Network


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

Hotspot


Dive into the research topics where Sameh Basta is active.

Publication


Featured researches published by Sameh Basta.


Nature Medicine | 2001

A novel influenza A virus mitochondrial protein that induces cell death

Weisan Chen; Paul A. Calvo; Daniela Malide; James Gibbs; Ulrich Schubert; Igor Bacik; Sameh Basta; Robert E. O'Neill; Jeanne H. Schickli; Peter Palese; Peter Henklein; Jack R. Bennink; Jonathan W. Yewdell

While searching for alternative reading-frame peptides encoded by influenza A virus that are recognized by CD8+ T cells, we found an abundant immunogenic peptide encoded by the +1 reading frame of PB1. This peptide derives from a novel conserved 87-residue protein, PB1-F2, which has several unusual features compared with other influenza gene products in addition to its mode of translation. These include its absence from some animal (particularly swine) influenza virus isolates, variable expression in individual infected cells, rapid proteasome-dependent degradation and mitochondrial localization. Exposure of cells to a synthetic version of PB1-F2 induces apoptosis, and influenza viruses with targeted mutations that interfere with PB1-F2 expression induce less extensive apoptosis in human monocytic cells than those with intact PB1-F2. We propose that PB1-F2 functions to kill host immune cells responding to influenza virus infection.


Vaccine | 2008

TLR ligands and antigen need to be coencapsulated into the same biodegradable microsphere for the generation of potent cytotoxic T lymphocyte responses

Eva Schlosser; Marc Mueller; Stefan Fischer; Sameh Basta; Dirk H. Busch; Bruno Gander; Marcus Groettrup

Dendritic cells phagocytose pathogens leading to maturation and cross-presentation on MHC class I. We found that the efficiency of cross-priming in mice after vaccination with biodegradable poly(D,L-lactide-co-glycolide) microspheres (MSs) was enhanced when ovalbumin was coencapsulated together with either a CpG oligonucleotide or polyI:C as compared to co-inoculation of ovalbumin-bearing MS with soluble or separately encapsulated adjuvants. A single immunization with MS containing coencaspsulated CpG and ovalbumin yielded 9% SIINFEKL/H-2K(b) tetramer positive CTLs, production of IFN-gamma, efficient cytolysis, and protection from vaccinia virus infection. Taken together, coencapsulation of adjuvant and antigen is an important paradigm for the generation of potent CTL responses.


Cell | 2011

IL-7 Engages Multiple Mechanisms to Overcome Chronic Viral Infection and Limit Organ Pathology

Marc Pellegrini; Thomas Calzascia; Jesse G. Toe; Simon Preston; Amy E Lin; Alisha R. Elford; Arda Shahinian; Philipp A. Lang; Karl S. Lang; Michel Morre; Brigitte Assouline; Katharina Lahl; Tim Sparwasser; Thomas F. Tedder; Ji Hye Paik; Ronald A. DePinho; Sameh Basta; Pamela S. Ohashi; Tak W. Mak

Understanding the factors that impede immune responses to persistent viruses is essential in designing therapies for HIV infection. Mice infected with LCMV clone-13 have persistent high-level viremia and a dysfunctional immune response. Interleukin-7, a cytokine that is critical for immune development and homeostasis, was used here to promote immunity toward clone-13, enabling elucidation of the inhibitory pathways underlying impaired antiviral immune response. Mechanistically, IL-7 downregulated a critical repressor of cytokine signaling, Socs3, resulting in amplified cytokine production, increased T cell effector function and numbers, and viral clearance. IL-7 enhanced thymic output to expand the naive T cell pool, including T cells that were not LCMV specific. Additionally, IL-7 promoted production of cytoprotective IL-22 that abrogated liver pathology. The IL-7-mediated effects were dependent on endogenous IL-6. These attributes of IL-7 have profound implications for its use as a therapeutic in the treatment of chronic viral diseases.


Glycoconjugate Journal | 2009

Dependence of pathogen molecule-induced Toll-like receptor activation and cell function on Neu1 sialidase

Schammim Ray Amith; Preethi Jayanth; Susan Franchuk; Sarah Siddiqui; Volkan Seyrantepe; Katrina Gee; Sameh Basta; Rudi Beyaert; Alexey V. Pshezhetsky; Myron R. Szewczuk

The signaling pathways of mammalian Toll-like receptors (TLR) are well characterized, but the initial molecular mechanisms activated following ligand interactions with the receptors remain poorly defined. Here, we show a membrane controlling mechanism that is initiated by ligand binding to TLR-2, -3 and-4 to induce Neu1 sialidase activity within minutes in live primary bone marrow (BM) macrophage cells and macrophage and dendritic cell lines. Central to this process is that Neu1 and not Neu2,-3 and-4 forms a complex with TLR-2,-3 and-4 on the cell surface of naïve macrophage cells. Neuraminidase inhibitors BCX1827, 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (DANA), zanamivir and oseltamivir carboxylate have a limited significant inhibition of the LPS-induced sialidase activity in live BMC-2 macrophage cells but Tamiflu (oseltamivir phosphate) completely blocks this activity. Tamiflu inhibits LPS-induced sialidase activity in live BMC-2 cells with an IC50 of 1.2 μM compared to an IC50 of 1015 μM for its hydrolytic metabolite oseltamivir carboxylate. Tamiflu blockage of LPS-induced Neu1 sialidase activity is not affected in BMC-2 cells pretreated with anticarboxylesterase agent clopidogrel. Endotoxin LPS binding to TLR4 induces Neu1 with subsequent activation of NFκB and the production of nitric oxide and pro-inflammatory IL-6 and TNFα cytokines in primary and macrophage cell lines. Hypomorphic cathepsin A mice with a secondary Neu1 deficiency respond poorly to LPS-induced pro-inflammatory cytokines compared to the wild-type or hypomorphic cathepsin A with normal Neu1 mice. Our findings establish an unprecedented mechanism for pathogen molecule-induced TLR activation and cell function, which is critically dependent on Neu1 sialidase activity associated with TLR ligand treated live primary macrophage cells and macrophage and dendritic cell lines.


Journal of Immunology | 2004

Reversal in the Immunodominance Hierarchy in Secondary CD8+ T Cell Responses to Influenza A Virus: Roles for Cross-Presentation and Lysis-Independent Immunodomination

Weisan Chen; Ken C. Pang; Kelly-Anne Masterman; Gina Kennedy; Sameh Basta; Nektaria Dimopoulos; Felicita Hornung; Mark J. Smyth; Jack R. Bennink; Jonathan W. Yewdell

Immunodominance is a central feature of CD8+ T cell (TCD8+) responses to pathogens, transplants, and tumors. Determinants occupy a stable position in an immunodominance hierarchy (α-, β-, etc.) defined by the frequencies of responding TCD8+. In this paper, we study the mechanistic basis for place-swapping between α- (acid polymerase (PA)224–233) and β-determinants (nuclear protein 366–374) in primary vs secondary anti-influenza A virus (IAV) responses in mice. This phenomena was recently correlated with the inability of IAV-infected nondendritic cells (DCs) to generate PA224–233, and it was proposed that secondary TCD8+ are principally activated by IAV-infected epithelial cells, while primary TCD8+ are activated by IAV-infected DCs. In this study, we show that the inability of non-DCs to generate PA224–232 is relative rather than absolute, and that the preferential use of cross-priming in secondary anti-IAV responses can also account for the revised hierarchy. We further show that immunodomination of PA224–233-specific TCD8+ by nucleoprotein 366–374-specific TCD8+ plays a critical role in the phenomena, and that this is unlikely to be mediated by TCD8+ lysis of APCs or other cells.


Viral Immunology | 2003

A Survival Game of Hide and Seek: Cytomegaloviruses and MHC Class I Antigen Presentation Pathways

Sameh Basta; Jack R. Bennink

Cytomegaloviruses (CMV) are members of the ubiquitous family of herpesviruses, which escape immunological clearance and persist throughout life in the infected host. Cytomegaloviruses have developed numerous strategies that permit them to co-exist with their host even as an anti-virus immune response endangers their long-term survival. A considerable number of these strategies are aimed at MHC class I presentation of viral proteins to CD8+ T cells (TCD8+ ). Although the gamut of CMV immune evasion will be briefly examined, the primary focus of this review is on the host ability to counteract the strategies developed by CMV to inhibit antigen processing and presentation. A primary mechanism used by the immune system is the recognition of very early virus proteins including recognition of the immunomodulatory proteins themselves. We further speculate that cross-presentation of antigen is an adaptive immune response to the inhibition of direct presentation. Other mechanisms, such as the evolution of pAPC subsets, may also allow the immune system to adapt to a variety of different infectious pathogens while preventing cytopathic infection of all pAPCs.


Journal of Immunology | 2005

Cross-Presentation of the Long-Lived Lymphocytic Choriomeningitis Virus Nucleoprotein Does Not Require Neosynthesis and Is Enhanced via Heat Shock Proteins

Sameh Basta; Ricarda Stoessel; Michael Basler; Maries van den Broek; Marcus Groettrup

Many viral proteins that contain MHC class I-restricted peptides are long-lived, and it is elusive how they can give rise to class I epitopes. Recently, we showed that direct presentation of an epitope of the long-lived lymphocytic choriomeningitis virus nucleoprotein (LCMV-NP) required neosynthesis in accordance with the defective ribosomal products hypothesis. In this study, we report that LCMV-NP can be cross-primed in mice using either LCMV-NP-transfected human HEK293 or BALB/c-derived B8 cells as Ag donor cells. In addition, we establish that contrary to direct presentation, cross-presentation required accumulation of the mature LCMV-NP and could not be sustained by the newly synthesized LCMV-NP protein, intermediate proteasomal degradation products, or the minimal NP396 epitope. Nevertheless, NP cross-presentation was enhanced by heat shock and was blunted by inhibitors of heat shock protein 90 and gp96. We propose that cross-presentation has evolved to sustain the presentation of stable viral proteins when their neosynthesis has ceased in infected donor cells.


Scandinavian Journal of Immunology | 2007

The Cross‐priming Pathway: A Portrait of an Intricate Immune System

Sameh Basta; Attiya Alatery

Antigen presentation by professional antigen‐presenting cells (pAPCs) to cytotoxic CD8+ T cells can occur via two processing routes – the direct and cross‐presentation pathways. Cross‐presentation of exogenous antigens in the context of major histocompatibility complex (MHC) class I molecules has recently attracted a lot of research interest because it may prove crucial for vaccine development. This alternative pathway has been implicated in priming CD8+ T‐cell responses to pathogens as well as tumours in vivo (cross‐priming). In cross‐presentation, the internalized antigens can be processed through diverse intracellular routes. As many unresolved questions regarding the molecular basis that controls the cross‐priming process still exist, it is essential to explore the various elements involved therein, to better elucidate this pathway. In this review, we summarize current data that explore how the source and nature of antigens could affect their cross‐presentation. Moreover, we will discuss and outline how recent advances regarding pAPCs’ properties have increased our appreciation of the complex nature of the cross‐priming pathway in vivo. In conclusion, we contemplate how the direct and cross‐presentation pathways can function to allow the immune system to deal efficiently with diverse pathogens.


Journal of Immunology | 2002

Inhibitory Effects of Cytomegalovirus Proteins US2 and US11 Point to Contributions from Direct Priming and Cross-Priming in Induction of Vaccinia Virus-Specific CD8 + T Cells

Sameh Basta; Weisan Chen; Jack R. Bennink; Jonathan W. Yewdell

The extent to which naive CD8+ CTLs (TCD8+) are primed by APCs presenting endogenous Ags (direct priming) or Ags acquired from other infected cells (cross-priming) is a critical topic in basic and applied immunology. To examine the contribution of direct priming in the induction of VV-specific TCD8+, we generated recombinant vaccinia viruses that express human CMV proteins (US2 and US11) that induce the destruction of newly synthesized MHC class I molecules. Expression of US2 or US11 was associated with a 24–63% decrease in numbers of primary or secondary VV-specific TCD8+ responding to i.p. infection. Using HPLC-isolated peptides from VV-infected cells, we show that US2 and US11 selectively inhibit TCD8+ responses to a subset of immunogenic VV determinants. Moreover, VV-US2 and lysates from VV-infected histoincompatible cells elicit TCD8+ specific for a similar subset of VV determinants. These findings indicate that US2 and US11 can function in vivo to interfere with the activation of virus-specific TCD8+. Furthermore, they suggest that 1) both cross-priming and direct priming contribute significantly to the generation of VV-specific TCD8+, 2) the sets of immunogenic vaccinia virus determinants generated by cross-priming and direct priming are not completely overlapping, and 3) cross-priming overrides the effects of cis-acting viral interference with the class I Ag presentation pathway.


Journal of Immunology | 2012

IL-27 Enhances LPS-Induced Proinflammatory Cytokine Production via Upregulation of TLR4 Expression and Signaling in Human Monocytes

Christina Guzzo; Amit Ayer; Sameh Basta; Bruce W. Banfield; Katrina Gee

IL-27, which is produced by activated APCs, bridges innate and adaptive immunity by regulating the development of Th cells. Recent evidence supports a role for IL-27 in the activation of monocytic cells in terms of inflammatory responses. Indeed, proinflammatory and anti-inflammatory activities are attributed to IL-27, and IL-27 production itself is modulated by inflammatory agents such as LPS. IL-27 primes LPS responses in monocytes; however, the molecular mechanism behind this phenomenon is not understood. In this study, we demonstrate that IL-27 priming results in enhanced LPS-induced IL-6, TNF-α, MIP-1α, and MIP-1β expression in human primary monocytes. To elucidate the molecular mechanisms responsible for IL-27 priming, we measured levels of CD14 and TLR4 required for LPS binding. We determined that IL-27 upregulates TLR4 in a STAT3- and NF-κB–dependent manner. Immunofluorescence microscopy revealed enhanced membrane expression of TLR4 and more distinct colocalization of CD14 and TLR4 upon IL-27 priming. Furthermore, IL-27 priming enhanced LPS-induced activation of NF-κB family members. To our knowledge, this study is the first to show a role for IL-27 in regulating TLR4 expression and function. This work is significant as it reveals new mechanisms by which IL-27 can enhance proinflammatory responses that can occur during bacterial infections.

Collaboration


Dive into the Sameh Basta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jack R. Bennink

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jonathan W. Yewdell

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge