Network


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

Hotspot


Dive into the research topics where Leonid A. Pobezinsky is active.

Publication


Featured researches published by Leonid A. Pobezinsky.


Blood | 2012

Basis of CTLA-4 function in regulatory and conventional CD4+ T cells

Xuguang Tai; François Van Laethem; Leonid A. Pobezinsky; Terry I. Guinter; Susan O. Sharrow; Anthony Adams; Larry Granger; Michael J. Kruhlak; Tullia Lindsten; Craig B. Thompson; Lionel Feigenbaum; Alfred Singer

CTLA-4 proteins contribute to the suppressor function of regulatory T cells (Tregs), but the mechanism by which they do so remains incompletely understood. In the present study, we assessed CTLA-4 protein function in both Tregs and conventional (Tconv) CD4(+) T cells. We report that CTLA-4 proteins are responsible for all 3 characteristic Treg functions of suppression, TCR hyposignaling, and anergy. However, Treg suppression and anergy only required the external domain of CTLA-4, whereas TCR hyposignaling required its internal domain. Surprisingly, TCR hyposignaling was neither required for Treg suppression nor anergy because costimulatory blockade by the external domain of CTLA-4 was sufficient for both functions. We also report that CTLA-4 proteins were localized in Tregs in submembrane vesicles that rapidly recycled to/from the cell surface, whereas CTLA-4 proteins in naive Tconv cells were retained in Golgi vesicles away from the cell membrane and had no effect on Tconv cell function. However, TCR signaling of Tconv cells released CTLA-4 proteins from Golgi retention and caused activated Tconv cells to acquire suppressor function. Therefore, the results of this study demonstrate the importance of intracellular localization for CTLA-4 protein function and reveal that CTLA-4 protein externalization imparts suppressor function to both regulatory and conventional CD4(+) T cells.


Immunity | 2012

αβ T cell receptors that do not undergo major histocompatibility complex-specific thymic selection possess antibody-like recognition specificities.

Anastasia N. Tikhonova; François Van Laethem; Ken-ichi Hanada; Jinghua Lu; Leonid A. Pobezinsky; Changwan Hong; Terry I. Guinter; Susanna Jeurling; Günter Bernhardt; Jung-Hyun Park; James Chih-Hsin Yang; Peter D. Sun; Alfred Singer

Major histocompatibility complex (MHC) restriction is the cardinal feature of T cell antigen recognition and is thought to be intrinsic to αβ T cell receptor (TCR) structure because of germline-encoded residues that impose MHC specificity. Here, we analyzed αβTCRs from T cells that had not undergone MHC-specific thymic selection. Instead of recognizing peptide-MHC complexes, the two αβTCRs studied here resembled antibodies in recognizing glycosylation-dependent conformational epitopes on a native self-protein, CD155, and they did so with high affinity independently of MHC molecules. Ligand recognition was via the αβTCR combining site and involved the identical germline-encoded residues that have been thought to uniquely impose MHC specificity, demonstrating that these residues do not only promote MHC binding. This study demonstrates that, without MHC-specific thymic selection, αβTCRs can resemble antibodies in recognizing conformational epitopes on MHC-independent ligands.


Nature Immunology | 2013

IL-7 signaling must be intermittent, not continuous, during CD8 + T cell homeostasis to promote cell survival instead of cell death

Motoko Kimura; Leonid A. Pobezinsky; Terry I. Guinter; Julien Thomas; Anthony Adams; Jung-Hyun Park; Xuguang Tai; Alfred Singer

The maintenance of naive CD8+ T cells is necessary for lifelong immunocompetence but for unknown reasons requires signaling via both interleukin 7 (IL-7) and the T cell antigen receptor (TCR). We now report that naive CD8+ T cells required IL-7 signaling to be intermittent, not continuous, because prolonged IL-7 signaling induced naive CD8+ T cells to proliferate, produce interferon-γ (IFN-γ) and undergo IFN-γ-triggered cell death. Homeostatic engagement of the TCR interrupted IL-7 signaling and thereby supported the survival and quiescence of CD8+ T cells. However, CD8+ T cells with insufficient TCR affinity for self ligands received prolonged IL-7 signaling and died during homeostasis. In this study we identified regulation of the duration of IL-7 signaling by homeostatic engagement of the TCR as the basis for in vivo CD8+ T cell homeostasis.


Nature Immunology | 2015

Let-7 microRNAs target the lineage-specific transcription factor PLZF to regulate terminal NKT cell differentiation and effector function

Leonid A. Pobezinsky; Ruth Etzensperger; Susanna Jeurling; Amala Alag; Tejas Kadakia; Tom McCaughtry; Motoko Kimura; Susan O. Sharrow; Terry I. Guinter; Lionel Feigenbaum; Alfred Singer

Lethal-7 (let-7) microRNAs (miRNAs) are the most abundant miRNAs in the genome, but their role in developing thymocytes is unclear. We found that let-7 miRNAs targeted Zbtb16 mRNA, which encodes the lineage-specific transcription factor PLZF, to post-transcriptionally regulate PLZF expression and thereby the effector functions of natural killer T cells (NKT cells). Dynamic upregulation of let-7 miRNAs during the development of NKT thymocytes downregulated PLZF expression and directed their terminal differentiation into interferon-γ (IFN-γ)-producing NKT1 cells. Without upregulation of let-7 miRNAs, NKT thymocytes maintained high PLZF expression and terminally differentiated into interleukin 4 (IL-4)-producing NKT2 cells or IL-17-producing NKT17 cells. Upregulation of let-7 miRNAs in developing NKT thymocytes was signaled by IL-15, vitamin D and retinoic acid. Such targeting of a lineage-specific transcription factor by miRNA represents a previously unknown level of developmental regulation in the thymus.


Cell | 2013

Lck Availability during Thymic Selection Determines the Recognition Specificity of the T Cell Repertoire

François Van Laethem; Anastasia N. Tikhonova; Leonid A. Pobezinsky; Xuguang Tai; Motoko Kimura; Cecile Le Saout; Terry I. Guinter; Anthony Adams; Susan O. Sharrow; Günter Bernhardt; Lionel Feigenbaum; Alfred Singer

Thymic selection requires signaling by the protein tyrosine kinase Lck to generate T cells expressing αβ T cell antigen receptors (TCR). For reasons not understood, the thymus selects only αβTCR that are restricted by major histocompatibility complex (MHC)-encoded determinants. Here, we report that Lck proteins that were coreceptor associated promoted thymic selection of conventionally MHC-restricted TCR, but Lck proteins that were coreceptor free promoted thymic selection of MHC-independent TCR. Transgenic TCR with MHC-independent specificity for CD155 utilized coreceptor-free Lck to signal thymic selection in the absence of MHC, unlike any transgenic TCR previously described. Thus, the thymus can select either MHC-restricted or MHC-independent αβTCR depending on whether Lck is coreceptor associated or coreceptor free. We conclude that the intracellular state of Lck determines the specificity of thymic selection and that Lck association with coreceptor proteins during thymic selection is the mechanism by which MHC restriction is imposed on a randomly generated αβTCR repertoire.


Journal of Experimental Medicine | 2005

Restricted MHC-peptide repertoire predisposes to autoimmunity.

Nadezda N. Logunova; Christophe Viret; Leonid A. Pobezinsky; Sara A. Miller; Dmitri B. Kazansky; John P. Sundberg; Alexander V. Chervonsky

MHC molecules associated with autoimmunity possess known structural features that limit the repertoire of peptides that they can present. Such limitation gives a selective advantage to TCRs that rely on interaction with the MHC itself, rather than with the peptide residues. At the same time, negative selection is impaired because of the lack of negatively selecting peptide ligands. The combination of these factors may predispose to autoimmunity. We found that mice with an MHC class II–peptide repertoire reduced to a single complex demonstrated various autoimmune reactions. Transgenic mice bearing a TCR (MM14.4) cloned from such a mouse developed severe autoimmune dermatitis. Although MM14.4 originated from a CD4+ T cell, dermatitis was mediated by CD8+ T cells. It was established that MM14.4+ is a highly promiscuous TCR with dual MHC class I/MHC class II restriction. Furthermore, mice with a limited MHC–peptide repertoire selected elevated numbers of TCRs with dual MHC class I/MHC class II restriction, a likely source of autoreactivity. Our findings may help to explain the link between MHC class I responses that are involved in major autoimmune diseases and the well-established genetic linkage of these diseases with MHC class II.


Science Signaling | 2014

T cell receptor stimulation impairs IL-7 receptor signaling by inducing expression of the microRNA miR-17 to target Janus kinase 1.

Gil Katz; Leonid A. Pobezinsky; Susanna Jeurling; Miho Shinzawa; François Van Laethem; Alfred Singer

Induction of a microRNA that targets a critical kinase enables the T cell receptor to inhibit cytokine receptor signaling. Cutting Out a Kinase for T cell Survival Although the survival of T cells requires signaling by both the T cell receptor (TCR) and the interleukin-7 receptor (IL-7R), stimulation of the TCR intermittently inhibits IL-7R signaling, because constant IL-7R signaling causes T cells to die. The kinase Jak1 (Janus kinase 1) is required for IL-7R signaling, and Katz et al. found that Jak1 was unstable and low in abundance in T cells. Furthermore, activation of the TCR induced the expression of a microRNA that prevented the synthesis of new Jak1 protein, thereby interfering with the ability of IL-7R to signal. T cell receptor (TCR)–mediated inhibition of interleukin-7 (IL-7) signaling is important for lineage fate determination in the thymus and for T cell survival in the periphery because uninterrupted IL-7 signaling results in T cell death. The initial event in IL-7 signaling is the transactivation of Janus kinases 1 and 3 (Jak1 and Jak3), which are associated with the cytosolic tails of the IL-7 receptor α chain (IL-7Rα) and the γc subunit, the two cell surface proteins that constitute IL-7R. We found that Jak1 is a highly unstable protein with a half-life of only 1.5 hours, so that continuous Jak1 protein synthesis is required to maintain Jak1 protein in sufficient abundance to support IL-7 signaling. However, we also found that Jak1 protein synthesis was acutely reduced by TCR-responsive microRNAs in the miR-17 family, which targeted Jak1 mRNA (messenger RNA) to inhibit its translation. Thus, this study identifies a molecular mechanism by which TCR engagement acutely disrupts IL-7 signaling.


eLife | 2017

Modulation of let-7 miRNAs controls the differentiation of effector CD8 T cells

Alexandria C Wells; Keith A. Daniels; Constance C Angelou; Eric Fagerberg; Amy S. Burnside; Michele Markstein; Dominique Alfandari; Raymond M. Welsh; Elena Pobezinskaya; Leonid A. Pobezinsky

The differentiation of naive CD8 T cells into effector cytotoxic T lymphocytes upon antigen stimulation is necessary for successful antiviral, and antitumor immune responses. Here, using a mouse model, we describe a dual role for the let-7 microRNAs in the regulation of CD8 T cell responses, where maintenance of the naive phenotype in CD8 T cells requires high levels of let-7 expression, while generation of cytotoxic T lymphocytes depends upon T cell receptor-mediated let-7 downregulation. Decrease of let-7 expression in activated T cells enhances clonal expansion and the acquisition of effector function through derepression of the let-7 targets, including Myc and Eomesodermin. Ultimately, we have identified a novel let-7-mediated mechanism, which acts as a molecular brake controlling the magnitude of CD8 T cell responses. DOI: http://dx.doi.org/10.7554/eLife.26398.001


Molecular Biology | 2008

Coreceptor function of CD4 in response to the MHC class I molecule

E. S. Zvezdova; T. S. Grinenko; E. L. Pobezinskaya; Leonid A. Pobezinsky; D. B. Kazansky

The specificity of the T-cell receptor (TCR) and its interaction with coreceptors play a crucial role in T-cell passing through developmental checkpoints and, eventually, determine the efficiency of adaptive immunity. The genes for the α and β chains of TCR were cloned from T-cell hybridoma 1D1, which was obtained by fusion of BWZ.36CD8α cells with CD8+ memory cells specific for the H-2Kb MHC class I molecule. Retroviral transduction of the 1D1 TCR genes and the CD4 and CD8 coreceptor genes was used to obtain 4G4 thymoma variants that exposed the CD3/TCR complex together with CD4, CD8, or both of the coreceptors on their surface. Although the main function of CD4 is to stabilize the interaction of TCR with MHC class II molecules, CD4 was found to mediate the activation of transfected cells via TCR specific for the H-2Kb MHC class I molecule. Moreover, CD4 proved to dominate over CD8, since the response of CD4+CD8+ transfectants was suppressed by antibodies against CD4 and the Ab MHC class II molecule but not to CD8. The response of CD4+ transfectants was not due to a cross-reaction of 1D1 TCR with MHC class II molecules, because the transfectants did not respond to splenocytes of H-2b knockout mice, which were defective in the assembly of the MHC class I molecule/β2 microglobulin/peptide complex and did not expose the complex on cell surface. The domination was not due to sequestration of p56lck kinase, since CD4 devoid of the kinase-binding site was functional in 4G4 thymoma cells. The results were used to explain some features of intrathymic cell selection and assumed to provide an experimental basis for developing new methods of anticancer gene therapy.


Science Signaling | 2014

Science Signaling Podcast: 26 August 2014

Alfred Singer; Leonid A. Pobezinsky; Gil Katz; Annalisa M. VanHook

Signaling through the T cell receptor stimulates production of a microRNA that inhibits interleukin-7 receptor signaling. This Podcast features an interview with Alfred Singer, Gil Katz, and Leonid Pobezinsky, authors of a Research Article that appears in the 26 August 2014 issue of Science Signaling, about a microRNA that plays a role in the survival of T cells. During development in the thymus and after maturation and migration into the periphery, T cells require signaling through both the T cell receptor (TCR) and the interleukin-7 receptor (IL-7R) to survive. However, because sustained IL-7R signaling kills T cells, TCR signaling periodically inhibits IL-7R signaling to enable T cell survival. Katz et al. have identified a microRNA that is induced by T cell receptor activation and inhibits IL-7R signaling by reducing the abundance of Jak1, a kinase that transduces signaling downstream of IL-7R.

Collaboration


Dive into the Leonid A. Pobezinsky's collaboration.

Top Co-Authors

Avatar

Alfred Singer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Terry I. Guinter

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

François Van Laethem

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lionel Feigenbaum

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Xuguang Tai

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Anthony Adams

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jung-Hyun Park

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Susan O. Sharrow

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Susanna Jeurling

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge