Elsevier

Immunobiology

Volume 223, Issue 11, November 2018, Pages 608-617
Immunobiology

Human keratinocyte cultures (HaCaT) can be infected by DENV, triggering innate immune responses that include IFNλ and LL37

https://doi.org/10.1016/j.imbio.2018.07.006Get rights and content

Abstract

The skin is the first anatomical region that dengue virus (DENV) encounters during the natural infection. Although the role of some skin resident cells like dendritic cells and fibroblasts has been demonstrated to be crucial to elucidate the role of resident cells and molecules participating during the early events of the innate immune response, the participation of keratinocytes during DENV infection has not been fully elucidated. In this paper we aimed to evaluate the use of the HaCaT cell line as a model to study the immune responses of skin keratinocytes to DENV infection. We demonstrated productive DENV-2 infection of HaCaT cells and their capability to establish an antiviral response through production of type I and type III interferons (IFN-β and IFN-λ). The production of these cytokines by HaCaT cells correlated with upregulation of IFN-inducible transmembrane protein-3 (IFITM3) and viperin in bystander, uninfected cells. We also observed an increase in secretion of IL-6 and IL-8. Skin keratinocytes are known to secrete antimicrobial peptides (AMPs) during viral infections. In our model, DENV-2 infected HaCaT cells upregulate the production of cytoplasmic LL-37. We evaluated the dual role of LL-37, HBD2, and HBD3 antiviral activity and immunoregulation during DENV-2 infection of HaCaT cells and found that LL-37 significantly reduced DENV-2 replication. This indicates that the HaCaT cell line can be used as a model for studying the innate response of keratinocytes to DENV infection. Our results also suggest that skin keratinocytes play an important role in the skin microenvironment after DENV infection by secreting molecules like type I and type III IFNs, pro-inflammatory molecules, and LL-37, which may contribute to the protection against arboviral infections.

Introduction

Dengue viruses (DENV) are transmitted to humans by the Aedes mosquito, and all four serotypes (DENV 1–4) can cause disease (Gubler and Kuno, 1997). The skin is the first organ that DENV encounters after mosquitoe probing and blood feeding. Two of the most abundant cell types in the skin are keratinocytes and fibroblasts. We hypothesized that these cells could help restrict replication during DENV infection, depending on the inoculation route and intrinsic host variability (Garcia et al., 2017; Kupper and Fuhlbrigge, 2004; Nestle et al., 2009). Although dendritic cells and monocytes in the skin are considered important targets of DENV infection, the abundance of these cells is considerably low compared to keratinocytes and fibroblasts (Wu et al., 2000). In the dermis, fibroblasts have been found to be permissive to DENV infection and capable of establishing an antiviral microenvironment (Bustos-Arriaga et al., 2011; Kurane et al., 1992; Limon-Flores et al., 2005). Furthermore, data from a model of non-cadaveric human skin explants infected with DENV-2 suggested that keratinocytes were permissive to DENV infection in situ (Limon-Flores et al., 2005). Additionally, Surasombatpattana et. al. confirmed that primary cultures of keratinocytes are permissive to DENV infection (Surasombatpattana et al., 2011, 2012). The autocrine and paracrine activity of type I and type III interferons during viral infection is critical for restricting virus dissemination. In a previous study from our group, we observed that the release of IFN-β appears to be important part of the early immune response and effective control of DENV infection in primary fibroblasts (Bustos-Arriaga et al., 2011).

Three classes of IFN-λ have been identified (IFN-λ1, IFN-λ2, and IFN-λ3) and all of them play a role in the innate immune response to viral infections. Their receptors are largely restricted to cells of epithelial origin, such as keratinocytes (Ank et al., 2006; Donnelly and Kotenko, 2010; Kotenko et al., 2003). It has been shown that IFN-λ has antiviral activity against chikungunya virus, West Nile virus, and hepatitis C virus-the last two being members of the Flaviviridae family (Hsu et al., 2016; Kotenko et al., 2003; Liu et al., 2015; Ma et al., 2009). Recent reports indicate that human dendritic cells and human lung epithelial cells infected with DENV trigger the production of IFN-λ1 dependent toll-like receptor (TLR)-3, signaling pathway (Castaneda-Sanchez et al., 2016; Hsu et al., 2016; Palma-Ocampo et al., 2015) Type I, II, and III IFNs can induce transcription of thousands of IFN-stimulated genes (ISGs) (Helbig et al., 2013; MacMicking, 2012; Sadler and Williams, 2008). Some of these ISGs have broad-spectrum antiviral activity and can interfere with multiple steps of the virus life cycle (Chan et al., 2012; Jiang et al., 2010; Onoguchi et al., 2007).

Antimicrobial peptides (AMPs) constitute an early defence in epithelial and mucosal tissues, providing a rapid and broad-spectrum effect against pathogens, including viruses (Klotman and Chang, 2006; Nestle et al., 2009). AMPs can also modulate the immune response (Quinones-Mateu et al., 2003; Yang et al., 1999). Among the AMPs present in the skin, defensins and cathelicidin LL-37 are primarily expressed by keratinocytes. In addition to its microbicide activity, LL-37 also acts as a chemoattractant for neutrophils, monocytes, and T-cells (De et al., 2000). It has been reported that both THP-1 cells and human neutrophils produce HBD-1 and LL-37 following DENV infection (Castaneda-Sanchez et al., 2016). Furthermore, the treatment of DENV with LL37 prior infection of Vero cells significantly reduced levels of DENV genomic RNA and viral antigen (Alagarasu et al., 2017).

HaCaT cells are a widely used model in research involving keratinocytes and can produce type I and II IFNs, AMPs, HBDs, and LL-37. Therefore, to enrich the currently available information regarding the antiviral response in the skin and particularly the role of keratinocytes in the early stages of DENV infection, we evaluated the innate immune response to DENV infection of HaCaT cells.

Section snippets

Cells lines and virus

HaCaT cells, a non-tumorigenic, spontaneously immortalised human skin keratinocyte line were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Carlsbad, CA, USA) with 10% fetal bovine serum (FBS; Gibco) and l-glutamine (Gibco) in a humidified atmosphere of 5% CO2 at 37 °C.

C6/36 cells (Aedes albopictus mosquito cells) were maintained in supplemented minimum essential medium (MEM; Invitrogen, Grand Island, NY, USA) at 34 °C in the absence of CO2. The stock preparation and titration of

Infection of keratinocytes by DENV-2

To confirm productive infection of HaCaT cells with DENV-2, we performed immunofluorescence analysis at different time points. We observed the presence of the non-structural proteins the NS3 protease in the cytoplasm and NS5 RNA polymerase in the nucleus and cytoplasm. No signal was detected in mock-infected and uninfected controls (Fig. 1A). The percentage of infected cells was quantified by flow cytometry. At 24 h post infection, 10% of the cells expressed viral E protein and this increased

Discussion

Elucidating the contributions of each cell type present in the skin and their active interplay with DENV-infected and resident bystander cells will provide more insight into how DENV establishes infection in the skin (Bustos-Arriaga et al., 2015). Early stages of the in situ innate immunity to DENV infection might be crucial not just for the local response but may also be involved in defining the quality and intensity of the subsequent adaptive immunity.

Keratinocytes are crucial for innate

Competing interests

We declare that all authors, have no financial or personal relationships with other people or organizations that could create a potential conflict of interest or the appearance of a conflict of interest with regard to the work.

Acknowledgments

The authors would like to thanks to Victor Rosales-Garcia for the technical assistance Leticia Aleman Lazarini for the microscopy services and Sarah Alicia Gutierrez Cedillo for illustrate the Fig. 7 model. The National Council for Science and Technology (CONACyT Grant 0115401 CB). Additionally, MLG and DEMS received fellowships from CONACyT. JGC, LSA, LFR and LCB, are members of the National System of Researchers, SNI.

References (47)

  • J. Bustos-Arriaga et al.

    Activation of the innate immune response against DENV in normal non-transformed human fibroblasts

    PLoS Negl. Trop. Dis.

    (2011)
  • J. Bustos-Arriaga et al.

    Soluble mediators produced by the crosstalk between microvascular endothelial cells and dengue-infected primary dermal fibroblasts inhibit dengue virus replication and increase leukocyte transmigration

    Immunol. Res.

    (2015)
  • J.I. Castaneda-Sanchez et al.

    Expression of antimicrobial peptides in human monocytic cells and neutrophils in response to dengue virus type 2

    Intervirology

    (2016)
  • Y.K. Chan et al.

    IFITM proteins restrict antibody-dependent enhancement of dengue virus infection

    PLoS One

    (2012)
  • Y. De et al.

    LL-37, the neutrophil granule- and epithelial cell-derived cathelicidin, utilizes formyl peptide receptor-like 1 (FPRL1) as a receptor to chemoattract human peripheral blood neutrophils, monocytes, and T cells

    J. Exp. Med.

    (2000)
  • M.S. Diamond et al.

    Modulation of dengue virus infection in human cells by alpha, beta, and gamma interferons

    J. Virol.

    (2000)
  • R.P. Donnelly et al.

    Interferon-lambda: a new addition to an old family

    J. Interferon Cytokine Res.

    (2010)
  • P. Duangkhae et al.

    Interplay between keratinocytes and myeloid cells drives dengue virus spread in human skin

    J. Invest. Dermatol.

    (2017)
  • A. Egli et al.

    The impact of the interferon-lambda family on the innate and adaptive immune response to viral infections

    Emerg. Microbes Infect.

    (2014)
  • M. Garcia et al.

    Skin innate immune response to flaviviral infection

    Eur. Cytokine Netw.

    (2017)
  • J. Garcia-Cordero et al.

    Production and characterization of a monoclonal antibody specific for NS3 protease and the ATPase region of dengue-2 virus

    Hybridoma (Larchmt)

    (2005)
  • J. Garcia-Cordero et al.

    Generation and characterization of a rat monoclonal antibody against the RNA polymerase protein from dengue virus-2

    Immunol. Invest.

    (2014)
  • D.J. Gubler et al.

    Dengue and Dengue Hemorrhagic Fever

    (1997)
  • Cited by (17)

    View all citing articles on Scopus
    View full text