Network


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

Hotspot


Dive into the research topics where Brian A. Aguado is active.

Publication


Featured researches published by Brian A. Aguado.


Nature Communications | 2015

In vivo capture and label-free detection of early metastatic cells

Samira M. Azarin; Ji Yi; Robert Michael Gower; Brian A. Aguado; Megan E. Sullivan; Ashley G. Goodman; Eric J. Jiang; Shreyas S. Rao; Yinying Ren; Susan L. Tucker; Vadim Backman; Jacqueline S. Jeruss; Lonnie D. Shea

Breast cancer is a leading cause of death for women, with mortality resulting from metastasis. Metastases are often detected once tumor cells affect the function of solid organs, with a high disease burden limiting effective treatment. Here we report a method for the early detection of metastasis using an implanted scaffold to recruit and capture metastatic cells in vivo, which achieves high cell densities and reduces the tumor burden within solid organs 10-fold. Recruitment is associated with infiltration of immune cells, which include Gr1hiCD11b+ cells. We identify metastatic cells in the scaffold through a label-free detection system using inverse-spectroscopic optical coherence tomography, which identifies changes to nanoscale tissue architecture associated with the presence of tumor cells. For patients at risk of recurrence, scaffold implantation following completion of primary therapy has the potential to identify metastatic disease at the earliest stage, enabling initiation of therapy while the disease burden is low.


Acta Biomaterialia | 2013

In vivo acute and humoral response to three-dimensional porous soy protein scaffolds.

Karen B. Chien; Brian A. Aguado; Paul J. Bryce; Ramille N. Shah

Increasing interest in using soy biomaterials for tissue engineering applications has prompted investigation into the in vivo biocompatibility of soy implants. In this study, the biocompatibility of soy protein scaffolds fabricated using freeze-drying and 3-D printing was assessed using a subcutaneous implant model in BALB/c mice. The main objectives of this study were: (1) to compare soy protein with bovine collagen, a well-characterized natural protein implant, by implanting scaffolds of the same protein weight, and (2) to observe the effects of soy scaffold microstructure and amount of protein loading, which also alters the degradation properties, on the acute and humoral immune responses towards soy. Results showed that freeze-dried soy scaffolds fully degraded after 14 days, whereas collagen scaffolds (of the same protein weight) remained intact for 56 days. Furthermore, Massons trichrome staining showed little evidence of damage or fibrosis at the soy implant site. Scaffolds of higher soy protein content, however, were still present after 56 days. H&E staining revealed that macrophage infiltration was hindered in the denser bioplotted soy scaffolds, causing slower degradation. Analysis of soy-specific antibodies in mouse serum after implantation revealed levels of IgG1 that correlated with higher scaffold weight and protein density. However, no soy-specific IgE was detected, indicating the absence of an allergic response to the soy implants. These results demonstrate that soy protein could be an acceptable biocompatible implant for tissue regeneration, and that scaffold porosity, soy protein density and scaffold degradation rate significantly affect the acute and humoral immune response.


Nature Biomedical Engineering | 2017

Engineering the pre-metastatic niche

Brian A. Aguado; Grace G. Bushnell; Shreyas S. Rao; Jacqueline S. Jeruss; Lonnie D. Shea

The pre-metastatic niche — the accumulation of aberrant immune cells and extracellular matrix proteins in target organs — primes the initially healthy organ microenvironment and renders it amenable for subsequent metastatic cell colonization. By attracting metastatic cancer cells, mimics of the pre-metastatic niche offer both diagnostic and therapeutic potential. However, deconstructing the complexity of the niche by identifying the interactions between cell populations and the mediatory roles of the immune system, soluble factors, extracellular matrix proteins, and stromal cells has proved challenging. Experimental models need to recapitulate niche-population biology in situ and mediate in vivo tumour-cell homing, colonization and proliferation. In this Review, we outline the biology of the pre-metastatic niche and discuss advances in engineered niche-mimicking biomaterials that regulate the behaviour of tumour cells at an implant site. Such oncomaterials offer strategies for early detection of metastatic events, inhibiting the formation of the pre-metastatic niche, and attenuating metastatic progression.


Acta Biomaterialia | 2016

Extracellular matrix mediators of metastatic cell colonization characterized using scaffold mimics of the pre-metastatic niche.

Brian A. Aguado; Jordan R. Caffe; Dhaval Nanavati; Shreyas S. Rao; Grace G. Bushnell; Samira M. Azarin; Lonnie D. Shea

UNLABELLED Metastatic tumor cells colonize the pre-metastatic niche, which is a complex microenvironment consisting partially of extracellular matrix (ECM) proteins. We sought to identify and validate novel contributors to tumor cell colonization using ECM-coated poly(ε-caprolactone) (PCL) scaffolds as mimics of the pre-metastatic niche. Utilizing orthotopic breast cancer mouse models, fibronectin and collagen IV-coated scaffolds implanted in the subcutaneous space captured colonizing tumor cells, showing a greater than 2-fold increase in tumor cell accumulation at the implant site compared to uncoated scaffolds. As a strategy to identify additional ECM colonization contributors, decellularized matrix (DCM) from lungs and livers containing metastatic tumors were characterized. In vitro, metastatic cell adhesion was increased on DCM coatings from diseased organs relative to healthy DCM. Furthermore, in vivo implantations of diseased DCM-coated scaffolds had increased tumor cell colonization relative to healthy DCM coatings. Mass-spectrometry proteomics was performed on healthy and diseased DCM to identify candidates associated with colonization. Myeloperoxidase was identified as abundantly present in diseased organs and validated as a contributor to colonization using myeloperoxidase-coated scaffold implants. This work identified novel ECM proteins associated with colonization using decellularization and proteomics techniques and validated candidates using a scaffold to mimic the pre-metastatic niche. STATEMENT OF SIGNIFICANCE The pre-metastatic niche consists partially of ECM proteins that promote metastatic cell colonization to a target organ. We present a biomaterials-based approach to mimic this niche and identify ECM mediators of colonization. Using murine breast cancer models, we implanted microporous PCL scaffolds to recruit colonizing tumor cells in vivo. As a strategy to modulate colonization, we coated scaffolds with various ECM proteins, including decellularized lung and liver matrix from tumor-bearing mice. After characterizing the organ matrices using proteomics, myeloperoxidase was identified as an ECM protein contributing to colonization and validated using our scaffold. Our scaffold provides a platform to identify novel contributors to colonization and allows for the capture of colonizing tumor cells for a variety of downstream clinical applications.


Scientific Reports | 2015

Secretome identification of immune cell factors mediating metastatic cell homing.

Brian A. Aguado; Jia J. Wu; Samira M. Azarin; Dhaval Nanavati; Shreyas S. Rao; Grace G. Bushnell; Chaitanya B. Medicherla; Lonnie D. Shea

Metastatic cell homing is a complex process mediated in part by diffusible factors secreted from immune cells found at a pre-metastatic niche. We report on connecting secretomics and TRanscriptional Activity CEll aRray (TRACER) data to identify functional paracrine interactions between immune cells and metastatic cells as novel mediators of homing. Metastatic breast cancer mouse models were used to generate a diseased splenocyte conditioned media (D-SCM) containing immune cell secreted factors. MDA-MB-231 metastatic cell activity including cell invasion, migration, transendothelial migration, and proliferation were increased in D-SCM relative to control media. Our D-SCM secretome analysis yielded 144 secreted factor candidates that contribute to increased metastatic cell activity. The functional mediators of homing were identified using MetaCore software to determine interactions between the immune cell secretome and the TRACER-identified active transcription factors within metastatic cells. Among the 5 candidate homing factors identified, haptoglobin was selected and validated in vitro and in vivo as a key mediator of homing. Our studies demonstrate a novel systems biology approach to identify functional signaling factors associated with a cellular phenotype, which provides an enabling tool that complements large-scale protein identification provided by proteomics.


Cancer Research | 2016

Enhanced survival with implantable scaffolds that capture metastatic breast cancer cells in vivo

Shreyas S. Rao; Grace G. Bushnell; Samira M. Azarin; Graham Spicer; Brian A. Aguado; Jenna Stoehr; Eric J. Jiang; Vadim Backman; Lonnie D. Shea; Jacqueline S. Jeruss

The onset of distant organ metastasis from primary breast cancer marks the transition to a stage IV diagnosis. Standard imaging modalities often detect distant metastasis when the burden of disease is high, underscoring the need for improved methods of detection to allow for interventions that would impede disease progression. Here, microporous poly(ε-caprolactone) scaffolds were developed that capture early metastatic cells and thus serve as a sentinel for early detection. These scaffolds were used to characterize the dynamic immune response to the implant spanning the acute and chronic foreign body response. The immune cell composition had stabilized at the scaffold after approximately 1 month and changed dramatically within days to weeks after tumor inoculation, with CD11b(+)Gr1(hi)Ly6C(-) cells having the greatest increase in abundance. Implanted scaffolds recruited metastatic cancer cells that were inoculated into the mammary fat pad in vivo, which also significantly reduced tumor burden in the liver and brain. Additionally, cancer cells could be detected using a label-free imaging modality termed inverse spectroscopic optical coherence tomography, and we tested the hypothesis that subsequent removal of the primary tumor after early detection would enhance survival. Surgical removal of the primary tumor following cancer cell detection in the scaffold significantly improved disease-specific survival. The enhanced disease-specific survival was associated with a systemic reduction in the CD11b(+)Gr1(hi)Ly6C(-) cells as a consequence of the implant, which was further supported by Gr-1 depletion studies. Implementation of the scaffold may provide diagnostic and therapeutic options for cancer patients in both the high-risk and adjuvant treatment settings. Cancer Res; 76(18); 5209-18. ©2016 AACR.


Science Translational Medicine | 2018

Engineering precision biomaterials for personalized medicine

Brian A. Aguado; Joseph C. Grim; Adrianne M. Rosales; Jana J. Watson-Capps; Kristi S. Anseth

A unit operations approach for designing precision biomaterial-based medical devices may lead to more effective personalized therapies. As the demand for precision medicine continues to rise, the “one-size-fits-all” approach to designing medical devices and therapies is becoming increasingly outdated. Biomaterials have considerable potential for transforming precision medicine, but individual patient complexity often necessitates integrating multiple functions into a single device to successfully tailor personalized therapies. Here, we introduce an engineering strategy based on unit operations to provide a unified vocabulary and contextual framework to aid the design of biomaterial-based devices and accelerate their translation.


ACS Applied Materials & Interfaces | 2017

Injectable Carbon Nanotube-Functionalized Reverse Thermal Gel Promotes Cardiomyocytes Survival and Maturation

Brisa Peña; Susanna Bosi; Brian A. Aguado; Daniele Borin; Nikki L. Farnsworth; Evgenia Dobrinskikh; Teisha J. Rowland; Valentina Martinelli; Mark Y. Jeong; Matthew R.G. Taylor; Carlin S. Long; Robin Shandas; Orfeo Sbaizero; Maurizio Prato; Kristi S. Anseth; Daewon Park; Luisa Mestroni

The ability of the adult heart to regenerate cardiomyocytes (CMs) lost after injury is limited, generating interest in developing efficient cell-based transplantation therapies. Rigid carbon nanotubes (CNTs) scaffolds have been used to improve CMs viability, proliferation, and maturation, but they require undesirable invasive surgeries for implantation. To overcome this limitation, we developed an injectable reverse thermal gel (RTG) functionalized with CNTs (RTG-CNT) that transitions from a solution at room temperature to a three-dimensional (3D) gel-based matrix shortly after reaching body temperature. Here we show experimental evidence that this 3D RTG-CNT system supports long-term CMs survival, promotes CMs alignment and proliferation, and improves CMs function when compared with traditional two-dimensional gelatin controls and 3D plain RTG system without CNTs. Therefore, our injectable RTG-CNT system could potentially be used as a minimally invasive tool for cardiac tissue engineering efforts.


ACS central science | 2018

A Reversible and Repeatable Thiol–Ene Bioconjugation for Dynamic Patterning of Signaling Proteins in Hydrogels

Joseph C. Grim; Tobin E. Brown; Brian A. Aguado; Douglas A. Chapnick; Alexandrea L. Viert; Xuedong Liu; Kristi S. Anseth

Biomolecule-functionalized hydrogels have emerged as valuable cell culture platforms to recapitulate the mechanical and biochemical properties of the extracellular niche. The typical strategy to functionalize hydrogels with biomolecules involves directly tethering them to the hydrogel backbone resulting in a static material. Thus, this approach fails to capture the dynamic changes in biomolecule composition that occur during biological processes or that may be required for regenerative medicine applications. Moreover, it also limits the scope of biomolecules to simple peptides, as signaling proteins generally have poor stability under cell culture conditions and lose their bioactivity over time. To that end, we sought to develop a bioconjugation reaction that would enable reversible and repeatable tethering of signaling proteins to hydrogels, so that spent protein could be released on-demand and replaced with fresh protein as needed. Specifically, we designed an allyl sulfide chain-transfer agent that enables a reversible, photomediated, thiol–ene bioconjugation of signaling proteins to hydrogels. Upon addition of a thiolated protein to the allyl sulfide moiety, the previously tethered protein is released, and the “ene” functionality is regenerated. Using this approach, we demonstrate that protein patterning can be achieved in hydrogels through a thiol–ene reaction, and the patterned protein can then be released through a subsequent thiol–ene reaction of a PEG thiol. Importantly, this process is repeatable through multiple iterations and proceeds at physiologically relevant signaling protein concentrations. Finally, we demonstrate that whole signaling proteins can be patterned and released in the presence of cells, and that cells respond to their presentation with spatial fidelity. Combined, these data represent the first example of a methodology that enables fully reversible and repeatable patterning and release of signaling proteins from hydrogels.


Advanced Healthcare Materials | 2018

Biomaterial Scaffolds as Pre‐metastatic Niche Mimics Systemically Alter the Primary Tumor and Tumor Microenvironment

Brian A. Aguado; Rachel M. Hartfield; Grace G. Bushnell; Joseph T. Decker; Samira M. Azarin; Dhaval Nanavati; Matthew J. Schipma; Shreyas S. Rao; Robert S. Oakes; Yining Zhang; Jacqueline S. Jeruss; Lonnie D. Shea

Primary tumor (PT) immune cells and pre-metastatic niche (PMN) sites are critical to metastasis. Recently, synthetic biomaterial scaffolds used as PMN mimics are shown to capture both immune and metastatic tumor cells. Herein, studies are performed to investigate whether the scaffold-mediated redirection of immune and tumor cells would alter the primary tumor microenvironment (TME). Transcriptomic analysis of PT cells from scaffold-implanted and mock-surgery mice identifies differentially regulated pathways relevant to invasion and metastasis progression. Transcriptomic differences are hypothesized to result from scaffold-mediated modulations of immune cell trafficking and phenotype in the TME. Culturing tumor cells with conditioned media generated from PT immune cells of scaffold-implanted mice decrease invasion in vitro more than two-fold relative to mock surgery controls and reduce activity of invasion-promoting transcription factors. Secretomic characterization of the conditioned media delineates interactions between immune cells in the TME and tumor cells, showing an increase in the pan-metastasis inhibitor decorin and a concomitant decrease in invasion-promoting chemokine (C-C motif) ligand 2 (CCL2) in scaffold-implanted mice. Flow cytometric and transcriptomic profiling of PT immune cells identify phenotypically distinct tumor-associated macrophages (TAMs) in scaffold-implanted mice, which may contribute to an invasion-suppressive TME. Taken together, this study demonstrates biomaterial scaffolds systemically influence metastatic progression through manipulation of the TME.

Collaboration


Dive into the Brian A. Aguado's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kristi S. Anseth

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge