Network


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

Hotspot


Dive into the research topics where Angela L. Zarling is active.

Publication


Featured researches published by Angela L. Zarling.


Journal of Immunology | 2003

Tapasin Is a Facilitator, Not an Editor, of Class I MHC Peptide Binding

Angela L. Zarling; Chance John Luckey; Jarrod A. Marto; Forest M. White; Cynthia J. Brame; Anne M. Evans; Paul J. Lehner; Peter Cresswell; Jeffrey Shabanowitz; Donald F. Hunt; Victor H. Engelhard

Tapasin has been proposed to function as a peptide editor to displace lower affinity peptides and/or to favor the binding of high affinity peptides. Consistent with this, cell surface HLA-B8 molecules in tapasin-deficient cells were less stable and the peptide repertoire was substantially altered. However, the binding affinities of peptides expressed in the absence of tapasin were unexpectedly higher, not lower. The peptide repertoire from cells expressing soluble tapasin was similar in both appearance and affinity to that presented in the presence of full-length tapasin, but the HLA-B8 molecules showed altered cell surface stability characteristics. Similarly, the binding affinities of HLA-A*0201-associated peptides from tapasin+ and tapasin− cells were equivalent, although steady state HLA-A*0201 cell surface expression was decreased and the molecules demonstrated reduced cell surface stability on tapasin− cells. These data are inconsistent with a role for tapasin as a peptide editor. Instead, we propose that tapasin acts as a peptide facilitator. In this role, it stabilizes the peptide-free conformation of class I MHC molecules in the endoplasmic reticulum and thus increases the number and variety of peptides bound to class I MHC. Full-length tapasin then confers additional stability on class I MHC molecules that are already associated with peptides.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Identification of class I MHC-associated phosphopeptides as targets for cancer immunotherapy.

Angela L. Zarling; Joy M. Polefrone; Anne M. Evans; Leann M. Mikesh; Jeffrey Shabanowitz; Sarah Lewis; Victor H. Engelhard; Donald F. Hunt

Alterations in phosphorylation of cellular proteins are a hallmark of malignant transformation. Degradation of these phosphoproteins could generate cancer-specific class I MHC-associated phosphopeptides recognizable by CD8+ T lymphocytes. In a comparative analysis of phosphopeptides presented on the surface of melanoma, ovarian carcinoma, and B lymphoblastoid cells, we find 5 of 36 that are restricted to the solid tumors and common to both cancers. Differential presentation of these peptides can result from differential phosphorylation of the source proteins. Recognition of the peptides on cancer cells by phosphopeptide-specific CD8+ T lymphocytes validates the potential of these phosphopeptides as immunotherapeutic targets.


Molecular Immunology | 2002

Insights into antigen processing gained by direct analysis of the naturally processed class I MHC associated peptide repertoire

Victor H. Engelhard; Anthony G. Brickner; Angela L. Zarling

MHC class I molecules are responsible for the presentation of antigenic peptides to CD8+ T lymphocytes. Based on their relatively promiscuous binding of peptides, these molecules display information derived from a large fraction of proteins that are made inside the cell. This review describes our characterization of the peptides comprising this repertoire, with particular attention given to their complexity and quantities, their post-translational modification, and the pathways leading to their expression.


Science Translational Medicine | 2013

MHC class-I associated phosphopeptides are the targets of memory-like immunity in leukemia

Mark Cobbold; Hugo De La Peña; Andrew Norris; Joy M. Polefrone; Jie Qian; Ann Michelle English; Kara L. Cummings; Sarah Penny; James E. Turner; Jennifer Cottine; Jennifer G. Abelin; Stacy A. Malaker; Angela L. Zarling; Hsing-Wen Huang; Oliver Goodyear; Sylvie Freeman; Jeffrey Shabanowitz; Guy Pratt; Charles Craddock; Michael E. Williams; Donald F. Hunt; Victor H. Engelhard

Immunity against phosphopeptide antigens lacking in leukemia patients can be restored with stem cell transplantation. Adding to the Toolkit for Cancer Therapy The immune system is increasingly being used as a tool for cancer therapy. Researchers have harnessed the body’s own defense system to specifically target tumors. However, one limitation of immune-targeting strategies is the relative lack of targets. Because cancer cells are derived from normal human tissue, ideal antigens would be specifically or differentially expressed by tumor cells and healthy tissues. Now, Cobbold et al. find that phosphoproteins may broaden the pool of tumor antigens that can be targeted with immunotherapy. One difference between cancer and normal cells is the way in which they are regulated. Indeed, signal transduction pathways are frequently dysregulated in cancer cells. The authors now use a hallmark of signal transduction—protein phosphorylation—to identify and characterize new phosphoantigens that stimulate immune cells. They identified 95 phosphopeptides presented on the surface of leukemic cells and demonstrated that they could be recognized and killed by phosphopeptide-specific cytotoxic T lymphocytes. Somewhat surprisingly, healthy individuals had high levels of responses to phosphopeptides, but these responses were muted in leukemia patients with poor prognosis. What’s more, allogeneic stem cell transplant could restore phosphoprotein immune response in patients. These data suggest that phosphopeptides could be developed as new targets for cancer immunotherapy. Deregulation of signaling pathways is a hallmark of malignant transformation. Signaling-associated phosphoproteins can be degraded to generate cancer-specific phosphopeptides that are presented by major histocompatibility complex (MHC) class I and II molecules and recognized by T cells; however, the contribution of these phosphoprotein-specific T cells to immune surveillance is unclear. We identified 95 phosphopeptides presented on the surface of primary hematological tumors and normal tissues, including 61 that were tumor-specific. Phosphopeptides were more prevalent on more aggressive and malignant samples. CD8+ T cell lines specific for these phosphopeptides recognized and killed both leukemia cell lines and human leukocyte antigen–matched primary leukemia cells ex vivo. Notably, healthy individuals showed robust CD8+ T cell responses against many of these phosphopeptides within the circulating memory compartment. This immunity was significantly reduced or absent in some leukemia patients. This reduction correlated with clinical outcome; however, immunity was restored after allogeneic stem cell transplantation. These results suggest that phosphopeptides may be targets of cancer immune surveillance in humans, and point to their importance for development of vaccine-based and T cell adoptive transfer immunotherapies.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Identification of tumor-associated, MHC class II-restricted phosphopeptides as targets for immunotherapy

Florence R. Depontieu; Jie Qian; Angela L. Zarling; Tracee L. McMiller; Theresa M. Salay; Andrew Norris; A. Michelle English; Jeffrey Shabanowitz; Victor H. Engelhard; Donald F. Hunt; Suzanne L. Topalian

The activation and recruitment of CD4+ T cells are critical for the development of efficient antitumor immunity and may allow for the optimization of current cancer immunotherapy strategies. Searching for more optimal and selective targets for CD4+ T cells, we have investigated phosphopeptides, a new category of tumor-derived epitopes linked to proteins with vital cellular functions. Although MHC I-restricted phosphopeptides have been identified, it was previously unknown whether human MHC II molecules present phosphopeptides for specific CD4+ T cell recognition. We first demonstrated the fine specificity of human CD4+ T cells to discriminate a phosphoresidue by using cells raised against the candidate melanoma antigen mutant B-Raf or its phosphorylated counterpart. Then, we assessed the presence and complexity of human MHC II-associated phosphopeptides by analyzing 2 autologous pairs of melanoma and EBV-transformed B lymphoblastoid lines. By using sequential affinity isolation, biochemical enrichment, mass spectrometric sequencing, and comparative analysis, a total of 175 HLA-DR-associated phosphopeptides were characterized. Many were derived from source proteins that may have roles in cancer development, growth, and metastasis. Most were expressed exclusively by either melanomas or transformed B cells, suggesting the potential to define cell type-specific phosphatome “fingerprints.” We then generated HLA-DRβ1*0101-restricted CD4+ T cells specific for a phospho-MART-1 peptide identified in both melanoma cell lines. These T cells showed specificity for phosphopeptide-pulsed antigen-presenting cells as well as for intact melanoma cells. This previously undescribed demonstration of MHC II-restricted phosphopeptides recognizable by human CD4+ T cells provides potential new targets for cancer immunotherapy.


Nature Immunology | 2008

Phosphorylation-dependent interaction between antigenic peptides and MHC class I: a molecular basis for the presentation of transformed self

Fiyaz Mohammed; Mark Cobbold; Angela L. Zarling; Mahboob Salim; Gregory A. Barrett-Wilt; Jeffrey Shabanowitz; Donald F. Hunt; Victor H. Engelhard; Benjamin E. Willcox

Protein phosphorylation generates a source of phosphopeptides that are presented by major histocompatibility complex class I molecules and recognized by T cells. As deregulated phosphorylation is a hallmark of malignant transformation, the differential display of phosphopeptides on cancer cells provides an immunological signature of transformed self. Here we demonstrate that phosphorylation can considerably increase peptide binding affinity for HLA-A2. To understand this, we solved crystal structures of four phosphopeptide–HLA-A2 complexes. These identified a novel peptide-binding motif centered on a solvent-exposed phosphate anchor. Our findings indicate that deregulated phosphorylation can create neoantigens by promoting binding to major histocompatibility complex molecules or by affecting the antigenic identity of presented epitopes. These results highlight the potential of phosphopeptides as novel targets for cancer immunotherapy.


Immunological Reviews | 2008

Strategies and challenges in eliciting immunity to melanoma

Andrew R. Ferguson; Lisa A. Nichols; Angela L. Zarling; Elizabeth Thompson; C. Colin Brinkman; Kristian M. Hargadon; Timothy Bullock; Victor H. Engelhard

Summary: The ability of CD8+ T cells to recognize melanoma tumors has led to the development of immunotherapeutic approaches that use the antigens CD8+ T cells recognize. However, clinical response rates have been disappointing. Here we summarize our work to understand the mechanisms of self‐tolerance that limit responses to currently utilized antigens and our approach to identify new antigens directly tied to malignancy. We also explore several aspects of the anti‐tumor immune response induced by peptide‐pulsed dendritic cells (DCs). DCs differentially augment the avidity of recall T cells specific for self‐antigens and overcome a process of aberrant CD8+ T‐cell differentiation that occurs in tumor‐draining lymph nodes. DC migration is constrained by injection route, resulting in immune responses in localized lymphoid tissue, and differential control of tumors depending on their location in the body. We demonstrate that CD8+ T‐cell differentiation in different lymphoid compartments alters the expression of homing receptor molecules and leads to the presence of systemic central memory cells. Our studies highlight several issues that must be addressed to improve the efficacy of tumor immunotherapy.


Cancer Research | 2014

MHC-Restricted Phosphopeptides from Insulin Receptor Substrate-2 and CDC25b Offer Broad-Based Immunotherapeutic Agents for Cancer

Angela L. Zarling; Rebecca C. Obeng; A. Nicole Desch; Joel Pinczewski; Kara L. Cummings; Donna H. Deacon; Mark R. Conaway; Craig L. Slingluff; Victor H. Engelhard

Cancer cells display novel phosphopeptides in association with MHC class I and II molecules. In this study, we evaluated two HLA-A2-restricted phosphopeptides derived from the insulin receptor substrate (IRS)-2 and the cell-cycle regulator CDC25b. These proteins are both broadly expressed in multiple malignancies and linked to cancer cell survival. Two phosphopeptides, termed pIRS-21097-1105 and pCDC25b38-46, served as targets of strong and specific CD8 T-cell memory responses in normal human donors. We cloned T-cell receptor (TCR) cDNAs from murine CD8 T-cell lines specific for either pIRS-21097-1105 or pCDC25b38-46. Expression of these TCRs in human CD8 T cells imparted high-avidity phosphopeptide-specific recognition and cytotoxic and cytokine-secreting effector activities. Using these cells, we found that endogenously processed pIRS-21097-1105 was presented on HLA-A2(+) melanomas and breast, ovarian, and colorectal carcinomas. Presentation was correlated with the level of the Ser(1100)-phosphorylated IRS-2 protein in metastatic melanoma tissues. The highest expression of this protein was evident on dividing malignant cells. Presentation of endogenously processed pCDC25b38-46 was narrower, but still evident on HLA-A2(+) melanoma, breast carcinoma, and lymphoblastoid cells. Notably, pIRS-21097-1105-specific and pCDC25b38-46-specific TCR-expressing human CD8 T cells markedly slowed tumor outgrowth in vivo. Our results define two new antigens that may be developed as immunotherapeutic agents for a broad range of HLA-A2(+) cancers.


