Network


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

Hotspot


Dive into the research topics where Julie S. Nielsen is active.

Publication


Featured researches published by Julie S. Nielsen.


Journal of Cell Science | 2008

Novel functions of the CD34 family

Julie S. Nielsen; Kelly M. McNagny

For almost 30 years, the cell-surface protein CD34 has been widely used as a marker to assist in the identification and isolation of hematopoietic stem cells (HSCs) and progenitors in preparation for bone-marrow transplantation. In addition, it has increasingly been used as a marker to help identify other tissue-specific stem cells, including muscle satellite cells and epidermal precursors. Despite its utility as a stem-cell marker, however, the function of CD34 has remained remarkably elusive. This is probably because: (1) it is subject to a range of tissue-specific post-transcriptional and post-translational modifications that are expected to alter its function dramatically; (2) the simple interpretation of CD34 gain- and loss-of-function experiments has been confounded by the overlapping expression of the two recently discovered CD34-related proteins podocalyxin and endoglycan; and (3) there has been a glaring lack of robust in vitro and in vivo functional assays that permit the structural and functional analysis of CD34 and its relatives. Here, we provide a brief review of the domain structure, genomic organization, and tissue distribution of the CD34 family. We also describe recent insights from gain- and loss-of-function experiments and improved assays, which are elucidating a fascinating role for these molecules in cell morphogenesis and migration.


Clinical Cancer Research | 2012

CD20+ tumor-infiltrating lymphocytes have an atypical CD27- memory phenotype and together with CD8+ T cells promote favorable prognosis in ovarian cancer.

Julie S. Nielsen; Rob A. Sahota; Katy Milne; Sara E.F. Kost; Nancy J. Nesslinger; Peter H. Watson; Brad H. Nelson

Purpose: Tumor-infiltrating lymphocytes (TIL), in particular CD8+ T cells and CD20+ B cells, are strongly associated with survival in ovarian cancer and other carcinomas. Although CD8+ TIL can mediate direct cytolytic activity against tumors, the role of CD20+ TIL is poorly understood. Here, we investigate the possible contributions of CD20+ TIL to humoral and cellular tumor immunity. Experimental Design: Tumor and serum specimens were obtained from patients with high-grade serous ovarian cancer. CD8+ and CD20+ TIL were analyzed by immunohistochemistry and flow cytometry. Immunoglobulin molecules were evaluated by DNA sequencing. Serum autoantibody responses to the tumor antigens p53 and NY-ESO-1 were measured by ELISA. Results: The vast majority of CD20+ TIL were antigen experienced, as evidenced by class-switching, somatic hypermutation, and oligoclonality, yet they failed to express the canonical memory marker CD27. CD20+ TIL showed no correlation with serum autoantibodies to p53 or NY-ESO-1. Instead, they colocalized with activated CD8+ TIL and expressed markers of antigen presentation, including MHC class I, MHC class II, CD40, CD80, and CD86. The presence of both CD20+ and CD8+ TIL correlated with increased patient survival compared with CD8+ TIL alone. Conclusions: In high-grade serous ovarian tumors, CD20+ TIL have an antigen–experienced but atypical CD27− memory B-cell phenotype. They are uncoupled from serum autoantibodies, express markers of antigen-presenting cells, and colocalize with CD8+ T cells. We propose that the association between CD20+ TIL and patient survival may reflect a supportive role in cytolytic immune responses. Clin Cancer Res; 18(12); 3281–92. ©2012 AACR.


Cancer Research | 2004

Overexpression of the Anti-Adhesin Podocalyxin Is an Independent Predictor of Breast Cancer Progression

Aruna Somasiri; Julie S. Nielsen; Nikita Makretsov; Marcia L. McCoy; Leah M Prentice; C. Blake Gilks; Stephen Chia; Karen A. Gelmon; David B. Kershaw; David Huntsman; Kelly M. McNagny; Calvin D. Roskelley

Podocalyxin is a CD34-related cell surface molecule with anti-adhesive qualities. We probed a tissue microarray (n = 272) linked to long-term outcome data and found that podocalyxin was highly overexpressed in a distinct subset of invasive breast carcinomas (n = 15; 6%). Univariate disease-specific (P < 0.01) and multivariate regression (P < 0.0005) analyses indicated that this overexpression is an independent indicator of poor outcome. Forced podocalyxin expression perturbed cell junctions between MCF-7 breast carcinoma cells, and it caused cell shedding from confluent monolayers. Therefore, podocalyxin overexpression is a novel predictor of breast cancer progression that may contribute to the process by perturbing tumor cell adhesion.


Journal of The American Society of Nephrology | 2009

The Role of Podocalyxin in Health and Disease

Julie S. Nielsen; Kelly M. McNagny

Podocalyxin, a sialomucin most closely related to CD34 and endoglycan, is expressed by kidney podocytes, hematopoietic progenitors, vascular endothelia, and a subset of neurons; aberrant expression has recently been implicated in a range of cancers. Through interactions with several intracellular proteins and at least one extracellular ligand, podocalyxin regulates both adhesion and cell morphology. In the developing kidney, podocalyxin plays an essential role in the formation and maintenance of podocyte foot processes, and its absence results in perinatal lethality. Podocalyxin expression in the hematopoietic system correlates with cell migration and the seeding of new hematopoietic tissues. In addition, it is abnormally expressed in subsets of breast, prostate, liver, pancreatic, and kidney cancer as well as leukemia. Strikingly, it is often associated with the most aggressive cases, and it is likely involved in metastasis. Thus, a thorough investigation of the normal activities of podocalyxin may facilitate the development of new cancer treatment strategies.


Clinical Cancer Research | 2014

Surveillance of the Tumor Mutanome by T Cells during Progression from Primary to Recurrent Ovarian Cancer

Darin A. Wick; John R. Webb; Julie S. Nielsen; Spencer D. Martin; David R. Kroeger; Katy Milne; Mauro Castellarin; Kwame Twumasi-Boateng; Peter H. Watson; Robert A. Holt; Brad H. Nelson

Purpose: Cancers accumulate mutations over time, each of which brings the potential for recognition by the immune system. We evaluated T-cell recognition of the tumor mutanome in patients with ovarian cancer undergoing standard treatment. Experimental Design: Tumor-associated T cells from 3 patients with ovarian cancer were assessed by ELISPOT for recognition of nonsynonymous mutations identified by whole exome sequencing of autologous tumor. The relative levels of mutations and responding T cells were monitored in serial tumor samples collected at primary surgery and first and second recurrence. Results: The vast majority of mutations (78/79) were not recognized by tumor-associated T cells; however, a highly specific CD8+ T-cell response to the mutation hydroxysteroid dehydrogenase–like protein 1 (HSDL1)L25V was detected in one patient. In the primary tumor, the HSDL1L25V mutation had low prevalence and expression, and a corresponding T-cell response was undetectable. At first recurrence, there was a striking increase in the abundance of the mutation and corresponding MHC class I epitope, and this was accompanied by the emergence of the HSDL1L25V-specific CD8+ T-cell response. At second recurrence, the HSDL1L25V mutation and epitope continued to be expressed; however, the corresponding T-cell response was no longer detectable. Conclusion: The immune system can respond to the evolving ovarian cancer genome. However, the T-cell response detected here was rare, was transient, and ultimately failed to prevent disease progression. These findings reveal the limitations of spontaneous tumor immunity in the setting of standard treatments and suggest a high degree of ignorance of tumor mutations that could potentially be reversed by immunotherapy. Clin Cancer Res; 20(5); 1125–34. ©2013 AACR.


Microcirculation | 2009

CD34 is a Key Regulator of Hematopoietic Stem Cell Trafficking to Bone Marrow and Mast Cell Progenitor Trafficking in the Periphery

Julie S. Nielsen; Kelly M. McNagny

CD34 is a cell‐surface sialomucin widely used for hematopoietic stem cell purification and as a marker of most vascular endothelial cells, including those of capillaries in the majority of tissues. Surprisingly, despite extensive research, the function of this sialomucin has remained elusive, with proposed roles ranging from enhancing proliferation or inhibiting differentiation to acting as a proadhesive L‐selectin ligand. Here, we review our recent studies, which suggest that CD34 does, indeed, play a role in leukocyte and HSC trafficking, but that this is through its action as a regulated blocker of cell adhesion and enhancer of migration.


PLOS Biology | 2011

ESRRA-C11orf20 is a recurrent gene fusion in serous ovarian carcinoma.

Julia Salzman; Robert J. Marinelli; Peter L. Wang; Ann E. Green; Julie S. Nielsen; Brad H. Nelson; Charles W. Drescher; Patrick O. Brown

Many ovarian cancers have a chromosomal rearrangement that fuses two neighboring genes, ESRRA and c11orf20. Similar rearrangements may be common, important features of cancer genomes that have largely escaped detection.


PLOS ONE | 2008

Tumor-Infiltrating T Cells Correlate with NY-ESO-1-Specific Autoantibodies in Ovarian Cancer

Katy Milne; Rebecca O. Barnes; Adam Girardin; Melanie A. Mawer; Nancy J. Nesslinger; Alvin Ng; Julie S. Nielsen; Robert Sahota; Eric Tran; John R. Webb; May Q. Wong; Darin A. Wick; Andrew Wray; Elissa McMurtrie; Martin Köbel; Steven E. Kalloger; C. Blake Gilks; Peter H. Watson; Brad H. Nelson

Background Tumor-infiltrating CD8+ T cells are correlated with prolonged progression-free and overall survival in epithelial ovarian cancer (EOC). A significant fraction of EOC patients mount autoantibody responses to various tumor antigens, however the relationship between autoantibodies and tumor-infiltrating T cells has not been investigated in EOC or any other human cancer. We hypothesized that autoantibody and T cell responses may be correlated in EOC and directed toward the same antigens. Methodology and Principal Findings We obtained matched serum and tumor tissue from 35 patients with high-grade serous ovarian cancer. Serum samples were assessed by ELISA for autoantibodies to the common tumor antigen NY-ESO-1. Tumor tissue was examined by immunohistochemistry for expression of NY-ESO-1, various T cell markers (CD3, CD4, CD8, CD25, FoxP3, TIA-1 and Granzyme B) and other immunological markers (CD20, MHC class I and MHC class II). Lymphocytic infiltrates varied widely among tumors and included cells positive for CD3, CD8, TIA-1, CD25, FoxP3 and CD4. Twenty-six percent (9/35) of patients demonstrated serum IgG autoantibodies to NY-ESO-1, which were positively correlated with expression of NY-ESO-1 antigen by tumor cells (r = 0.57, p = 0.0004). Autoantibodies to NY-ESO-1 were associated with increased tumor-infiltrating CD8+, CD4+ and FoxP3+ cells. In an individual HLA-A2+ patient with autoantibodies to NY-ESO-1, CD8+ T cells isolated from solid tumor and ascites were reactive to NY-ESO-1 by IFN-γ ELISPOT and MHC class I pentamer staining. Conclusion and Significance We demonstrate that tumor-specific autoantibodies and tumor-infiltrating T cells are correlated in human cancer and can be directed against the same target antigen. This implies that autoantibodies may collaborate with tumor-infiltrating T cells to influence clinical outcomes in EOC. Furthermore, serological screening methods may prove useful for identifying clinically relevant T cell antigens for immunotherapy.


OncoImmunology | 2012

Tumor-infiltrating B cells and T cells: Working together to promote patient survival.

Julie S. Nielsen; Brad H. Nelson

We recently reported a novel cooperative relationship between tumor-infiltrating B cells and CD8+ T cells in ovarian cancer, leading to increased patient survival. Here, we discuss the mechanisms whereby B cells might enhance cellular immunity, including serving as antigen-presenting cells, organizing tertiary lymphoid structures and secreting polarizing cytokines. The enhancement of both B and T-cell responses may result in more potent and sustained antitumor immunity.


Breast Cancer Research | 2015

Podocalyxin enhances breast tumor growth and metastasis and is a target for monoclonal antibody therapy

Kimberly Snyder; Michael R. Hughes; Bradley Hedberg; Jill Brandon; Diana Canals Hernaez; Peter J. Bergqvist; Frederic Cruz; Kelvin Po; Marcia L. Graves; Michelle E Turvey; Julie S. Nielsen; John A. Wilkins; John Babcook; Calvin D. Roskelley; Kelly M. McNagny

IntroductionPodocalyxin (gene name PODXL) is a CD34-related sialomucin implicated in the regulation of cell adhesion, migration and polarity. Upregulated expression of podocalyxin is linked to poor patient survival in epithelial cancers. However, it is not known if podocalyxin has a functional role in tumor progression.MethodsWe silenced podocalyxin expression in the aggressive basal-like human (MDA-MB-231) and mouse (4T1) breast cancer cell lines and also overexpressed podocalyxin in the more benign human breast cancer cell line, MCF7. We evaluated how podocalyxin affects tumorsphere formation in vitro and compared the ability of podocalyxin-deficient and podocalyxin-replete cell lines to form tumors and metastasize using xenogenic or syngeneic transplant models in mice. Finally, in an effort to develop therapeutic treatments for systemic cancers, we generated a series of antihuman podocalyxin antibodies and screened these for their ability to inhibit tumor progression in xenografted mice.ResultsAlthough deletion of podocalyxin does not alter gross cell morphology and growth under standard (adherent) culture conditions, expression of PODXL is required for efficient formation of tumorspheres in vitro. Correspondingly, silencing podocalyxin resulted in attenuated primary tumor growth and invasiveness in mice and severely impaired the formation of distant metastases. Likewise, in competitive tumor engraftment assays where we injected a 50:50 mixture of control and shPODXL (short-hairpin RNA targeting PODXL)-expressing cells, we found that podocalyxin-deficient cells exhibited a striking decrease in the ability to form clonal tumors in the lung, liver and bone marrow. Finally, to validate podocalyxin as a viable target for immunotherapy, we screened a series of novel antihuman podocalyxin antibodies for their ability to inhibit tumor progression in vivo. One of these antibodies, PODOC1, potently blocked tumor growth and metastasis.ConclusionsWe show that podocalyxin plays a key role in the formation of primary tumors and distant tumor metastasis. In addition, we validate podocalyxin as potential target for monoclonal antibody therapy to inhibit primary tumor growth and systemic dissemination.

Collaboration


Dive into the Julie S. Nielsen's collaboration.

Top Co-Authors

Avatar

Brad H. Nelson

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Kelly M. McNagny

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Spencer D. Martin

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Calvin D. Roskelley

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Eric Tran

University of Victoria

View shared research outputs
Top Co-Authors

Avatar

Michael R. Hughes

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge