Network


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

Hotspot


Dive into the research topics where Jack D. Bui is active.

Publication


Featured researches published by Jack D. Bui.


Journal of Experimental Medicine | 2012

Cancer immunoediting by the innate immune system in the absence of adaptive immunity

Timothy E. O’Sullivan; Robert Saddawi-Konefka; William Vermi; Catherine M. Koebel; Cora D. Arthur; J. Michael White; Ravi Uppaluri; Daniel M. Andrews; Shin Foong Ngiow; Michele W. L. Teng; Mark J. Smyth; Robert D. Schreiber; Jack D. Bui

In the absence of adaptive immunity, NK cells polarize M1 macrophages to facilitate cancer immunoediting.


Immunologic Research | 2005

Interferon-γ and cancer immunoediting

Gavin P. Dunn; Hiroaki Ikeda; Allen T. Bruce; Catherine M. Koebel; Ravi Uppaluri; Jack D. Bui; Ruby Chan; Mark S. Diamond; J. Michael White; Kathleen C. F. Sheehan; Robert D. Schreiber

Over the last 12 yr, we have shown that interferony and lymphocytes collaborate to regulate tumor development in mice. Specifically, we found that the immune system not only prevents the growth of primary (carcinogen-induced and spontaneous) and transplanted tumors but also sculpts the immunogenicity of tumors that form. These observations led us to refine the old and controversial “cancer immuno-surveillance” hypothesis of Burnet and Thomas into one that we termed cancer immunoediting that better emphasizes the paradoxical host-protective and tumor-sculpting roles of immunity on developing tumors. Our current work focuses on defining the molecular mechanisms that underlie cancer immunoediting and exploring the implications of this process for cancer immunotherapy.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Production of unique immunotoxin cancer therapeutics in algal chloroplasts

Miller Tran; Christina Van; Daniel J. Barrera; Pär L. Pettersson; Carlos Peinado; Jack D. Bui; Stephen P. Mayfield

The idea of targeted therapy, whereby drug or protein molecules are delivered to specific cells, is a compelling approach to treating disease. Immunotoxins are one such targeted therapeutic, consisting of an antibody domain for binding target cells and molecules of a toxin that inhibits the proliferation of the targeted cell. One major hurdle preventing these therapies from reaching the market has been the lack of a suitable production platform that allows the cost-effective production of these highly complex molecules. The chloroplast of the green alga Chlamydomonas reinhardtii has been shown to contain the machinery necessary to fold and assemble complex eukaryotic proteins. However, the translational apparatus of chloroplasts resembles that of a prokaryote, allowing them to accumulate eukaryotic toxins that otherwise would kill a eukaryotic host. Here we show expression and accumulation of monomeric and dimeric immunotoxin proteins in algal chloroplasts. These fusion proteins contain an antibody domain targeting CD22, a B-cell surface epitope, and the enzymatic domain of exotoxin A from Pseudomonas aeruginosa. We demonstrated that algal-produced immunotoxins accumulate as soluble and enzymatically active proteins that bind target B cells and efficiently kill them in vitro. We also show that treatment with either the mono- or dimeric immunotoxins significantly prolongs the survival of mice with implanted human B-cell tumors.


Journal of Immunology | 2009

Cutting Edge: Down-Regulation of MHC Class I-Related Chain A on Tumor Cells by IFN-γ-Induced MicroRNA

Deepak Yadav; Jennifer Ngolab; Rod Seung-Hwan Lim; Siddharth Krishnamurthy; Jack D. Bui

NKG2D is a receptor used by NK cells to detect virally infected and transformed cells. It recognizes ligands that are expressed constitutively on primary tumors and tumor cell lines. In this report, we have identified four microRNAs (miRNAs) that each was sufficient to reduce the expression of the NKG2D ligand MHC class I-related chain A (MICA). One of these miRNAs (miR-520b) was induced by IFN-γ, leading to a reduction in MICA surface protein levels. Interestingly, miR-520b acted on both the MICA 3′-untranslated region and the promoter region and caused a decrease in the levels of MICA transcript. In contrast, an antisense oligonucleotide inhibitor of miR-520b increased the expression of a reporter construct containing the MICA 3′-untranslated region but not the MICA promoter region. These findings demonstrate the novel regulation of an NKG2D ligand by an endogenous microRNA that is itself induced by IFN-γ.


Journal of Immunology | 2006

IFN-Dependent Down-Regulation of the NKG2D Ligand H60 on Tumors

Jack D. Bui; Leonidas N. Carayannopoulos; Lewis L. Lanier; Wayne M. Yokoyama; Robert D. Schreiber

In this study, we show that IFN-γ or IFN-α reduce expression of H60 on 3′-methylcholanthrene (MCA) sarcomas from 129/Sv mice. As determined by flow cytometry using either NKG2D tetramers or NKG2D ligand-specific mAb, H60 was identified as the NKG2D ligand most frequently expressed on these sarcomas, and its expression was selectively down-regulated by either IFN-γ or IFN-α in a manner that was dose- and time-dependent and reversible. Down-regulation occurred at the transcript level and was STAT1-dependent. It also had functional consequences. IFN-γ-treated MCA sarcomas with high levels of H60 were resistant to killing by IL-2-activated NK cells. Resistance was not solely dependent on enhanced MHC class I expression but rather also required H60 down-regulation. IFN-γ-treated tumor cells also displayed diminished capacity to down-regulate NKG2D on freshly isolated NK cells. Transplanted tumor cells reisolated from immunocompetent mice displayed reduced H60 expression and increased MHC class I expression compared with tumor cells that were either left unmanipulated or reisolated from mice treated with neutralizing IFN-γ-specific mAb. This report thus represents the first demonstration that certain cytokines and specifically the IFNs regulate expression of specific NKG2D ligands on murine tumors. This process most likely helps to specify the type of immune effector cell populations that participate in host-protective antitumor responses.


Cancer Research | 2012

ERK1/2 regulation of CD44 modulates oral cancer aggressiveness

Nancy P. Judd; Ashley E. Winkler; Oihana Murillo-Sauca; Joshua J. Brotman; Jonathan H. Law; James S. Lewis; Gavin P. Dunn; Jack D. Bui; John B. Sunwoo; Ravindra Uppaluri

Carcinogen-induced oral cavity squamous cell carcinoma (OSCC) incurs significant morbidity and mortality and constitutes a global health challenge. To gain further insight into this disease, we generated cell line models from 7,12-dimethylbenz(a)anthracene-induced murine primary OSCC capable of tumor formation upon transplantation into immunocompetent wild-type mice. Whereas several cell lines grew rapidly and were capable of metastasis, some grew slowly and did not metastasize. Aggressively growing cell lines displayed ERK1/2 activation, which stimulated expression of CD44, a marker associated with epithelial to mesenchymal transition and putative cancer stem cells. MEK (MAP/ERK kinase) inhibition upstream of ERK1/2 decreased CD44 expression and promoter activity and reduced cell migration and invasion. Conversely, MEK1 activation enhanced CD44 expression and promoter activity, whereas CD44 attenuation reduced in vitro migration and in vivo tumor formation. Extending these findings to freshly resected human OSCC, we confirmed a strict relationship between ERK1/2 phosphorylation and CD44 expression. In summary, our findings identify CD44 as a critical target of ERK1/2 in promoting tumor aggressiveness and offer a preclinical proof-of-concept to target this pathway as a strategy to treat head and neck cancer.


Transplantation | 2008

Prolongation of cardiac and islet allograft survival by a blocking hamster anti-mouse CXCR3 monoclonal antibody.

Ravindra Uppaluri; Kathleen C. F. Sheehan; Liqing Wang; Jack D. Bui; Joshua J. Brotman; Bao Lu; Craig Gerard; Wayne W. Hancock; Robert D. Schreiber

Background. Acute allograft rejection requires a multifaceted immune response involving trafficking of immune cells into the transplant and expression of effector cell functions leading to graft destruction. The chemokine receptor CXCR3 and its ligands, CXCL9, CXCL10 and CXCL11, constitute an important pathway for effector cell recruitment posttransplant. However, analysis of CXCR3 expression and function has been hampered by a general lack of availability of a neutralizing anti-CXCR3 monoclonal antibody (mAb) for use in experimental models. Methods. We report the generation, characterization, and use of CXCR3-173, a new hamster mAb specific for mouse CXCR3 that recognizes CXCR3 on cells from wild-type but not CXCR3−/− mice. Results. Using CXCR3-173 mAb, we demonstrate CXCR3 expression on primary memory phenotype CD4+ and CD8+ T cells, naturally occurring CD4+CD25+ Foxp3+ regulatory T cells, natural killer T cells, and approximately 25% of NK cells. CXCR3-173 blocked chemotaxis in vitro in response to CXCL10 or CXCL11 but not CXCL9. When injected into mice, this mAb significantly prolonged both cardiac and islet allograft survival. When combined with a subtherapeutic regimen of rapamycin, CXCR3-173 mAb induced long-term (>100 day) survival of cardiac and islet allografts. The in vivo effects of CXCR3-173 mAb were not associated with effector lymphocyte depletion. Conclusion. These data highlight the utility of CXCR3-173 mAb in developing immunotherapeutic approaches to inhibit transplant rejection and potentially other immune-mediated diseases in murine models.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Modulation of natural killer cell antitumor activity by the aryl hydrocarbon receptor

June Ho Shin; Luhua Zhang; Oihana Murillo-Sauca; Junmo Kim; Holbrook Kohrt; Jack D. Bui; John B. Sunwoo

The aryl hydrocarbon receptor (AhR) has become increasingly recognized for its role in the differentiation and activity of immune cell subsets; however, its role in regulating the activity of natural killer (NK) cells has not been described. Here, we show that AhR expression is induced in murine NK cells upon cytokine stimulation. We show that in the absence of AhR, NK cells have reduced cytolytic activity and reduced capacity to control RMA-S tumor formation in vivo, despite having normal development and maturation markers. Although AhR was first identified to bind the xenobiotic compound dioxin, AhR is now known to bind a variety of natural exogenous (e.g., dietary) and endogenous ligands. We show that activation of AhR with an endogenous tryptophan derivative, 6-formylindolo[3,2-b]carbazole, potentiates NK cell IFN-γ production and cytolytic activity. Further, administration of 6-formylindolo[3,2-b]carbazole in vivo enhances NK cell control of tumors in an NK cell- and AhR-dependent manner. Finally, similar effects on NK cell potency occur with AhR dietary ligands, potentially explaining the numerous associations that have been observed in the past between diet and NK cell function. Our studies introduce AhR as another regulator of NK cell activity in vivo.


Journal of Immunology | 2010

The Host Defense Peptide Cathelicidin Is Required for NK Cell-Mediated Suppression of Tumor Growth

Amanda S. Büchau; Shin Morizane; Janet M. Trowbridge; Jürgen Schauber; Paul Kotol; Jack D. Bui; Richard L. Gallo

Tumor surveillance requires the interaction of multiple molecules and cells that participate in innate and the adaptive immunity. Cathelicidin was initially identified as an antimicrobial peptide, although it is now clear that it fulfills a variety of immune functions beyond microbial killing. Recent data have suggested contrasting roles for cathelicidin in tumor development. Because its role in tumor surveillance is not well understood, we investigated the requirement of cathelicidin in controlling transplantable tumors in mice. Cathelicidin was observed to be abundant in tumor-infiltrating NK1.1+ cells in mice. The importance of this finding was demonstrated by the fact that cathelicidin knockout mice (Camp −/−) permitted faster tumor growth than wild type controls in two different xenograft tumor mouse models (B16.F10 and RMA-S). Functional in vitro analyses found that NK cells derived from Camp −/− versus wild type mice showed impaired cytotoxic activity toward tumor targets. These findings could not be solely attributed to an observed perforin deficiency in freshly isolated Camp −/− NK cells, because this deficiency could be partially restored by IL-2 treatment, whereas cytotoxic activity was still defective in IL-2-activated Camp −/− NK cells. Thus, we demonstrate a previously unrecognized role of cathelicidin in NK cell antitumor function.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Engagement of myelomonocytic Siglecs by tumor-associated ligands modulates the innate immune response to cancer

Heinz Läubli; Oliver M. T. Pearce; Flavio Schwarz; Shoib S. Siddiqui; Lingquan Deng; Michal A. Stanczak; Liwen Deng; Andrea Verhagen; Patrick Secrest; Chrissy Lusk; Ann G. Schwartz; Nissi M. Varki; Jack D. Bui; Ajit Varki

Significance In vitro and in vivo data indicate that hypersialylated tumor cells can engage Siglec-9 on myelomonocytic cells and influence the outcome of the interaction, depending on the stage of tumor growth and the microenvironment. On one hand, engagement of Siglec-9 or Siglec-E by tumor-associated ligands inhibited immunosurveillance and tumor cell killing during establishment of autologous tumors and new metastatic foci. On the other hand, inhibition of tumor-associated macrophages through Siglec-9 led to M1 polarization and reduced growth-promoting inflammation within the tumor microenvironment. This demonstrates a previously unidentified dualistic function of Siglec-9 during cancer progression. A functional polymorphism of Siglec-9 correlated with altered survival of lung cancer patients, suggesting that Siglec-9 might be therapeutically targeted. Certain pathogenic bacteria are known to modulate the innate immune response by decorating themselves with sialic acids, which can engage the myelomonocytic lineage inhibitory receptor Siglec-9, thereby evading immunosurveillance. We hypothesized that the well-known up-regulation of sialoglycoconjugates by tumors might similarly modulate interactions with innate immune cells. Supporting this hypothesis, Siglec-9–expressing myelomonocytic cells found in human tumor samples were accompanied by a strong up-regulation of Siglec-9 ligands. Blockade of Siglec-9 enhanced neutrophil activity against tumor cells in vitro. To investigate the function of inhibitory myelomonocytic Siglecs in vivo we studied mouse Siglec-E, the murine functional equivalent of Siglec-9. Siglec-E–deficient mice showed increased in vivo killing of tumor cells, and this effect was reversed by transgenic Siglec-9 expression in myelomonocytic cells. Siglec-E–deficient mice also showed enhanced immunosurveillance of autologous tumors. However, once tumors were established, they grew faster in Siglec-E–deficient mice. In keeping with this, Siglec-E–deficient macrophages showed a propensity toward a tumor-promoting M2 polarization, indicating a secondary role of CD33-related Siglecs in limiting cancer-promoting inflammation and tumor growth. Thus, we define a previously unidentified impact of inhibitory myelomonocytic Siglecs in cancer biology, with distinct roles that reflect the dual function of myelomonocytic cells in cancer progression. In keeping with this, a human polymorphism that reduced Siglec-9 binding to carcinomas was associated with improved early survival in non–small-cell lung cancer patients, which suggests that Siglec-9 might be therapeutically targeted within the right time frame and stage of disease.

Collaboration


Dive into the Jack D. Bui's collaboration.

Top Co-Authors

Avatar

Robert D. Schreiber

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Emilie Gross

University of California

View shared research outputs
Top Co-Authors

Avatar

Ajit Varki

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlos Peinado

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ruth Seelige

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John T. Chang

University of California

View shared research outputs
Top Co-Authors

Avatar

Ravindra Uppaluri

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge