):7959–67.

34. Honda T, Egawa G, Kabashima K. Antigen presentation and adaptive immune responses in skin. International Immunology. 2019 Jul 13;31(7):423–9.

35. Tintelnot J, Ufer F, Engler JB, Winkler H, Lücke K, Mittrücker H-W, et al. Arc/Arg3.1 defines dendritic cells and Langerhans cells with superior migratory ability independent of phenotype and ontogeny in mice. European Journal of Immunology. 2019;49(5):724–36.

36. Mathers AR, Janelsins BM, Rubin JP, Tkacheva OA, Shufesky WJ, Watkins SC, et al. Differential capability of human cutaneous dendritic cell subsets to initiate Th17 responses. J Immunol. 2009 Jan 15;182(2):921–33.

37. Kobayashi T, Glatz M, Horiuchi K, Kawasaki H, Akiyama H, Kaplan DH, et al. Dysbiosis and Staphylococcus aureus Colonization Drives Inflammation in Atopic Dermatitis. Immunity. 2015 Apr 21;42(4):756–66.

38. Janela B, Patel AA, Lau MC, Goh CC, Msallam R, Kong WT, et al. A Subset of Type I Conventional Dendritic Cells Controls Cutaneous Bacterial Infections through VEGFa-Mediated Recruitment of Neutrophils. Immunity. 2019 Apr;50(4):1069-1083.e8.

39. Dalla Serra M, Coraiola M, Viero G, Comai M, Potrich C, Ferreras M, et al. Staphylococcus aureus Bicomponent ?-Hemolysins, HlgA, HlgB, and HlgC, Can Form Mixed Pores Containing All Components. J Chem Inf Model. 2005 Nov;45(6):1539–45.

40. Meyer F, Girardot R, Piémont Y, Prévost G, Colin DA. Analysis of the Specificity of Panton- Valentine Leucocidin and Gamma-Hemolysin F Component Binding. Infect Immun. 2009 Jan;77(1):266–73.

41. DuMont AL, Yoong P, Liu X, Day CJ, Chumbler NM, James DBA, et al. Identification of a Crucial Residue Required for Staphylococcus aureus LukAB Cytotoxicity and Receptor Recognition. Infect Immun. 2014 Mar;82(3):1268–76.

42. Spaan AN, van Strijp JAG, Torres VJ. Leukocidins: Staphylococcal bi-component pore-forming toxins find their receptors. Nat Rev Microbiol. 2017 Jul;15(7):435–47.

43. Berends ETM, Zheng X, Zwack EE, Ménager MM, Cammer M, Shopsin B, et al. Staphylococcus aureus Impairs the Function of and Kills Human Dendritic Cells via the LukAB Toxin. mBio. 2019 Jan 2;10(1):e01918-18.

44. Bera A, Herbert S, Jakob A, Vollmer W, Götz F. Why are pathogenic staphylococci so lysozyme resistant? The peptidoglycan O-acetyltransferase OatA is the major determinant for lysozyme resistance of Staphylococcus aureus. Molecular Microbiology. 2005;55(3):778–87.

45. Cosgrove K, Coutts G, Jonsson I-M, Tarkowski A, Kokai-Kun JF, Mond JJ, et al. Catalase (KatA) and Alkyl Hydroperoxide Reductase (AhpC) Have Compensatory Roles in Peroxide Stress Resistance and Are Required for Survival, Persistence, and Nasal Colonization in Staphylococcus aureus. J Bacteriol. 2007 Feb;189(3):1025–35.

46. Grosz M, Kolter J, Paprotka K, Winkler A-C, Schäfer D, Chatterjee SS, et al. Cytoplasmic replication of Staphylococcus aureus upon phagosomal escape triggered by phenol-soluble modulin a. Cell Microbiol. 2014 Apr;16(4):451–65.

47. de Jong NWM, Ramyar KX, Guerra FE, Nijland R, Fevre C, Voyich JM, et al. Immune evasion by a staphylococcal inhibitor of myeloperoxidase. Proc Natl Acad Sci U S A. 2017 Aug 29;114(35):9439– 44.

48. Armbruster NS, Richardson JR, Schreiner J, Klenk J, Günter M, Autenrieth SE. Staphylococcus aureus PSM peptides induce tolerogenic dendritic cells upon treatment with ligands of extracellular and intracellular TLRs. Int J Med Microbiol. 2016 Dec;306(8):666–74.

49. Richardson JR, Armbruster NS, Günter M, Henes J, Autenrieth SE. Staphylococcus aureus PSM Peptides Modulate Human Monocyte- Derived Dendritic Cells to Prime Regulatory T Cells. Front Immunol. 2018 Nov 13;9:2603.

50. Hendriks A, van Dalen R, Ali S, Gerlach D, van der Marel GA, Fuchsberger FF, et al. Impact of Glycan Linkage to Staphylococcus aureus Wall Teichoic Acid on Langerin Recognition and Langerhans Cell Activation. ACS Infect Dis. 2021 Mar 12;7(3):624–35.

51. Saito S, Okuno A, Cao D-Y, Peng Z, Wu H-Y, Lin S-H. Bacterial Lipoteichoic Acid Attenuates Toll-Like Receptor Dependent Dendritic Cells Activation and Inflammatory Response. Pathogens. 2020 Oct 8;9(10):825.

52. Cruciani M, Etna MP, Camilli R, Giacomini E, Percario ZA, Severa M, et al. Staphylococcus aureus Esx Factors Control Human Dendritic Cell Functions Conditioning Th1/Th17 Response. Front Cell Infect Microbiol. 2017 Jul 21;7:330.

53. Deplanche M, Alekseeva L, Semenovskaya K, Fu C-L, Dessauge F, Finot L, et al. Staphylococcus aureus Phenol-Soluble Modulins Impair Interleukin Expression in Bovine Mammary Epithelial Cells. Infect Immun. 2016 May 24;84(6):1682–92.

54. Breuer K, Wittmann M, Kempe K, Kapp A, Mai U, Dittrich-Breiholz O, et al. Alpha-toxin is produced by skin colonizing Staphylococcus aureus and induces a T helper type 1 response in atopic dermatitis. Clinical & Experimental Allergy. 2005;35(8):1088–95.

55. Iwamoto K, Moriwaki M, Niitsu Y, Saino M, Takahagi S, Hisatsune J, et al. Staphylococcus aureus from atopic dermatitis skin alters cytokine production triggered by monocyte-derived Langerhans cell. Journal of Dermatological Science. 2017 Dec 1;88(3):271–9.

56. Iwamoto K, Nümm TJ, Koch S, Herrmann N, Leib N, Bieber T. Langerhans and inflammatory dendritic epidermal cells in atopic dermatitis are tolerized toward TLR2 activation. Allergy. 2018;73(11):2205–13.

57. Koymans KJ, Goldmann O, Karlsson CAQ, Sital W, Thänert R, Bisschop A, et al. The TLR2 Antagonist Staphylococcal Superantigen- Like Protein 3 Acts as a Virulence Factor to Promote Bacterial Pathogenicity in vivo. J Innate Immun. 2017;9(6):561–73.

58. Al-Shangiti AM, Nair SP, Chain BM. The interaction between staphylococcal superantigen-like proteins and human dendritic cells. Clin Exp Immunol. 2005 Jun;140(3):461–9.

59. Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent infections. Science. 1999 May 21;284(5418):1318–22.

60. Paharik AE, Horswill AR. The Staphylococcal Biofilm: Adhesins, Regulation, and Host Response. Microbiology Spectrum. 2016 Mar 18;4(2):4.2.06.

61. Smith K, Perez A, Ramage G, Lappin D, Gemmell CG, Lang S. Biofilm formation by Scottish clinical isolates of Staphylococcus aureus. J Med Microbiol. 2008 Aug;57(Pt 8):1018–23.

62. Allen HB, Vaze ND, Choi C, Hailu T, Tulbert BH, Cusack CA, et al. The Presence and Impact of Biofilm-Producing Staphylococci in Atopic Dermatitis. JAMA Dermatology. 2014 Mar 1;150(3):260–5.

63. Stewart PS, Costerton JW. Antibiotic resistance of bacteria in biofilms. The Lancet. 2001 Jul 14;358(9276):135–8.

64. Donlan RM, Costerton JW. Biofilms: Survival Mechanisms of Clinically Relevant Microorganisms. Clin Microbiol Rev. 2002 Apr;15(2):167–93.

65. Forestier C, Billard E, Milon G, Gueirard P. Unveiling and Characterizing Early Bilateral Interactions between Biofilm and the Mouse Innate Immune System. Front Microbiol. 2017 Nov 21;8:2309.

66. Youn C, Archer NK, Miller LS. Research Techniques Made Simple: Mouse Bacterial Skin Infection Models for Immunity Research. J Invest Dermatol. 2020 Aug;140(8):1488-1497.e1.

67. Klopfenstein N, Cassat JE, Monteith A, Miller A, Drury S, Skaar E, et al. Murine Models for Staphylococcal Infection. Current Protocols. 2021;1(3):e52.

68. Tsuji R, Fujii T, Nakamura Y, Yazawa K, Kanauchi O. Staphylococcus aureus Epicutaneous Infection Is Suppressed by Lactococcus lactis Strain Plasma via Interleukin 17A Elicitation. J Infect Dis. 2019 Jul 31;220(5):892–901.

69. Schulz A, Jiang L, Vor L de, Ehrström M, Wermeling F, Eidsmo L, et al. Neutrophil Recruitment to Noninvasive MRSA at the Stratum Corneum of Human Skin Mediates Transient Colonization. Cell Reports. 2019 Oct 29;29(5):1074-1081.e5.

70. Arnold-Schrauf C, Berod L, Sparwasser T. Dendritic cell specific targeting of MyD88 signalling pathways in vivo. European Journal of Immunology. 2015;45(1):32–9.

71. Chan LC, Rossetti M, Miller LS, Filler SG, Johnson CW, Lee HK, et al. Protective immunity in recurrent Staphylococcus aureus infection reflects localized immune signatures and macrophage-conferred memory. Proc Natl Acad Sci U S A. 2018 20;115(47):E11111–9.

72. Chan LC, Chaili S, Filler SG, Miller LS, Solis NV, Wang H, et al. Innate Immune Memory Contributes to Host Defense against Recurrent Skin and Skin Structure Infections Caused by Methicillin-Resistant Staphylococcus aureus. Infect Immun. 2017 Jan 26;85(2):e00876-16.

73. Yee R, Yuan Y, Shi W, Brayton C, Tarff A, Feng J, et al. Infection with Persister Forms of Staphylococcus aureus Causes a Persistent Skin Infection with More Severe Lesions in Mice: Failure to Clear the Infection by the Current Standard of Care Treatment. Discovery Medicine. 2019 Jul 26;28(151):7–16.

74. Abdul Hamid AI, Nakusi L, Givskov M, Chang Y-T, Marquès C, Gueirard P. A mouse ear skin model to study the dynamics of innate immune responses against Staphylococcus aureus biofilms. BMC Microbiol. 2020 Jan 29;20(1):22.

75. Sauvat L, Hamid AIA, Blavignac C, Josse J, Lesens O, Gueirard P. Biofilm-coated microbeads and the mouse ear skin: An innovative model for analysing anti-biofilm immune response in vivo. PLOS ONE. 2020 Dec 4;15(12):e0243500.

76. Woischnig A-K, Gonçalves LM, Ferreira M, Kuehl R, Kikhney J, Moter A, et al. Acrylic microparticles increase daptomycin intracellular and in vivo anti-biofilm activity against Staphylococcus aureus. Int J Pharm. 2018 Oct 25;550(1–2):372–9.

77. Markel DC, Bergum C, Wu B, Bou-Akl T, Ren W. Does Suture Type Influence Bacterial Retention and Biofilm Formation After Irrigation in a Mouse Model? Clin Orthop Relat Res. 2019 Jan;477(1):116–26.

78. Le H, Arnoult C, Dé E, Schapman D, Galas L, Le Cerf D, et al. Antibody-Conjugated Nanocarriers for Targeted Antibiotic Delivery: Application in the Treatment of Bacterial Biofilms. Biomacromolecules. 2021 Apr 12;22(4):1639–53.

79. Yamada KJ, Heim CE, Aldrich AL, Gries CM, Staudacher AG, Kielian T. Arginase-1 Expression in Myeloid Cells Regulates Staphylococcus aureus Planktonic but Not Biofilm Infection. Infect Immun. 2018 Jun 21;86(7):e00206-18.

80. He L, Le KY, Khan BA, Nguyen TH, Hunt RL, Bae JS, et al. Resistance to leukocytes ties benefits of quorum-sensing dysfunctionality to biofilm infection. Nat Microbiol. 2019 Jul;4(7):1114–9.

81. Narayana JL, Mishra B, Lushnikova T, Golla RM, Wang G. Modulation of antimicrobial potency of human cathelicidin peptides against the ESKAPE pathogens and in vivo efficacy in a murine catheter-associated biofilm model. Biochim Biophys Acta Biomembr. 2019 Sep 1;1861(9):1592–602.

82. Anderson LS, Reynolds MB, Rivara KR, Miller LS, Simon SI. A Mouse Model to Assess Innate Immune Response to Staphylococcus aureus Infection. J Vis Exp. 2019 Feb 28;(144):10.3791/59015.

83. Hoffmann JP, Friedman JK, Wang Y, McLachlan JB, Sammarco MC, Morici LA, et al. In situ Treatment With Novel Microbiocide Inhibits Methicillin Resistant Staphylococcus aureus in a Murine Wound Infection Model. Front Microbiol. 2020 Jan 23;10:3106.

84. Li J, Zhong W, Zhang K, Wang D, Hu J, Chan-Park MB. Biguanide-Derived Polymeric Nanoparticles Kill MRSA Biofilm and Suppress Infection In Vivo. ACS Appl Mater Interfaces. 2020 May 13;12(19):21231–41.

85. Wang B, Yao Y, Wei P, Song C, Wan S, Yang S, et al. Housefly Phormicin inhibits Staphylococcus aureus and MRSA by disrupting biofilm formation and altering gene expression in vitro and in vivo. International Journal of Biological Macromolecules. 2021 Jan 15;167:1424–34.

86. Archer NK, Wang Y, Ortines RV, Liu H, Nolan SJ, Liu Q, et al. Preclinical Models and Methodologies for Monitoring Staphylococcus aureus Infections Using Noninvasive Optical Imaging. Methods Mol Biol. 2020;2069:197–228.

87. Quan K, Jiang G, Liu J, Zhang Z, Ren Y, Busscher HJ, et al. Influence of interaction between surface-modified magnetic nanoparticles with infectious biofilm components in artificial channel digging and biofilm eradication by antibiotics in vitro and in vivo. Nanoscale. 2021 Mar 4;13(8):4644–53.

88. Redman WK, Welch GS, Rumbaugh KP. Assessing Biofilm Dispersal in Murine Wounds. J Vis Exp. 2021 Aug 7;(174).

89. Gries CM, Rivas Z, Chen J, Lo DD. Intravital Multiphoton Examination of Implant-AssociatedStaphylococcus aureus Biofilm Infection. Front Cell Infect Microbiol. 2020 Oct 15;10:574092.

90. Abdul Hamid AI, Cara A, Diot A, Laurent F, Josse J, Gueirard P. Differential Early in vivo Dynamics and Functionality of Recruited Polymorphonuclear Neutrophils After Infection by Planktonic or Biofilm Staphylococcus aureus. Frontiers in Microbiology. 2021;12:2471.

91. Abtin A, Jain R, Mitchell AJ, Roediger B, Brzoska AJ, Tikoo S, et al. Perivascular macrophages mediate neutrophil recruitment during bacterial skin infection. Nat Immunol. 2014 Jan;15(1):45–53.

92. Ortiz G, Chao C, Jamali A, Seyed-Razavi Y, Kenyon B, Harris DL, et al. Effect of Dry Eye Disease on the Kinetics of Lacrimal Gland Dendritic Cells as Visualized by Intravital Multi-Photon Microcopy. Front Immunol. 2020 Aug 12;11:1713.

93. Jamali A, Seyed-Razavi Y, Chao C, Ortiz G, Kenyon B, Blanco T, et al. Intravital Multiphoton Microscopy of the Ocular Surface: Alterations in Conventional Dendritic Cell Morphology and Kinetics in Dry Eye Disease. Front Immunol. 2020 May 7;11:742.

94. Kitano M, Yamazaki C, Takumi A, Ikeno T, Hemmi H, Takahashi N, et al. Imaging of the cross- presenting dendritic cell subsets in the skin-draining lymph node. Proc Natl Acad Sci U S A. 2016 Jan 26;113(4):1044–9.

95. Okada T, Takahashi S, Ishida A, Ishigame H. In vivo multiphoton imaging of immune cell dynamics. Pflugers Arch. 2016;468(11):1793–801.

96. van Panhuys N. Studying Dendritic Cell-T Cell Interactions Under In Vivo Conditions. Methods Mol Biol. 2017;1584:569–83.

97. Akkaya B, Kamenyeva O, Kabat J, Kissinger R. Visualizing the Dynamics of T Cell-Dendritic Cell Interactions in Intact Lymph Nodes by Multiphoton Confocal Microscopy. Methods Mol Biol. 2021;2304:243–63.

98. Hanke ML, Heim CE, Angle A, Sanderson SD, Kielian T. Targeting macrophage activation for the prevention and treatment of Staphylococcus aureus biofilm infections. J Immunol. 2013 Mar 1;190(5):2159–68.

99. Yamada KJ, Heim CE, Xi X, Attri KS, Wang D, Zhang W, et al. Monocyte metabolic reprogramming promotes pro-inflammatory activity and Staphylococcus aureus biofilm clearance. PLOS Pathogens. 2020 Mar 6;16(3):e1008354.

100. Gardner A, de Mingo Pulido Á, Ruffell B. Dendritic Cells and Their Role in Immunotherapy. Front Immunol. 2020 May 21;11:924.

Author Information X