Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
AAPS J ; 24(2): 42, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35288790

ABSTRACT

The COVID-19 pandemic has strained the biological matrix supply chain. An upsurge in demand driven by numerous COVID-19 therapeutic and vaccine development programs to combat the pandemic, along with logistical challenges sourcing and transporting matrix, has led to increased lead times for multiple matrices. Biological matrix shortages can potentially cause significant delays in drug development programs across the pharmaceutical and biotechnology industry. Given the current circumstances, discussion is warranted around what will likely be increased use of surrogate matrices in support of pharmacokinetic (PK), immunogenicity, and biomarker assays for regulatory filings. Regulatory authorities permit the use of surrogate matrix in bioanalytical methods in instances where matrix is rare or difficult to obtain, as long as the surrogate is appropriately selected and scientifically justified. Herein, the scientific justification and possible regulatory implications of employing surrogate matrix in PK, immunogenicity, and biomarker assays are discussed. In addition, the unique challenges that cell and gene therapy (C>) and other innovative therapeutic modalities place on matrix supply chains are outlined. Matrix suppliers and contract research organizations (CROs) are actively implementing mitigation strategies to alleviate the current strain on the matrix supply chain and better prepare the industry for any future unexpected strains. To maintain ethical standards, these mitigation strategies include projecting matrix needs with suppliers at least 6 months in advance and writing or updating study protocols to allow for additional matrix draws from study subjects and/or re-purposing of subject matrix from one drug development program to another.


Subject(s)
COVID-19 , Pandemics , Humans
2.
Cancer Immunol Res ; 5(12): 1062-1073, 2017 12.
Article in English | MEDLINE | ID: mdl-29097419

ABSTRACT

Although CD8+ T cells are critical for controlling tumors, how they are recruited and home to primary and metastatic lesions is incompletely understood. We characterized the homing receptor (HR) ligands on tumor vasculature to determine what drives their expression and their role in T-cell entry. The anatomic location of B16-OVA tumors affected the expression of E-selectin, MadCAM-1, and VCAM-1, whereas the HR ligands CXCL9 and ICAM-1 were expressed on the vasculature regardless of location. VCAM-1 and CXCL9 expression was induced by IFNγ-secreting adaptive immune cells. VCAM-1 and CXCL9/10 enabled CD8+ T-cell effectors expressing α4ß1 integrin and CXCR3 to enter both subcutaneous and peritoneal tumors, whereas E-selectin enabled E-selectin ligand+ effectors to enter subcutaneous tumors. However, MadCAM-1 did not mediate α4ß7+ effector entry into peritoneal tumors due to an unexpected lack of luminal expression. These data establish the relative importance of certain HRs expressed on activated effectors and certain HR ligands expressed on tumor vasculature in the effective immune control of tumors. Cancer Immunol Res; 5(12); 1062-73. ©2017 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/etiology , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Adaptive Immunity , Animals , Biomarkers , Cell Line, Tumor , Chemokine CXCL9/genetics , E-Selectin/genetics , E-Selectin/metabolism , Integrin alpha4beta1/genetics , Ligands , Melanoma, Experimental , Mice , Models, Biological , Receptors, Lymphocyte Homing/metabolism , Vascular Cell Adhesion Molecule-1/genetics
3.
Nat Commun ; 6: 6771, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25857745

ABSTRACT

Lymphatic endothelial cells (LECs) directly express peripheral tissue antigens and induce CD8 T-cell deletional tolerance. LECs express MHC-II molecules, suggesting they might also tolerize CD4 T cells. We demonstrate that when ß-galactosidase (ß-gal) is expressed in LECs, ß-gal-specific CD8 T cells undergo deletion via the PD-1/PD-L1 and LAG-3/MHC-II pathways. In contrast, LECs do not present endogenous ß-gal in the context of MHC-II molecules to ß-gal-specific CD4 T cells. Lack of presentation is independent of antigen localization, as membrane-bound haemagglutinin and I-Eα are also not presented by MHC-II molecules. LECs express invariant chain and cathepsin L, but not H2-M, suggesting that they cannot load endogenous antigenic peptides onto MHC-II molecules. Importantly, LECs transfer ß-gal to dendritic cells, which subsequently present it to induce CD4 T-cell anergy. Therefore, LECs serve as an antigen reservoir for CD4 T-cell tolerance, and MHC-II molecules on LECs are used to induce CD8 T-cell tolerance via LAG-3.


Subject(s)
Antigen Presentation/genetics , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Peripheral Tolerance/genetics , Animals , Antigens/genetics , Antigens/immunology , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cathepsin L/genetics , Cathepsin L/immunology , Clonal Anergy/genetics , Dendritic Cells/cytology , Endothelial Cells/cytology , Gene Expression , Hemagglutinins/genetics , Hemagglutinins/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , beta-Galactosidase/genetics , beta-Galactosidase/immunology , Lymphocyte Activation Gene 3 Protein
4.
PLoS One ; 9(2): e87740, 2014.
Article in English | MEDLINE | ID: mdl-24503860

ABSTRACT

Peripheral self-tolerance eliminates lymphocytes specific for tissue-specific antigens not encountered in the thymus. Recently, we demonstrated that lymphatic endothelial cells in mice directly express peripheral tissue antigens, including tyrosinase, and induce deletion of specific CD8 T cells via Programmed Death Ligand-1 (PD-L1). Here, we demonstrate that high-level expression of peripheral tissue antigens and PD-L1 is confined to lymphatic endothelial cells in lymph nodes, as opposed to tissue (diaphragm and colon) lymphatics. Lymphatic endothelial cells in the lymph node medullary sinus express the highest levels of peripheral tissue antigens and PD-L1, and are the only subpopulation that expresses tyrosinase epitope. The representation of lymphatic endothelial cells in the medullary sinus expressing high-level PD-L1, which is necessary for normal CD8 T cell deletion kinetics, is controlled by lymphotoxin-ß receptor signaling and B cells. Lymphatic endothelial cells from neonatal mice do not express high-level PD-L1 or present tyrosinase epitope. This work uncovers a critical role for the lymph node microenvironment in endowing lymphatic endothelial cells with potent tolerogenic properties.


Subject(s)
Cellular Microenvironment , Endothelial Cells/metabolism , Endothelium, Lymphatic/metabolism , Immune Tolerance , Lymph Nodes/immunology , Lymph Nodes/metabolism , Animals , Animals, Newborn , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Adhesion Molecules/metabolism , Cellular Microenvironment/immunology , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Knockout , Monophenol Monooxygenase/metabolism , Mucoproteins , Phenotype , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
5.
Article in English | MEDLINE | ID: mdl-25580369

ABSTRACT

Lymphatic endothelial cells are most often thought of as structural cells that form the lymphatic vasculature, which transports fluid out of peripheral tissues and transports antigens and antigen presenting cells to lymph nodes. Recently, it has been shown that lymphatic endothelial cells also dynamically respond to and influence the immune response in several ways. Here, we describe how lymphatic endothelial cells induce peripheral T-cell tolerance and how this relates to tolerance induced by other types of antigen presenting cells. Furthermore, the ability of lymphatic endothelial cells to alter immune responses under steady-state or inflammatory conditions is explored, and the therapeutic potential of bypassing lymphatic endothelial cell-induced tolerance to enhance cancer immunotherapy is discussed.

6.
J Reprod Immunol ; 100(1): 66-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24080233

ABSTRACT

CD4+ CD25+ regulatory T cells (Tregs) strongly influence the early and late autoimmune responses to meiotic germ cell antigens (MGCA) and the gonadal immunopathology in vasectomized mice. This is supported by the published and recently acquired information presented here. Within 24h of unilateral vasectomy (uni-vx) the ipsilateral epididymis undergoes epithelial cell apoptosis followed by necrosis, severe inflammation, and granuloma formation. Unexpectedly, vasectomy alone induced MGCA-specific tolerance. In contrast, uni-vx plus simultaneous Treg depletion resulted in MGCA-specific autoimmune response and bilateral autoimmune orchitis. Both tolerance and autoimmunity were strictly linked to the early epididymal injury. We now discovered that testicular autoimmunity in uni-vx mice did not occur when Treg depletion was delayed by one week. Remarkably, this delayed Treg depletion also prevented tolerance induction. Therefore, tolerance depends on a rapid de novo Treg response to MGCA exposed after vasectomy. Moreover, tolerance was blunted in mice genetically deficient in PD-1 ligand, suggesting the involvement of induced Treg. We conclude that pre-existing natural Treg prevents post-vasectomy autoimmunity, whereas vasectomy-induced Treg maintains post-vasectomy tolerance. We further discovered that vasectomized mice were still resistant to autoimmune orchitis induction for at least 12-16 months; thus, tolerance is long-lasting. Although significant sperm autoantibodies of low titers became detectable in uni-vx mice at 7 months, the antibody titers fluctuated over time, suggesting a dynamic "balance" between the autoimmune and tolerance states. Finally, we observed severe epididymal fibrosis and hypo-spermatogenesis at 12 months after uni-vx: findings of highly critical clinical significance.


Subject(s)
Epididymis/pathology , Orchitis/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Vasectomy , Animals , Autoantibodies/blood , Autoimmunity/genetics , CD4 Antigens/metabolism , Fibrosis/etiology , Humans , Immune Tolerance/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Depletion , Male , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orchitis/etiology , Programmed Cell Death 1 Receptor/genetics , Vasectomy/adverse effects
7.
Front Immunol ; 3: 305, 2012.
Article in English | MEDLINE | ID: mdl-23060883

ABSTRACT

The lymphatic vasculature provides routes for dendritic cell and lymphocyte migration into and out of lymph nodes. Lymphatic endothelial cells (LEC) control these processes by expression of CCL21, sphingosine-1-phosphate, and adhesion molecules. LEC express MHC-I and MHC-II, but not costimulatory molecules, and present antigen on MHC-I via both direct and cross-presentation. Whether LEC present to CD4 T cells on MHC-II is unknown. Interestingly, LEC express antigens otherwise restricted to a small number of peripheral tissues in an autoimmune regulatory element-independent manner. Direct presentation of peripheral tissue antigens (PTA) to CD8 T cells results in abortive proliferation and deletion, due to both a lack of costimulation and active PD-L1 engagement. Autoimmunity develops when deletion is subverted, suggesting that LEC presentation of PTA could lead to human disease if PD-1 signaling were impaired by genetic polymorphisms, or aberrant costimulation occurred during inflammation. The expression of additional inhibitory molecules, which are not involved in LEC-mediated deletion, suggests that LEC may have additional immunoregulatory roles. LEC express receptors for several immunomodulatory molecules whose engagement alters their phenotype and function. In this review we describe the role of LEC in distinct anatomical locations in controlling immune cell trafficking, as well as their emerging role in the regulation of T cell tolerance and immunity.

8.
Blood ; 120(24): 4772-82, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22993390

ABSTRACT

Lymphatic endothelial cells (LECs) induce peripheral tolerance by direct presentation to CD8 T cells (T(CD8)). We demonstrate that LECs mediate deletion only via programmed cell death-1 (PD-1) ligand 1, despite expressing ligands for the CD160, B- and T-lymphocyte attenuator, and lymphocyte activation gene-3 inhibitory pathways. LECs induce activation and proliferation of T(CD8), but lack of costimulation through 4-1BB leads to rapid high-level expression of PD-1, which in turn inhibits up-regulation of the high-affinity IL-2 receptor that is necessary for T(CD8) survival. Rescue of tyrosinase-specific T(CD8) by interference with PD-1 or provision of costimulation results in autoimmune vitiligo, demonstrating that LECs are significant, albeit suboptimal, antigen-presenting cells. Because LECs express numerous peripheral tissue antigens, lack of costimulation coupled to rapid high-level up-regulation of inhibitory receptors may be generally important in systemic peripheral tolerance.


Subject(s)
B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Endothelial Cells/immunology , Immune Tolerance/immunology , Programmed Cell Death 1 Receptor/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Endothelial Cells/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphatic Vessels/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/immunology , Monophenol Monooxygenase/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, OX40/immunology , Receptors, OX40/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Vitiligo/genetics , Vitiligo/immunology , Vitiligo/metabolism
9.
J Exp Med ; 207(4): 681-8, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-20308365

ABSTRACT

Peripheral immune tolerance is generally thought to result from cross-presentation of tissue-derived proteins by quiescent tissue-resident dendritic cells to self-reactive T cells that have escaped thymic negative selection, leading to anergy or deletion. Recently, we and others have implicated the lymph node (LN) stroma in mediating CD8 T cell peripheral tolerance. We demonstrate that LN-resident lymphatic endothelial cells express multiple peripheral tissue antigens (PTAs) independent of the autoimmune regulator (Aire). They directly present an epitope derived from one of these, the melanocyte-specific protein tyrosinase, to tyrosinase-specific CD8 T cells, leading to their deletion. We also show that other LN stromal subpopulations express distinct PTAs by mechanisms that vary in their Aire dependence. These results establish lymphatic endothelial cells, and potentially other LN-resident cells, as systemic mediators of peripheral immune tolerance.


Subject(s)
Antigen Presentation/immunology , Endothelial Cells/immunology , Immune Tolerance/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Transcription Factors/genetics , Animals , Antigens, Neoplasm/genetics , Autoantigens/genetics , Autoantigens/immunology , Autoantigens/metabolism , Cell Proliferation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Gene Expression/genetics , Gene Expression/immunology , Glutamate Decarboxylase/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Immunophenotyping , Lymphocyte Activation/immunology , MART-1 Antigen , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/immunology , Monophenol Monooxygenase/metabolism , Neoplasm Proteins/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Stromal Cells/cytology , Stromal Cells/immunology , Stromal Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , AIRE Protein
10.
PLoS Pathog ; 5(5): e1000457, 2009 May.
Article in English | MEDLINE | ID: mdl-19478869

ABSTRACT

Virus-specific CD8(+) T cells (T(CD8+)) are initially triggered by peptide-MHC Class I complexes on the surface of professional antigen presenting cells (pAPC). Peptide-MHC complexes are produced by two spatially distinct pathways during virus infection. Endogenous antigens synthesized within virus-infected pAPC are presented via the direct-presentation pathway. Many viruses have developed strategies to subvert direct presentation. When direct presentation is blocked, the cross-presentation pathway, in which antigen is transferred from virus-infected cells to uninfected pAPC, is thought to compensate and allow the generation of effector T(CD8+). Direct presentation of vaccinia virus (VACV) antigens driven by late promoters does not occur, as an abortive infection of pAPC prevents production of these late antigens. This lack of direct presentation results in a greatly diminished or ablated T(CD8+) response to late antigens. We demonstrate that late poxvirus antigens do not enter the cross-presentation pathway, even when identical antigens driven by early promoters access this pathway efficiently. The mechanism mediating this novel means of viral modulation of antigen presentation involves the sequestration of late antigens within virus factories. Early antigens and cellular antigens are cross-presented from virus-infected cells, as are late antigens that are targeted to compartments outside of the virus factories. This virus-mediated blockade specifically targets the cross-presentation pathway, since late antigen that is not cross-presented efficiently enters the MHC Class II presentation pathway. These data are the first to describe an evasion mechanism employed by pathogens to prevent entry into the cross-presentation pathway. In the absence of direct presentation, this evasion mechanism leads to a complete ablation of the T(CD8+) response and a potential replicative advantage for the virus. Such mechanisms of viral modulation of antigen presentation must also be taken into account during the rational design of antiviral vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Vaccinia virus/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/virology , Cell Line , Histocompatibility Antigens Class I , Histocompatibility Antigens Class II , Humans
11.
Immunology ; 125(4): 480-91, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18489571

ABSTRACT

CD8(+) T cells (T(CD8+)) differentiate into effector cells following recognition of specific peptide-major histocompatibility complex (MHC) class I complexes (pMHC-I) on the surface of professional APCs (pAPCs), such as dendritic cells. Antigenic pMHC-I can be generated from two spatially distinct sources. The direct presentation pathway involves generation of peptide from protein substrate synthesized within the cell that is presenting the pMHC-I. Alternatively, the cross presentation pathway involves presentation of antigen that is not synthesized within the presenting cell, but is derived from exogenous proteins synthesized within other donor cells. The mechanisms by which cross presentation of exogenous antigens occur in vivo remain controversial. The C-type lectin scavenger receptor A (SR-A) has been implicated in a number of potential cross presentation pathways, including the presentation of peptide bound to heat shock proteins, such as glycoprotein 96 (gp96), and the transfer of pMHC-I from a donor cell to the pAPC. We demonstrate here that initiation of T(CD8+) responses is normal in mice lacking SR-A, and that the redundancy of ligand binding exhibited by the SR family is likely to be an important mechanism that ensures cross presentation in vivo. These observations emphasize the requirement to target multiple receptors and antigen-processing pathways during the rational design of vaccines aimed at eliciting protective T(CD8+).


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Membrane Glycoproteins/metabolism , Scavenger Receptors, Class A/metabolism , Adoptive Transfer/methods , Animals , Antigen Presentation , Calreticulin/immunology , Cell Line , Cross-Priming , Electroporation , Female , Histocompatibility Antigens Class I , Immunologic Memory , Interferon-gamma/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Orthomyxoviridae/immunology , Ovalbumin , Receptors, Antigen, T-Cell/genetics , Scavenger Receptors, Class A/genetics , Vaccinia virus/immunology
12.
J Exp Med ; 204(6): 1475-85, 2007 Jun 11.
Article in English | MEDLINE | ID: mdl-17548520

ABSTRACT

The deubiquitinating enzyme CYLD has recently been implicated in the regulation of signal transduction, but its physiological function and mechanism of action are still elusive. In this study, we show that CYLD plays a pivotal role in regulating T cell activation and homeostasis. T cells derived from Cyld knockout mice display a hyperresponsive phenotype and mediate the spontaneous development of intestinal inflammation. Interestingly, CYLD targets a ubiquitin-dependent kinase, transforming growth factor-beta-activated kinase 1 (Tak1), and inhibits its ubiquitination and autoactivation. Cyld-deficient T cells exhibit constitutively active Tak1 and its downstream kinases c-Jun N-terminal kinase and IkappaB kinase beta. These results emphasize a critical role for CYLD in preventing spontaneous activation of the Tak1 axis of T cell signaling and, thereby, maintaining normal T cell function.


Subject(s)
Cysteine Endopeptidases/metabolism , Lymphocyte Activation/immunology , MAP Kinase Kinase Kinases/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism , Adoptive Transfer , Animals , Deubiquitinating Enzyme CYLD , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Mice , Mice, Knockout , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Ubiquitination/immunology
13.
Immunology ; 119(1): 63-73, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16764686

ABSTRACT

CD8+ T cells play a crucial role in protective immunity to viruses and tumours. Antiviral CD8+ T cells are initially activated by professional antigen presenting cells (pAPCs) that are directly infected by viruses (direct-priming) or following uptake of exogenous antigen transferred from virus-infected or tumour cells (cross-priming). In order to efficiently target each of these antigen-processing pathways during vaccine design, it is necessary to delineate the properties of the natural substrates for either of these antigen-processing pathways. In this study, we utilized a novel T-cell receptor (TCR) transgenic mouse to examine the requirement for both antigen synthesis and synthesis of other cellular factors during direct or cross-priming. We found that direct presentation required ongoing synthesis of antigen, but that cross-priming favoured long-lived antigens and did not require ongoing antigen production. Even after prolonged blockade of protein synthesis in the donor cell, cross-priming was unaffected. In contrast, direct-presentation was almost undetectable in the absence of antigen neosynthesis and required ongoing protein synthesis. This suggests that the direct- and cross-priming pathways may utilize differing pools of antigen, an observation that has far-reaching implications for the rational design of vaccines aimed at the generation of protective CD8+ T cells.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/physiology , Virus Diseases/immunology , Adoptive Transfer/methods , Animals , Antigen-Presenting Cells/drug effects , Antigens/biosynthesis , Cell Line , Cycloheximide/pharmacology , Electroporation , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/prevention & control , Protein Synthesis Inhibitors/pharmacology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Virus Diseases/prevention & control
14.
Immunity ; 24(5): 503-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16713966

ABSTRACT

MHC class I binding peptides are generated via cytosolic degradation of a previously undefined substrate. In this issue of Immunity, pre-degradation polypeptide intermediates bound to a cytosolic chaperone is isolated.


Subject(s)
Histocompatibility Antigens Class I/immunology , Models, Immunological , Peptides/immunology , Signal Transduction/immunology , Animals , Antigen Presentation , Histocompatibility Antigens Class I/metabolism , Humans , Peptides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...