Journal of Proteome Research | 2017

Identification and Characterization of Complex Glycosylated Peptides Presented by the MHC Class II Processing Pathway in Melanoma

Stacy A. Malaker; Michael J. Ferracane; Florence R. Depontieu; Angela L. Zarling; Jeffrey Shabanowitz; Dina L. Bai; Suzanne L. Topalian; Victor H. Engelhard; Donald F. Hunt

The MHC class II (MHCII) processing pathway presents peptides derived from exogenous or membrane-bound proteins to CD4+ T cells. Several studies have shown that glycopeptides are necessary to modulate CD4+ T cell recognition, though glycopeptide structures in these cases are generally unknown. Here, we present a total of 93 glycopeptides from three melanoma cell lines and one matched EBV-transformed line with most found only in the melanoma cell lines. The glycosylation we detected was diverse and comprised 17 different glycoforms. We then used molecular modeling to demonstrate that complex glycopeptides are capable of binding the MHC and may interact with complementarity determining regions. Finally, we present the first evidence of disulfide-bonded peptides presented by MHCII. This is the first large scale study to sequence glyco- and disulfide bonded MHCII peptides from the surface of cancer cells and could represent a novel avenue of tumor activation and/or immunoevasion.


Oncotarget | 2017

The antigenic identity of human class I MHC phosphopeptides is critically dependent upon phosphorylation status.

Fiyaz Mohammed; Daniel H. Stones; Angela L. Zarling; Carrie R. Willcox; Jeffrey Shabanowitz; Kara L. Cummings; Donald F. Hunt; Mark Cobbold; Victor H. Engelhard; Benjamin E. Willcox

Dysregulated post-translational modification provides a source of altered self-antigens that can stimulate immune responses in autoimmunity, inflammation, and cancer. In recent years, phosphorylated peptides have emerged as a group of tumour-associated antigens presented by MHC molecules and recognised by T cells, and represent promising candidates for cancer immunotherapy. However, the impact of phosphorylation on the antigenic identity of phosphopeptide epitopes is unclear. Here we examined this by determining structures of MHC-bound phosphopeptides bearing canonical position 4-phosphorylations in the presence and absence of their phosphate moiety, and examining phosphopeptide recognition by the T cell receptor (TCR). Strikingly, two peptides exhibited major conformational changes upon phosphorylation, involving a similar molecular mechanism, which focussed changes on the central peptide region most critical for T cell recognition. In contrast, a third epitope displayed little conformational alteration upon phosphorylation. In addition, binding studies demonstrated TCR interaction with an MHC-bound phosphopeptide was both epitope-specific and absolutely dependent upon phosphorylation status. These results highlight the critical influence of phosphorylation on the antigenic identity of naturally processed class I MHC epitopes. In doing so they provide a molecular framework for understanding phosphopeptide-specific immune responses, and have implications for the development of phosphopeptide antigen-specific cancer immunotherapy approaches.Dysregulated post-translational modification provides a source of altered self-antigens that can stimulate immune responses in autoimmunity, inflammation, and cancer. In recent years, phosphorylated peptides have emerged as a group of tumour-associated antigens presented by MHC molecules and recognised by T cells, and represent promising candidates for cancer immunotherapy. However, the impact of phosphorylation on the antigenic identity of phosphopeptide epitopes is unclear. Here we examined this by determining structures of MHC-bound phosphopeptides bearing canonical position 4-phosphorylations in the presence and absence of their phosphate moiety, and examining phosphopeptide recognition by the T cell receptor (TCR). Strikingly, two peptides exhibited major conformational changes upon phosphorylation, involving a similar molecular mechanism, which focussed changes on the central peptide region most critical for T cell recognition. In contrast, a third epitope displayed little conformational alteration upon phosphorylation. In addition, binding studies demonstrated TCR interaction with an MHC-bound phosphopeptide was both epitope-specific and absolutely dependent upon phosphorylation status. These results highlight the critical influence of phosphorylation on the antigenic identity of naturally processed class I MHC epitopes. In doing so they provide a molecular framework for understanding phosphopeptide-specific immune responses, and have implications for the development of phosphopeptide antigen-specific cancer immunotherapy approaches.

Collaboration


Dive into the Angela L. Zarling's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark Cobbold

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jie Qian

University of Virginia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fiyaz Mohammed

University of Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge