Network


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

Hotspot


Dive into the research topics where Daniel W. McVicar is active.

Publication


Featured researches published by Daniel W. McVicar.


Nature Genetics | 2007

Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1

Maureen P. Martin; Ying Qi; Xiaojiang Gao; Eriko Yamada; Jeffrey N. Martin; Florencia Pereyra; Sara Colombo; Elizabeth E. Brown; W. Lesley Shupert; John P. Phair; James J. Goedert; Susan Buchbinder; Gregory D. Kirk; Amalio Telenti; Mark Connors; Stephen J. O'Brien; Bruce D. Walker; Peter Parham; Steven G. Deeks; Daniel W. McVicar; Mary Carrington

Allotypes of the natural killer (NK) cell receptor KIR3DL1 vary in both NK cell expression patterns and inhibitory capacity upon binding to their ligands, HLA-B Bw4 molecules, present on target cells. Using a sample size of over 1,500 human immunodeficiency virus (HIV)+ individuals, we show that various distinct allelic combinations of the KIR3DL1 and HLA-B loci significantly and strongly influence both AIDS progression and plasma HIV RNA abundance in a consistent manner. These genetic data correlate very well with previously defined functional differences that distinguish KIR3DL1 allotypes. The various epistatic effects observed here for common, distinct KIR3DL1 and HLA-B Bw4 combinations are unprecedented with regard to any pair of genetic loci in human disease, and indicate that NK cells may have a critical role in the natural history of HIV infection.


Current Opinion in Immunology | 2009

TREM and TREM-like receptors in inflammation and disease ☆ ☆☆

Jill W. Ford; Daniel W. McVicar

Since the discovery of triggering receptor expressed on myeloid cells (TREM)-1 in 2000, evidence documenting the profound ability of the TREM and TREM-like receptors to regulate inflammation has rapidly accumulated. Monocytes, macrophages, myeloid dendritic cells, plasmacytoid dendritic cells, neutrophils, microglia, osteoclasts, and platelets all express at least one member of the TREM family, underscoring the importance of these proteins in the regulation of innate resistance. Recent work on the TREM family includes: characterization of a new receptor expressed on plasmacytoid dendritic cells; definition of a key role for TREM in inflammatory bowel disease and multiple sclerosis; an expanded list of diseases associated with the release of soluble forms of TREM proteins; and identification of the first well characterized TREM ligand: B7-H3, a ligand for TREM-like Transcript (TLT)-2. Moreover, analysis of TREM signaling has now identified key regulatory components and defined pathways that may be responsible for the complex functional interactions between the TREM and toll-like receptors. In addition, there is expanding evidence of a role for TREM in the regulation of integrin function via Plexin-A1. Together these new findings define the TREM and TREM-like receptors as pluripotent modifiers of disease through the integration of inflammatory signals with those associated with leukocyte adhesion.


Nature Genetics | 2009

HLA-C cell surface expression and control of HIV/AIDS correlate with a variant upstream of HLA-C

Rasmi Thomas; Richard Apps; Ying Qi; Xiaojiang Gao; Victoria Male; Colm O'hUigin; Geraldine M. O'Connor; Dongliang Ge; Jacques Fellay; Jeffrey N. Martin; Joseph B. Margolick; James J. Goedert; Susan Buchbinder; Gregory D. Kirk; Maureen P. Martin; Amalio Telenti; Steven G. Deeks; Bruce D. Walker; David B. Goldstein; Daniel W. McVicar; Ashley Moffett; Mary Carrington

A variant 35 kb upstream of the HLA-C gene (-35C/T) was previously shown to associate with HLA-C mRNA expression level and steady-state plasma HIV RNA levels. We genotyped this variant in 1,698 patients of European ancestry with HIV. Individuals with known seroconversion dates were used for disease progression analysis and those with longitudinal viral load data were used for viral load analysis. We further tested cell surface expression of HLA-C in normal donors using an HLA-C-specific antibody. We show that the -35C allele is a proxy for high HLA-C cell surface expression, and that individuals with high-expressing HLA-C alleles progress more slowly to AIDS and control viremia significantly better than individuals with low HLA-C expressing alleles. These data strongly implicate high HLA-C expression levels in more effective control of HIV-1, potentially through better antigen presentation to cytotoxic T lymphocytes or recognition and killing of infected cells by natural killer cells.


Journal of Immunology | 2007

Cutting Edge: Allele-Specific and Peptide-Dependent Interactions between KIR3DL1 and HLA-A and HLA-B

Hathairat Thananchai; Geraldine Gillespie; Maureen P. Martin; Arman Bashirova; Nobuyo Yawata; Makoto Yawata; Philippa Easterbrook; Daniel W. McVicar; Katsumi Maenaka; Peter Parham; Mary Carrington; Tao Dong; Sarah Rowland-Jones

Although it is clear that KIR3DL1 recognizes Bw4+ HLA-B, the role of Bw4+ HLA-A allotypes as KIR3DL1 ligands is controversial. We therefore examined the binding of tetrameric HLA-A and –B complexes, including HLA*2402, a common Bw4+ HLA-A allotype, to KIR3DL1*001, *005, *007, and *1502 allotypes. Only Bw4+ tetramers bound KIR3DL1. Three of four HLA-A*2402 tetramers bound one or more KIR3DL1 allotypes and all four KIR3DL1 allotypes bound to one or more HLA-A*2402 tetramers, but with different binding specificities. Only KIR3DL1*005 bound both HLA-A*2402 and HLA-B*5703 tetramers. HLA-A*2402-expressing target cells were resistant to lysis by NK cells expressing KIR3DL1*001 or *005. This study shows that HLA-A*2402 is a ligand for KIR3DL1 and demonstrates how the binding of KIR3DL1 to Bw4+ ligands depends upon the bound peptide as well as HLA and KIR3DL1 polymorphism.


Nature Immunology | 2001

Mutation of Tec family kinases alters T helper cell differentiation.

Edward M. Schaeffer; George S. Yap; Carol Lewis; Michael J. Czar; Daniel W. McVicar; Allen W. Cheever; Alan Sher; Pamela L. Schwartzberg

The Tec kinases Rlk and Itk are critical for full T cell receptor (TCR)-induced activation of phospholipase C-γ and mitogen-activated protein kinase. We show here that the mutation of Rlk and Itk impaired activation of the transcription factors NFAT and AP-1 and production of both T helper type 1 (TH1) and TH2 cytokines. Consistent with these biochemical defects, Itk−/− mice did not generate effective TH2 responses when challenged with Schistosoma mansoni eggs. Paradoxically, the more severely impaired Rlk−/−Itk−/− mice were able to mount a TH2 response and produced TH2 cytokines in response to this challenge. In addition, Rlk−/−Itk−/− cells showed impaired TCR-induced repression of the TH2-inducing transcription factor GATA-3, suggesting a potential mechanism for TH2 development in these hyporesponsive cells. Thus, mutations that affect Tec kinases lead to complex alterations in CD4+ TH cell differentiation.


Nature | 2011

Killer cell immunoglobulin-like receptor 3DL1-mediated recognition of human leukocyte antigen B

Julian P. Vivian; Renee C Duncan; Richard M. Berry; Geraldine M. O'Connor; Hugh H. Reid; Travis Beddoe; Stephanie Gras; Philippa M. Saunders; Maya A Olshina; Jacqueline M. L. Widjaja; Christopher M. Harpur; Jie Lin; Sebastien Maloveste; David A. Price; Bernard A. P. Lafont; Daniel W. McVicar; Craig S. Clements; Andrew G. Brooks; Jamie Rossjohn

Members of the killer cell immunoglobulin-like receptor (KIR) family, a large group of polymorphic receptors expressed on natural killer (NK) cells, recognize particular peptide-laden human leukocyte antigen (pHLA) class I molecules and have a pivotal role in innate immune responses. Allelic variation and extensive polymorphism within the three-domain KIR family (KIR3D, domains D0–D1–D2) affects pHLA binding specificity and is linked to the control of viral replication and the treatment outcome of certain haematological malignancies. Here we describe the structure of a human KIR3DL1 receptor bound to HLA-B*5701 complexed with a self-peptide. KIR3DL1 clamped around the carboxy-terminal end of the HLA-B*5701 antigen-binding cleft, resulting in two discontinuous footprints on the pHLA. First, the D0 domain, a distinguishing feature of the KIR3D family, extended towards β2-microglobulin and abutted a region of the HLA molecule with limited polymorphism, thereby acting as an ‘innate HLA sensor’ domain. Second, whereas the D2–HLA-B*5701 interface exhibited a high degree of complementarity, the D1–pHLA-B*5701 contacts were suboptimal and accommodated a degree of sequence variation both within the peptide and the polymorphic region of the HLA molecule. Although the two-domain KIR (KIR2D) and KIR3DL1 docked similarly onto HLA-C and HLA-B respectively, the corresponding D1-mediated interactions differed markedly, thereby providing insight into the specificity of KIR3DL1 for discrete HLA-A and HLA-B allotypes. Collectively, in association with extensive mutagenesis studies at the KIR3DL1–pHLA-B*5701 interface, we provide a framework for understanding the intricate interplay between peptide variability, KIR3D and HLA polymorphism in determining the specificity requirements of this essential innate interaction that is conserved across primate species.


Immunological Reviews | 2001

The ever-expanding Ly49 gene family: repertoire and signaling.

Stephen K. Anderson; John R. Ortaldo; Daniel W. McVicar

Summary: The mouse lectin‐related Ly49 family and the human killer cell Ig‐like receptor (KIR) family represent structurally distinct, yet functionally analogous, class I MHC receptors that are expressed on natural killer cells and some T cells. The functional similarity of these two families has been borne out by the demonstration of identical signal transduction pathways associated with each receptor family. The Ly49 family therefore provides a useful model system to study the role of this class of receptors in the regulation of the immune system. Recent data relating to the Ly49 repertoire in several mouse strains has revealed an additional evolutionary parallel between KIR and Ly49 receptor families. There is now an appreciation of the variation in the number and type of Ly49s expressed in different mouse strains, similar to the previously demonstrated differences in the number of KIR genes found in humans. This review summarizes the current members of the Ly49 gene family, their MHC class I recognition and associated signal transduction pathways.


Nature | 2016

NAFLD causes selective CD4 + T lymphocyte loss and promotes hepatocarcinogenesis

Chi Ma; Aparna H. Kesarwala; Tobias Eggert; José Medina-Echeverz; David E. Kleiner; Ping Jin; David F. Stroncek; Masaki Terabe; Veena Kapoor; Mei ElGindi; Miaojun Han; Angela M. Thornton; Haibo Zhang; Michèle Egger; Ji Luo; Dean W. Felsher; Daniel W. McVicar; Achim Weber; Mathias Heikenwalder; Tim F. Greten

Hepatocellular carcinoma (HCC) is the second most common cause of cancer-related death. Non-alcoholic fatty liver disease (NAFLD) affects a large proportion of the US population and is considered to be a metabolic predisposition to liver cancer. However, the role of adaptive immune responses in NAFLD-promoted HCC is largely unknown. Here we show, in mouse models and human samples, that dysregulation of lipid metabolism in NAFLD causes a selective loss of intrahepatic CD4+ but not CD8+ T lymphocytes, leading to accelerated hepatocarcinogenesis. We also demonstrate that CD4+ T lymphocytes have greater mitochondrial mass than CD8+ T lymphocytes and generate higher levels of mitochondrially derived reactive oxygen species (ROS). Disruption of mitochondrial function by linoleic acid, a fatty acid accumulated in NAFLD, causes more oxidative damage than other free fatty acids such as palmitic acid, and mediates selective loss of intrahepatic CD4+ T lymphocytes. In vivo blockade of ROS reversed NAFLD-induced hepatic CD4+ T lymphocyte decrease and delayed NAFLD-promoted HCC. Our results provide an unexpected link between lipid dysregulation and impaired anti-tumour surveillance.


PLOS ONE | 2009

Visualization and Identification of IL-7 Producing Cells in Reporter Mice

Renata Mazzucchelli; Søren Warming; Scott M. Lawrence; Masaru Ishii; Mehrnoosh Abshari; A. Vallance Washington; Lionel Feigenbaum; Andrew C. Warner; Davis J. Sims; Wenqing Li; Julie A. Hixon; Daniel Gray; Benjamin E. Rich; Matthew P. Morrow; Miriam R. Anver; James Cherry; Dieter Naf; Lawrence R. Sternberg; Daniel W. McVicar; Andrew G. Farr; Ronald N. Germain; Keith Rogers; Nancy A. Jenkins; Neal G. Copeland; Scott K. Durum

Interleukin-7 (IL-7) is required for lymphocyte development and homeostasis although the actual sites of IL-7 production have never been clearly identified. We produced a bacterial artificial chromosome (BAC) transgenic mouse expressing ECFP in the Il7 locus. The construct lacked a signal peptide and ECFP (enhanced cyan fluorescent protein ) accumulated inside IL-7-producing stromal cells in thoracic thymus, cervical thymus and bone marrow. In thymus, an extensive reticular network of IL-7-containing processes extended from cortical and medullary epithelial cells, closely contacting thymocytes. Central memory CD8 T cells, which require IL-7 and home to bone marrow, physically associated with IL-7-producing cells as we demonstrate by intravital imaging.


Science Signaling | 2001

Intracellular Signaling by the Killer Immunoglobulin-Like Receptors and Ly49

Daniel W. McVicar; Deborah N. Burshtyn

Once thought to be promiscuous killers, it is now known that natural killer (NK) cells possess an elaborate array of receptors that regulate NK cytotoxic and secretory functions upon interaction with target cell MHC class I proteins. These receptors, known as killer cell immunoglobulin-like receptors (KIRs) in humans, and Ly49 receptors in the mouse, have become the focus of intense study in an effort to discern the underlying biology of these large receptor families. These receptor families include both inhibitory and activating receptors. Interrogation of a target expressing KIR ligands leads to coengagement of the inhibitory receptor with as-yet poorly defined activation receptors. Kinases activated during engagement mediate the phosphorylation of the KIR or Ly49 cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIMs). The phosphorylated ITIMs serve as efficient recruitment points for the cytosolic protein tyrosine phosphatases, SHP-1 and SHP-2, resulting in the dephosphorylation of substrates critical for cellular activation. In contrast, some KIRs and Ly49s lack the ITIM and possess a charged residue in their transmembrane domains that mediates interaction with the DAP12 signal transduction chain. DAP12 uses its cytoplasmic immunoreceptor tyrosine-based activation motif (ITAM) to mediate cellular activation. Engagement of a DAP12 coupled KIR or Ly49 results in phosphorylation of DAP12, and other key substrates, including the Syk tryosine kinase, phospholipase C, and c-Cbl. DAP12 activation then leads to the Mapk cascade and ultimately to enhanced degranulation, and production of cytokines and chemokines. Although the context in which inhibitory and activating KIR and Ly49s function is not yet known, the dissection of the activating and inhibitory signal transduction pathways should shed light on their method of integration into the activation sequela of NK cells. Ultimately, this work will lead to concrete understanding of the immunobiology of these seemingly antagonistic receptor systems. Natural killer (NK) cells patrol the blood and immune organs in search of infected or malignant cells. Research suggests that these cells query a potential target cell for surface proteins that identify it as a healthy, self-derived cell. If the NK cell fails to recognize self markers because they have been down-regulated due to malignant transformation or viral infection, the NK cell kills the target. Scientists now have a clearer understanding of how NK cells discriminate between healthy and diseased cells. NK surface proteins called killer cell immunoglobulin-like receptors (KIRs) in humans, and Ly49 receptors in the mouse, recognize the markers of self. Once engaged, specific intracellular regions of the receptors are modified to recruit enzymes capable of reversing the biochemical events within the NK cell that would otherwise lead it to kill the target. Surprisingly, some KIRs and Ly49 receptors recognize the same self markers but send activation signals to NK cells. It is unclear why these cells would want a system of receptors that both activate and inhibit, thus potentially canceling one another out, but the method of this madness is clear. Activating KIRs or Ly49s lack inhibitory characteristics and instead interact with a second protein called DAP12, which contains an immunoreceptor tyrosine-based activation motif that functions to recruit activating enzymes. The result is a biochemical system of Yin and Yang where some receptors shut down responses and others activate them. Through tight regulation of the expression of these receptors, the NK cell is careful to seek out and kill only those cells identified as infected, damaged, or malignant.

Collaboration


Dive into the Daniel W. McVicar's collaboration.

Top Co-Authors

Avatar

Laura Quigley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John R. Ortaldo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Geraldine M. O'Connor

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John J. O'Shea

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lisa A. Ridnour

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Maureen P. Martin

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

David A. Wink

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge