Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Elena Shklovskaya is active.

Publication


Featured researches published by Elena Shklovskaya.


Journal of Experimental Medicine | 2011

Cutaneous immunosurveillance by self-renewing dermal γδ T cells

Nital Sumaria; Ben Roediger; Lai Guan Ng; Jim Qin; Rachel Pinto; Lois L. Cavanagh; Elena Shklovskaya; Barbara Fazekas de St Groth; James A. Triccas; Wolfgang Weninger

The dermis contains a novel population of γδT cells that are distinct from epidermal γδT cells and produce IL-17 in response to mycobacterial infection.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Langerhans cells are precommitted to immune tolerance induction.

Elena Shklovskaya; Brendan J. O’Sullivan; Lai Guan Ng; Ben Roediger; Ranjeny Thomas; Wolfgang Weninger; Barbara Fazekas de St Groth

Antigen-dependent interactions between T lymphocytes and dendritic cells (DCs) can produce two distinct outcomes: tolerance and immunity. It is generally considered that all DC subsets are capable of supporting both tolerogenic and immunogenic responses, depending on their exposure to activating signals. Here, we tested whether epidermal Langerhans cells (LCs) can support immunogenic responses in vivo in the absence of antigen presentation by other DC subsets. CD4 T cells responding to antigen presentation by activated LCs initially proliferated but then failed to differentiate into effector/memory cells or to survive long term. The tolerogenic function of LCs was maintained after exposure to potent adjuvants and occurred despite up-regulation of the costimulatory molecules CD80, CD86, and IL-12, but was consistent with their failure to translocate the NF-κB family member RelB from the cytoplasm to the nucleus. Commitment of LCs to tolerogenic function may explain why commensal microorganisms expressing Toll-like receptor (TLR) ligands but confined to the skin epithelium are tolerated, whereas invading pathogens that breach the epithelial basement membrane and activate dermal DCs stimulate a strong immune response.


Journal of Immunology | 2008

Epidermal and Dermal Dendritic Cells Display Differential Activation and Migratory Behavior While Sharing the Ability to Stimulate CD4+ T Cell Proliferation In Vivo

Elena Shklovskaya; Ben Roediger; Barbara Fazekas de St Groth

Migrated Langerhans cells (m-LCs) have recently been shown to comprise only a minority of skin-derived dendritic cells (DCs) expressing Langerin in cutaneous lymph nodes. We have used BM chimeric mice that differ in CD45 and MHC class II alleles to unequivocally distinguish between radioresistant m-LCs and radiosensitive migrated dermal DCs (m-dDCs), to determine their phenotype, response to contact sensitization, and ability to activate naive CD4+ T cells in vivo. We have also characterized three subsets of dDCs and their migratory counterparts, as distinguished by expression of CD11b and Langerin. Each of the four subsets of skin DCs showed differential migration to draining LN in response to contact sensitizing agents. Migration of Langerin−CD11b+ and Langerin+CD11blow dDCs peaked after 1 day, followed by Langerin−CD11blow dDCs at 2 days and Langerin+ LCs at 4 days. Moreover, while m-LCs and m-dDCs had similar surface phenotypes in the steady state, they displayed unexpectedly different activation responses to contact sensitization: m-dDCs markedly up-regulated CD80 and CD86 at day 1, whereas only m-LCs up-regulated CD40, with delayed kinetics. Thus, m-dDCs are likely to be responsible for the initial response to skin immunization. However, when expression of cognate MHC class II was restricted to LCs and m-LCs, they were also capable of processing and presenting protein Ag to drive naive CD4 T cell proliferation in vivo. Thus, m-dDCs and m-LCs display distinct behavior in cutaneous lymph nodes while sharing the ability to interact specifically with T cells to control the immune response.


Journal of Immunology | 2009

Antigen Load Governs the Differential Priming of CD8 T Cells in Response to the Bacille Calmette Guérin Vaccine or Mycobacterium tuberculosis Infection

Anthony A. Ryan; Jonathan K. Nambiar; Teresa M. Wozniak; Ben Roediger; Elena Shklovskaya; Warwick J. Britton; Barbara Fazekas de St Groth; James A. Triccas

One reason proposed for the failure of Mycobacterium bovis bacille Calmette Guérin (BCG) vaccination to adequately control the spread of tuberculosis is a limited ability of the vaccine to induce effective CD8 T cell responses. However, the relative capacity of the BCG vaccine and virulent Mycobacterium tuberculosis to induce activation of CD8 T cells, and the factors that govern the initial priming of these cells after mycobacterial infection, are poorly characterized. Using a TCR transgenic CD8 T cell transfer model, we demonstrate significant activation of Ag-specific CD8 T cells by BCG, but responses were delayed and of reduced magnitude compared with those following infection with M. tuberculosis. The degree of CD8 T cell activation was critically dependent on the level of antigenic stimulation, as modifying the infectious dose to achieve comparable numbers of BCG or M. tuberculosis in draining lymph nodes led to the same pattern of CD8 T cell responses to both strains. Factors specific to M. tuberculosis infection did not influence the priming of CD8 T cells, as codelivery of M. tuberculosis with BCG did not alter the magnitude of BCG-induced T cell activation. Following transfer to RAG-1−/− recipients, BCG and M. tuberculosis-induced CD8 T cells conferred equivalent levels of protection against M. tuberculosis infection. These findings demonstrate that BCG is able to prime functional CD8 T cells, and suggest that effective delivery of Ag to sites of T cell activation by vaccines may be a key requirement for optimal CD8 T cell responses to control mycobacterial infection.


The Journal of Allergy and Clinical Immunology | 2015

IL-2 is a critical regulator of group 2 innate lymphoid cell function during pulmonary inflammation

Ben Roediger; Ryan Kyle; Szun Szun Tay; Andrew J. Mitchell; Holly A. Bolton; Thomas V. Guy; Sioh-Yang Tan; Elizabeth Forbes-Blom; Philip L. Tong; Yasmin Köller; Elena Shklovskaya; Makio Iwashima; Kathy D. McCoy; Graham Le Gros; Barbara Fazekas de St Groth; Wolfgang Weninger

BACKGROUND Group 2 innate lymphoid cells (ILC2) have been implicated in the pathogenesis of allergic lung diseases. However, the upstream signals that regulate ILC2 function during pulmonary inflammation remain poorly understood. ILC2s have been shown to respond to exogenous IL-2, but the importance of endogenous IL-2 in ILC2 function in vivo remains unclear. OBJECTIVE We sought to understand the role of IL-2 in the regulation of ILC2 function in the lung. METHODS We used histology, flow cytometry, immunohistochemistry, ELISA, and quantitative PCR with knockout and reporter mice to dissect pulmonary ILC2 function in vivo. We examined the role of ILC2s in eosinophilic crystalline pneumonia, an idiopathic type 2 inflammatory lung condition of mice, and the effect of IL-2 deficiency on this disease. We determined the effect of IL-2 administration on pulmonary ILC2 numbers and function in mice in the steady state and after challenge with IL-33. RESULTS We discovered an unexpected role for innate cell-derived IL-2 as a major cofactor of ILC2 function during pulmonary inflammation. Specifically, we found that IL-2 was essential for the development of eosinophilic crystalline pneumonia, a type 2 disease characterized by increased numbers of activated ILC2s. We show that IL-2 signaling serves 2 distinct functions in lung ILC2s, namely promoting cell survival/proliferation and serving as a cofactor for the production of type 2 cytokines. We further demonstrate that group 3 innate lymphoid cells are an innate immune source of IL-2 in the lung. CONCLUSION Innate cell-derived IL-2 is a critical cofactor in regulating ILC2 function in pulmonary type 2 pathology.


Journal of Immunology | 2007

Improved protection against disseminated tuberculosis by Mycobacterium bovis bacillus Calmette-Guérin secreting murine GM-CSF is associated with expansion and activation of APCs

Anthony A. Ryan; Teresa M. Wozniak; Elena Shklovskaya; Michael A. O’Donnell; Barbara Fazekas de St Groth; Warwick J. Britton; James A. Triccas

Modulating the host-immune response by the use of recombinant vaccines is a potential strategy to improve protection against microbial pathogens. In this study, we sought to determine whether secretion of murine GM-CSF by the bacillus Calmette-Guérin (BCG) vaccine influenced protective immunity against Mycobacterium tuberculosis. BCG-derived GM-CSF stimulated the in vitro generation of functional APCs from murine bone marrow precursors, as demonstrated by the infection-induced secretion of IL-12 by differentiated APCs, and the ability of these cells to present Ag to mycobacterium-specific T cells. Mice vaccinated with BCG-secreting murine GM-CSF (BCG:GM-CSF) showed increased numbers of CD11c+MHCII+ and CD11c−CD11b+F480+ cells compared with those vaccinated with control BCG, and this effect was most apparent in the draining lymph nodes at 7 and 14 days postvaccination. Vaccination with BCG:GM-CSF also resulted in enhanced expression of costimulatory molecules on migratory dendritic cells in the draining lymph nodes. The increased APC number was associated with an increase in the frequency of anti-mycobacterial IFN-γ-secreting T cells generated after BCG:GM-CSF vaccination compared with vaccination with control BCG, and this effect was sustained up to 17 wk in the spleens of immunized mice. Vaccination with BCG:GM-CSF resulted in an ∼10-fold increase in protection against disseminated M. tuberculosis infection compared with control BCG. This study demonstrates the potential of BCG-secreting immunostimulatory molecules as vaccines to protect against tuberculosis and suggests BCG:GM-CSF merits further appraisal as a candidate to control M. tuberculosis infection in humans.


Journal of Clinical Investigation | 2015

Selective Treg reconstitution during lymphopenia normalizes DC costimulation and prevents graft-versus-host disease.

Holly A. Bolton; Erhua Zhu; Alexandra M. Terry; Thomas V. Guy; Woon-Puay Koh; Sioh-Yang Tan; Carl A. Power; Patrick Bertolino; Katharina Lahl; Tim Sparwasser; Elena Shklovskaya; Barbara Fazekas de St Groth

Regulatory T cells (Tregs) have been shown to enhance immune reconstitution and prevent graft-versus-host disease (GVHD) after hematopoietic stem cell transplantation; however, it is unclear how Tregs mediate these effects. Here, we developed a model to examine the mechanism of Treg-dependent regulation of immune reconstitution. Lymphopenic mice were selectively reconstituted with Tregs prior to transfer of conventional CD4+ T cells. Full Treg reconstitution prevented the rapid oligoclonal proliferation that gives rise to pathogenic CD4 effector T cells, while preserving the slow homeostatic form of lymphopenia-induced peripheral expansion that repopulates a diverse peripheral T cell pool. Treg-mediated CTLA-4-dependent downregulation of CD80/CD86 on DCs was critical for inhibition of rapid proliferation and was a function of the Treg/DC ratio achieved by reconstitution. In an allogeneic BM transplant model, selective Treg reconstitution before T cell transfer also normalized DC costimulation and provided complete protection against GVHD. In contrast, cotransfer of Tregs was not protective. Our results indicate that achieving optimal recovery from lymphopenia should aim to improve early Treg reconstitution in order to increase the relative number of Tregs to DCs and thereby inhibit spontaneous oligoclonal T cell proliferation.


Infection and Immunity | 2007

Effects of DNA- and Mycobacterium bovis BCG-based delivery of the Flt3 ligand on protective immunity to Mycobacterium tuberculosis.

James A. Triccas; Elena Shklovskaya; Joanne M. Spratt; Anthony A. Ryan; Umaimainthan Palendira; Barbara Fazekas de StGroth; Warwick J. Britton

ABSTRACT The control of intracellular pathogens such as Mycobacterium tuberculosis is dependent on the activation and maintenance of pathogen-reactive T cells. Dendritic cells (DCs) are the major antigen-presenting cells initiating antimycobacterial T-cell responses in vivo. To investigate if immunization strategies that aim to optimize DC function can improve protective immunity against virulent mycobacterial infection, we exploited the ability of the hematopoietic growth factor Fms-like tyrosine kinase 3 ligand (Flt3L) to expand the number of DCs in vivo. A DNA fusion of the genes encoding murine Flt3L and M. tuberculosis antigen 85B stimulated enhanced gamma interferon (IFN-γ) release by T cells and provided better protection against virulent M. tuberculosis than DNA encoding the single components. Vaccination of mice with a recombinant Mycobacterium bovis BCG strain secreting Flt3L (BCG:Flt3L) led to early expansion of DCs compared to immunization with BCG alone, and this effect was associated with increased stimulation of BCG-reactive IFN-γ-secreting T cells. BCG and BCG:Flt3L provided similar protective efficacies against low-dose aerosol M. tuberculosis; however, immunization of immunodeficient mice revealed that BCG:Flt3L was markedly less virulent than conventional BCG. These results demonstrate the potential of in vivo targeting of DCs to improve antimycobacterial vaccine efficacy.


Journal of Immunology | 2006

Severely Impaired Clonal Deletion of CD4+ T Cells in Low-Dose Irradiated Mice: Role of T Cell Antigen Receptor and IL-7 Receptor Signals

Elena Shklovskaya; Barbara Fazekas de St Groth

Systemic administration of high doses of soluble Ag induces peripheral CD4+ T cell tolerance in unmanipulated hosts. To test whether tolerance is modified under conditions of transient lymphopenia, we tracked the response of 5C.C7 TCR-transgenic CD4+ T cells to i.v. moth cytochrome c peptide in mice that received low-dose gamma irradiation 10 days previously. This model was chosen because it does not support spontaneous lymphopenia-induced proliferation of 5C.C7 cells, allowing the study of Ag-specific responses without interference from simultaneous spontaneous proliferation. Clonal expansion in response to i.v. peptide was increased in irradiated mice, while clonal deletion was severely impaired in comparison with untreated animals. Amplified TCR triggering was observed in irradiated hosts, consistent with dendritic cell activation leading to enhanced Ag presentation. Failure of deletion was accompanied by persistent T cell activation and accumulation of Th1 effector cells. Up-regulated expression of IL-7R and the prosurvival protein Bcl-xL was associated with clonal persistence. Cells with memory and naive phenotypes were both represented within persistent clones, but no Th1 function could be demonstrated within the long-term memory population. Failure of clonal deletion in irradiated hosts represents a novel mechanism limiting TCR diversity in a lymphopenic environment and may contribute to subsequent autoimmunity.


Journal of Immunology | 2014

CD326 lo CD103 lo CD11b lo Dermal Dendritic Cells Are Activated by Thymic Stromal Lymphopoietin during Contact Sensitization in Mice

Sotaro Ochiai; Ben Roediger; Arby Abtin; Elena Shklovskaya; Barbara Fazekas de St Groth; Hidehiro Yamane; Wolfgang Weninger; Graham Le Gros; Franca Ronchese

The cytokine thymic stromal lymphopoietin (TSLP) is produced by epithelia exposed to the contact sensitizer dibutyl phthalate (DBP), and it is critical for the induction of Th2 immune responses by DBP-FITC. TSLP is thought to act on dendritic cells (DC), but the precise DC subsets involved in the response to TSLP remain to be fully characterized. In this study we show that a subset of CD326loCD103loCD11blo dermal DC, which we termed “triple-negative (TN) DC,” is highly responsive to TSLP. In DBP-FITC–treated mice, TN DC upregulated expression of CD86 and rapidly migrated to the draining lymph node to become the most abundant skin-derived DC subset at 24 and 48 h after sensitization. None of these responses was observed in TSLPR-deficient mice. In contrast, TN DC numbers were not increased after treatment with the allergen house dust mite or the bacteria Escherichia coli and bacillus Calmette–Guérin, which increased other DC subsets. In vivo, treatment with rTSLP preferentially increased the numbers of TN DC in lymph nodes. In vitro, TN DC responded to rTSLP treatment with a higher level of STAT5 phosphorylation compared with other skin-derived DC subsets. The TN DC subset shared the morphology, phenotype, and developmental requirements of conventional DC, depending on FLT3 expression for their optimal development from bone marrow precursors, and CCR7 for migration to the draining lymph node. Thus, TN DC represent a dermal DC subset that should be considered in future studies of TSLP-dependent contact sensitization and skin immune responses.

Collaboration


Dive into the Elena Shklovskaya's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge