Christy Willert's research while affiliated with Robarts Research Institute and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (6)


Fig. 1. CpG-CC mediated phenotypic and functional maturation and activation of DC in vitro is unaffected by SPIO labeling. Flow cytometry was performed to examine the surface expression of several DC maturation, functional and activation markers. (A) Gating strategy is shown. Letters within the gates are the cells analyzed in the corresponding letter panel. (B) Viable cells are gated on and CD11c expression is examined. (C) Viable CD11c cells were examined for their expression of CD86. (D) CD11c + CD86 + mature cells were then 
Fig. 2. CpG-CC-mediated up-regulation of DC migration surface markers is unaffected by SPIO labeling. Flow cytometry was performed to examine the surface expression of several DC migration markers. (A) Gating strategy is shown. Letters within the gates are the cells analyzed in the corresponding letter panel. (B) Viable CD11c + CD86 À cells are gated on and examined for their CD38 expression. (C) Viable CD11c + CD86 + cells were examined for their expression of CD38, CD11b, CCR7 and CD8 a . Numbers above the gate are the percentage of UT or SPIO mix DC positive for the marker indicated. Bold numbers below the gates are the percentage of CpG-CC or SPIO mix -CpG-CC cells positive for the marker 
Fig. 3. SPIO + -CpG-CC DC migrate more efficiently in vivo when compared with SPIO + DC according to cellular MRI. Either 3 3 10 5 or 1 3 10 6 
Fig. 4. Conventional histological analysis correlates well with cellular MRI data. Conventional histological analysis of mice scanned using cellular MRI was performed in order to confirm MRI data. (A and B) Representative fluorescence images of PKH + cells in popliteal LNs are shown for 
Fig. 5. Pulsing DC with SIINFEKL peptide results in efficient MHC I loading in vitro . UT, CpG-CC, SPIO + or SPIO + -CpG-CC DC were pulsed with SIINFEKL peptide for two h and collected for MHC I loading using flow cytometry. (A) Gating strategy for flow cytometry is shown. Histograms are gated on viable CD11c + cells. Representative histograms are from one of three independent experiments. Numbers above the gate are the percentage of SIINFEKL-UT or SIINFEKL-SPIO + cells positive for the marker indicated. Bold numbers below the gates are the percentage of SIINFEKL-CpG-CC or SIINFEKL-SPIO + -CpG-CC cells positive for the marker indicated. (B) CD11c + CD86 À DC from all populations had less MHC 

+2

Cellular MRI as a suitable, sensitive non-invasive modality for correlating in vivo migratory efficiencies of different dendritic cell populations with subsequent immunological outcomes
  • Article
  • Full-text available

January 2012

·

397 Reads

·

35 Citations

International Immunology

·

Christy Willert

·

Christiane Mallet

·

[...]

·

The clinical application of dendritic cells (DC) as adjuvants in immunotherapies such as the cell-based cancer vaccine continues to gain interest. The overall efficacy of this emerging immunotherapy, however, remains low. Studies suggest the stage of maturation and activation of ex vivo-prepared DC immediately prior to patient administration is critical to subsequent DC migration in vivo, which ultimately affects overall vaccine efficacy. While it is possible to generate mature and activated DC ex vivo using various stimulatory cocktails, in the case of cancer patients, the qualitative and quantitative assessment of which DC stimulatory cocktail works most effectively to enhance subsequent DC migration in vivo is difficult. Thus, a non-invasive imaging modality capable of monitoring the real-time migration of DC in long-term studies is required. In this paper, we address whether cellular magnetic resonance imaging (MRI) is sufficiently sensitive to quantitatively detect differences in the migratory abilities of two different DC preparations: untreated (resting) versus ex vivo matured in a mouse model. In order to distinguish our ex vivo-generated DC of interest from surrounding tissues in magnetic resonance (MR) images, DC were labeled in vitro with the superparamagnetic iron oxide (SPIO) nanoparticle FeREX®. Characterization of DC phenotype and function following addition of a cytokine maturation cocktail and the toll-like receptor ligand CpG, both in the presence and in the absence of SPIO, were also carried out. Conventional histological techniques were used to verify the quantitative data obtained from MR images. This study provides important information relevant to tracking the in vivo migration of ex vivo-prepared and stimulated DC.

Download
Share

Cellular magnetic resonance imaging of monocyte-derived dendritic cell migration from healthy donors and cancer patients as assessed in a scid mouse model

September 2011

·

30 Reads

·

27 Citations

Cytotherapy

BACKGROUND AIMS. The use of dendritic cells (DC) as an adjuvant in cell-based immunotherapeutic cancer vaccines is a growing field of interest. A reliable and non-invasive method to track the fate of autologous DC following their administration to patients is required in order to confirm that clinically sufficient numbers are reaching the lymph node (LN). We demonstrate that an immunocompromised mouse model can be used to conduct translational studies employing cellular magnetic resonance imaging (MRI). Such studies can provide clinically relevant information regarding the migration potential of clinical-grade DC used in cancer immunotherapies. METHODS. Human monocyte-derived dendritic cells (mo-DC) were generated from negatively selected monocytes obtained from either healthy donors or cancer patients. DC were labeled with superparamagnetic iron oxide (SPIO) nanoparticles in order to track them in vivo in a CB17scid mouse model using cellular MRI. SPIO did not have any adverse effects on DC phenotype or function, independent of donor type. Cellular MRI readily detected migration of SPIO-loaded DC in CB17scid mice. No differences in migration were observed between DC obtained from healthy donors and those obtained from donors undergoing autologous stem cell transplant for cancer therapy. CONCLUSIONS. Cellular MRI provided semi-quantitative image data that corresponded with data obtained by digital morphometry, validating cellular MRI's potential to assess DC migration in DC-based cancer immunotherapy clinical trials.


In Vivo Cellular MRI of Dendritic Cell Migration Using Micrometer-Sized Iron Oxide (MPIO) Particles

August 2011

·

34 Reads

·

40 Citations

Molecular imaging and biology: MIB: the official publication of the Academy of Molecular Imaging

This study seeks to assess the use of labeling with micron-sized iron oxide (MPIO) particles for the detection and quantification of the migration of dendritic cells (DCs) using cellular magnetic resonance imaging (MRI). DCs were labeled with red fluorescent MPIO particles for detection by cellular MRI and a green fluorescent membrane dye (PKH67) for histological detection. MPIO-labeled DCs or unlabeled control DCs were injected into mice footpads at two doses (0.1 × 10(6) and 1 × 10(6)). Images were acquired at 3 Tesla before DC injection and 2, 3, and 7 days post-DC injection. Labeling DCs with MPIO particles did not affect viability, but it did alter markers of DC activation and maturation. MRI and fluorescence microscopy allowed for the detection of MPIO-labeled DCs within the draining popliteal nodes after their injection into the footpad. This paper presents the first report of the successful use of fluorescent MPIO particles to label and track DC migration.


Labelling dendritic cells with SPIO has implications for subsequent in vivo migration as assessed with cellular MRI

July 2011

·

33 Reads

·

33 Citations

Contrast Media & Molecular Imaging

An optimized non-invasive imaging modality capable of tracking and quantifying in vivo DC migration in patients would provide clinicians with valuable information regarding therapeutic DC-based vaccine outcomes. Superparamagnetic iron oxide (SPIO) nanoparticles were used to label bone marrow-derived DC. In vivo DC migration was tracked and quantified non-invasively using cellular magnetic resonance imaging (MRI) in a mouse model. Labelling DC with SPIO reflects the kinetics of DC migration in vivo but appears to reduce overall DC migration, in part due to nanoparticle size. Magnetic separation of SPIO-labelled (SPIO(+)) DC from unlabelled (SPIO(-)) DC prior to injection improves SPIO(+) DC migration to the lymph node. Corresponding MR image data better correlate with the presence of DC in vivo; an improved immunological response is also seen. Cellular MRI is a viable, non-invasive imaging tool that can routinely track DC migration in vivo. Consideration should be given to optimizing MRI contrast agent-labelling of clinical-grade DC in order to accurately correlate DC fate to immunological outcomes in patients.


Semiquantitation of Mouse Dendritic Cell Migration In Vivo Using Cellular MRI

March 2009

·

114 Reads

·

45 Citations

Journal of Immunotherapy

Despite recent therapeutic advances, including the introduction of novel cytostatic drugs and therapeutic antibodies, many cancer patients will experience recurrent or metastatic disease. Current treatment options, particularly for those patients with metastatic breast, prostate, or skin cancers, are complex and have limited curative potential. Recent clinical trials, however, have shown that cell-based therapeutic vaccines may be used to generate broad-based, antitumor immune responses. Dendritic cells (DC) have proved to be the most efficacious cellular component for therapeutic vaccines, serving as both the adjuvant and antigen delivery vehicle. At present it is not possible to noninvasively determine the fate of DC-based vaccines after their administration to human subjects. In this study, we demonstrate that in vitro-generated mouse DC can be readily labeled with superparamagnetic iron oxide nanoparticles, Feridex, without altering cell morphology, or their phenotypic and functional maturation. Feridex-labeling enables the detection of DC in vivo after their migration to draining lymph nodes using a 1.5 T clinical magnetic resonance scanner. In addition, we report a semiquantitative approach for analysis of magnetic resonance images and show that the Feridex-induced signal void volume, and fractional signal loss, correlates with the delivery and migration of small numbers of in vitro-generated DC. These findings, together with ongoing preclinical studies, are key to gaining information critical for improving the efficacy of therapeutic vaccines for the treatment cancer, and potentially, chronic infectious diseases.


Inclusion of the viral anti-apoptotic molecule M11L in DNA vaccine vectors enhances HIV Env-specific T cell-mediated immunity

June 2008

·

26 Reads

·

7 Citations

Virology

A current goal of vaccine development against human immunodeficiency virus (HIV) is to develop a strategy that stimulates long-lasting memory T-cell responses, and provides immediate cytotoxicity in response to viral challenge. We demonstrate that the viral antiapoptotic molecule M11L promotes cellular immune responses to the HIV envelope protein. Coexpression of M11L in vitro inhibits gp140-mediated apoptosis and increases gp140 expression levels. Mice primed with M11L-pHERO DNA, followed by vCP205 boosting, exhibit significantly greater HIV-specific T-cell responses. Moreover, M11L synergizes with CpG motifs to augment anti-HIV responses and stimulates robust expansion of central memory and effector memory CD8(+) T-cells. Inclusion of M11L in a DNA vector increases the magnitude of T-cell responses, and promotes the generation of memory T-cells that provide rapid-responding CTL responses. This vaccine strategy may facilitate the generation of an efficacious vaccine for HIV, and other chronic diseases that require enhanced cell-mediated immunity, including HCV and metastatic cancer.

Citations (6)


... Among the vast repertoire of nanomaterials available, the possibility of modifying DCs with iron oxide MNPs has also been analyzed. From these studies, some general lessons were learned, regardless of the biomedical application for which they were intended: (1) at low concentrations (0-50 µg/mL Fe), MNPs do not exert significant cytotoxicity and they do not alter cell morphology, surface marker patterns, or the cytokine profile of previously matured DCs [202][203][204][205][206][207][208][209]. However, some reports did claim that MNPs can potentially reduce the viability of DCs at high concentrations (>400 µg/mL Fe), while not affecting their maturation [210]. ...

Reference:

Helios as a Potential Biomarker in Systemic Lupus Erythematosus and New Therapies Based on Immunosuppressive Cells
Cellular MRI as a suitable, sensitive non-invasive modality for correlating in vivo migratory efficiencies of different dendritic cell populations with subsequent immunological outcomes

International Immunology

... SPIO-labelled cell tracking with MRI is a powerful technique that can provide 3D resolution of therapy location in tandem with rich anatomical details of disease progression, such as tumour size, shape, and metastases. The applications of SPIO MRI contrast agents are broad -through in vivo tracking of tumours [4][5][6][7] , therapies 8 , immune cells [9][10][11][12][13] and vaccines 14 , as well as other uses, they can serve as valuable tools in the early detection of disease and the study of disease mechanisms and progression. ...

Cellular magnetic resonance imaging of monocyte-derived dendritic cell migration from healthy donors and cancer patients as assessed in a scid mouse model
  • Citing Article
  • September 2011

Cytotherapy

... Among the vast repertoire of nanomaterials available, the possibility of modifying DCs with iron oxide MNPs has also been analyzed. From these studies, some general lessons were learned, regardless of the biomedical application for which they were intended: (1) at low concentrations (0-50 µg/mL Fe), MNPs do not exert significant cytotoxicity and they do not alter cell morphology, surface marker patterns, or the cytokine profile of previously matured DCs [202][203][204][205][206][207][208][209]. However, some reports did claim that MNPs can potentially reduce the viability of DCs at high concentrations (>400 µg/mL Fe), while not affecting their maturation [210]. ...

Labelling dendritic cells with SPIO has implications for subsequent in vivo migration as assessed with cellular MRI
  • Citing Article
  • July 2011

Contrast Media & Molecular Imaging

... First, it cannot be excluded entirely that the labelling process does not affect some (known or unknown) functional features of the cells. It has been reported, e.g. that SPIO labelling of murine DCs can reduce their migratory ability in vivo [57]. Second, the imaging agent could be taken up by other phagocytic cells within the tissue, either after leaking from the cells or by clearing labelled dead cells. ...

In Vivo Cellular MRI of Dendritic Cell Migration Using Micrometer-Sized Iron Oxide (MPIO) Particles
  • Citing Article
  • August 2011

Molecular imaging and biology: MIB: the official publication of the Academy of Molecular Imaging

... From these studies, some general lessons were learned, regardless of the biomedical application for which they were intended: (1) at low concentrations (0-50 µg/mL Fe), MNPs do not exert significant cytotoxicity and they do not alter cell morphology, surface marker patterns, or the cytokine profile of previously matured DCs [202][203][204][205][206][207][208][209]. However, some reports did claim that MNPs can potentially reduce the viability of DCs at high concentrations (>400 µg/mL Fe), while not affecting their maturation [210]. In addition, DCs treated with MNPs can maintain a stable phenotype, but they may lose their optimal ability to stimulate CD4 + T lymphocytes [211]. ...

Semiquantitation of Mouse Dendritic Cell Migration In Vivo Using Cellular MRI
  • Citing Article
  • March 2009

Journal of Immunotherapy

... To counter host defenses, which could rapidly clear virally infected cells, many viruses express apoptosis-inhibiting factors. Proteins such as the myxoma virus encoded M11L protein (Everett et al., 2000;Su et al., 2008), vaccinia virus (VACV) encoded F1L protein (Wasilenko et al., 2003), deerpox virus encoded 022 protein (Banadyga et al., 2011), fowlpox virus encoded 039 protein (Banadyga et al., 2007), and ORFV encoded 125 protein (Westphal et al., 2007(Westphal et al., , 2009 have been shown to possess mitochondrial-targeting motifs and have anti-apoptotic functions. The investigation into ORFs 118-120 indicated that ORFV119 could be also confined to the mitochondria (Nagendraprabhu et al., 2017). ...

Inclusion of the viral anti-apoptotic molecule M11L in DNA vaccine vectors enhances HIV Env-specific T cell-mediated immunity
  • Citing Article
  • June 2008

Virology