Label-Free Functional Imaging of Single Molecules and Single Cells Using Surface- Enhanced Scattering Microscopy

193541-Thumbnail Image.png
Description
Recent breakthroughs in optical scattering-based imaging have enabledvisualization of entities as small as single proteins. Leveraging our innovation, Surface Enhanced Scattering Microscopy (SESM), detection of single protein binding kinetics and single DNA conformational changes have been achieved, showcasing the feasibility

Recent breakthroughs in optical scattering-based imaging have enabledvisualization of entities as small as single proteins. Leveraging our innovation, Surface Enhanced Scattering Microscopy (SESM), detection of single protein binding kinetics and single DNA conformational changes have been achieved, showcasing the feasibility of single molecule imaging. In this dissertation, I aim to harness the potential of SESM and extend its relevance in the biomedical realm. My first goal is to conduct multiplexed protein detection and parallel binding kinetics analysis with label-free digital single- molecule counting. My second goal is focused on accurate quantification of cell force. An elastic model has been developed to quantify the cell-substrate interactions and have continuously tracked cell force evolutions upon small-molecule drugs (for example, acetylcholine) stimulation, achieving a temporal resolution of approximately 60 ms over the course of 30 min without attenuating the signals. The third goal is to achieve real- time tracking of DNA self-assembly dynamics. I have demonstrated SESM's capability to image individual DNA origami monomers and established an on-chip temperature annealing system to monitor the real-time progression of DNA self-assembly. The applications of the imaging method, spanning single proteins, single DNA origami, and single cells, are poised to impact the field of biology
Date Created
2024
Agent

Nanoengineered Hydrogel Scaffolds for Enhanced Maturation and Functionality of hiPSC-derived Cardiac Tissues

193455-Thumbnail Image.png
Description
Cardiovascular diseases (CVDs), including myocardial infarction (MI), are the major cause of death globally. Considerable research has been devoted in recent years to developing in vitro cardiac tissue models utilizing human induced pluripotent stem cells (hiPSCs) for regenerative medicine, disease

Cardiovascular diseases (CVDs), including myocardial infarction (MI), are the major cause of death globally. Considerable research has been devoted in recent years to developing in vitro cardiac tissue models utilizing human induced pluripotent stem cells (hiPSCs) for regenerative medicine, disease modeling, and drug discovery applications. Notably, electroconductive hydrogel scaffolds have shown great promise in the development of functional hiPSC-derived cardiac tissues for both in vitro and in vivo cardiac research. However, the underlying mechanism(s) by which these nanoparticles contribute to the function and fate of stem cell-derived cardiac tissues have not been fully investigated. To address these knowledge gaps, this Ph.D. dissertation focuses on the mechanistic analysis of the impact of nanoengineered electroconductive hydrogel scaffolds on 2D and 3D hiPSC-derived cardiac tissues. Specifically, within the first phase of the project, hydrogel scaffolds were nanoengineered using either electroconductive or non-conductive nanoparticles to dissect the role of electroconductivity features of gold nanorods (GNRs) in the functionality of isogenic 2D hiPSC-derived cardiac patches. Extensive biological and electrophysiological assessments revealed that, while biophysical cues from the presence of nanoparticles could potentially play a role in cardiac tissue development, electroconductivity cues played a major role in enhancing the functional maturation of hiPSC-derived cardiac tissues in 2D cell-seeded cardiac patches. This dissertation further describes the application of GNRs in developing a biomimetic 3D electroconductive Heart-on-a-chip (eHOC) model. The 3D eHOC model was then leveraged to comprehensively investigate the cellular and molecular responses of isogenic human cardiac tissues to the electroconductive microenvironment through single-cell RNA sequencing (scRNAseq), an aspect not addressed in previous studies. The enhanced functional maturation of the 3D eHOC was demonstrated through extensive tissue-level and molecular-level assays. It was revealed that the GNR-based electroconductive microenvironment contributes to cardiac tissue development through the enrichment of calcium handling and cardiac contractile pathways.Overall, these findings offer additional insights into the role of electroconductive hydrogel scaffolds in regulating the functionalities of hiPSC-derived cardiac tissues. Furthermore, the proposed 3D eHOC platform could also serve as a more physiologically representative model of the in vivo microenvironment for in vitro applications, such as drug testing and disease modeling studies.
Date Created
2024
Agent

Microengineered Tumor-On-a-Chip Model to assess Tumor-Immune Interaction in Breast Cancer

189287-Thumbnail Image.png
Description
Evolving knowledge about the tumor microenvironment (TME) is driving innovation in designing novel therapies against hard-to-treat breast cancer. Addressing the immune elements within the tumor microenvironment (TME) has emerged as a highly encouraging strategy for treating cancer. Although current immunotherapies

Evolving knowledge about the tumor microenvironment (TME) is driving innovation in designing novel therapies against hard-to-treat breast cancer. Addressing the immune elements within the tumor microenvironment (TME) has emerged as a highly encouraging strategy for treating cancer. Although current immunotherapies have made advancements in reinstating the body's ability to fight tumors, the search for effective cancer treatments to combat tumor evasion remains a formidable challenge. In line with this objective, there is a pressing need to better understand the complex tumor-immune dynamics and crosstalk within the TME. To evaluate the cancer-immune interaction, this study aimed at investigating the crosstalk between naïve macrophages and cytotoxic T cells in driving tumor progression using an organotypic 3D ex vivo tumor on-a-chip model. The presented microfluidic platform consists of two distinct regions namely: The tumor region and the stroma region separated by trapezoidal microposts to ensure interconnectivity between regions thereby incorporating high spatial organization. In the established triculture platform, the complex Tumor Immune Microenvironment was successfully recapitulated by incorporating naïve macrophage and T cells within an appropriate 3D matrix. Through invasion and morphometric analyses, definitive outcomes were obtained that underscore the significant contribution of macrophages in facilitating tumor progression. Furthermore, the inclusion of T cells led to a notable decrease in the migratory speed of cancer cells and macrophages, underscoring the reciprocal communication between these two immune cell populations in the regulation of tumor advancement. Overall, this study highlights the complexity of TME and underscores the critical role of immune cells in regulating cancer progression.
Date Created
2023
Agent

Develop a Microfluidic Chip for Digital Counting of CAR-T Cells

187865-Thumbnail Image.png
Description
Chimeric antigen receptor (CAR)-T cell therapy is a type of cancer immunotherapy has shown promising results in engineering the T cells which targets a specific antigen. Despite their success rate, there are certain limitations to the use of CAR-T therapies

Chimeric antigen receptor (CAR)-T cell therapy is a type of cancer immunotherapy has shown promising results in engineering the T cells which targets a specific antigen. Despite their success rate, there are certain limitations to the use of CAR-T therapies that includes cytokine release syndrome (CRS), neurologic toxicity, lack of response in approximately 50% of treated patients, monitoring of patients treated with CAR-T therapy. However, rapid point- of- care testing helps in quantifying the circulating CAR T cells and can enhance the safety of patients, minimize the cost of CAR-T cell therapy, and ease the management process. Currently, the standard method to quantify CAR-T cell in patient blood samples are flow cytometry and quantitative polymerase chain reaction (qPCR). But these techniques are expensive and are not easily accessible and suitable for point- of- care testing to assist real- time clinical decisions. To overcome these hurdles, here I propose a solution to these problems by rapid optical imaging (ROI)- based principle to monitor and detect CAR-T cells. In this project, a microfluidic device is developed and integrated with two functions: (1) Centrifuge free, filter- based separation of white blood cells and plasma; (2) Optical imaging- based technique for digital counting of CAR T- cells. Here, I carried out proof- of- concept test on the laser cut prototype microfluidic chips as well as the surface chemistry for specific capture of CAR-T cells. These data show that the microfluidic chip can specifically capture CAR-T positive cells with concentration dependent counts of captured cells. Further development of the technology could lead to a new tool to monitor the CAR-T cells and help the clinicians to effectively measure the efficacy of CAR-T therapy treatment in a faster and safer manner.
Date Created
2023
Agent

Development and Implementation of an Automated Multiplexed Method to Study Genome-Wide DNA Methylation

171968-Thumbnail Image.png
Description
DNA methylation (DNAm) is an epigenetic mark with a critical role in regulating gene expression. Altered clinical states, including toxin exposure and viral infections, can cause aberrant DNA methylation in cells, which may persist during cell division. Current methods to

DNA methylation (DNAm) is an epigenetic mark with a critical role in regulating gene expression. Altered clinical states, including toxin exposure and viral infections, can cause aberrant DNA methylation in cells, which may persist during cell division. Current methods to study genome-wide methylome profiles of the cells require a long processing time and are expensive. Here, a novel technique called Multiplexed Methylated DNA Immunoprecipitation Sequencing (Mx-MeDIP-Seq), which is amenable to automation. Up to 15 different samples can be combined into the same run of Mx-MeDIP-Seq, using only 25 ng of DNA per sample. Mx-MeDIP-Seq was used to study DNAm profiles of peripheral blood mononuclear cells (PBMCs) in two biologically distinct RNA viral infections with different modes of transmission, symptoms, and interaction with the host immune system: human immunodeficiency virus1 (HIV-1) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Analysis of 90 hospitalized patients with SARS-CoV-2 and 57 healthy controls revealed that SARS-CoV-2 infection led to alterations in 920 methylated regions in PBMCs, resulting in a change in transcription that affects host immune response and cell survival. Analysis of publicly available RNA-Sequencing data in COVID-19 correlated with DNAm in several key pathways. These findings provide a mechanistic view toward further understanding of viral infections. Genome-wide DNAm changes post HIV-1-infection from 37 chronically ill patients compared to 17 controls revealed dysregulation of the actin cytoskeleton, which could contribute to the establishment of latency in HIV-1 infections. Longitudinal DNAm analysis identified several potentially protective and harmful genes that could contribute to disease suppression or progression.
Date Created
2022
Agent

Development of an Ultrasound-transparent Organ-on-chip Platform Towards Modeling Bubble-assisted Focused Ultrasound (BAFUS) Blood-brain-barrier (BBB) Disruption for Glioblastoma Therapy

171798-Thumbnail Image.png
Description
The blood-brain-barrier (BBB) is a significant obstacle for treating many neurological disorders. Bubble-assisted focused ultrasound (BAFUS) medicated BBB disruption is a promising technology that enables the delivery of large drug doses at targeted locations across the BBB. However, the current

The blood-brain-barrier (BBB) is a significant obstacle for treating many neurological disorders. Bubble-assisted focused ultrasound (BAFUS) medicated BBB disruption is a promising technology that enables the delivery of large drug doses at targeted locations across the BBB. However, the current lack of an in vitro model of this process hinders the full understanding of BAFUS BBB disruption for better translation into clinics. In this work, a US-transparent organ-on-chip device has been fabricated that can be critical for the in vitro modeling of the BAFUS BBB disruption. The transparency of the device window to focused ultrasound (FUS) was calculated theoretically and demonstrated by experiments. Nanobubbles were fabricated, characterized by cryogenic transmission electron microscopy (cryo-TEM), and showed bubble cavitation under FUS. Human colorectal adenocarcinoma (Caco-2) cells were used to form a good cellular barrier for BAFUS barrier disruption, as suggested by the measured permeability and transepithelial electrical resistance (TEER). Finally, barrier disruption and recovery were observed in BAFUS disrupted US-transparent organ-on-chips with Caco-2 barriers, showing great promise of the platform for future modeling BAFUS BBB disruption in vitro.
Date Created
2022
Agent

Utilizing Injection Molding to Generate Complex Three-Dimensional Cell Encapsulation Geometries

171760-Thumbnail Image.png
Description
Encapsulation is a promising technology to deliver cell-based therapies to patients safely and with reduced need for immunosuppression. Macroencapsulation devices are advantageous due to their ease of retrieval, and thus enhanced safety profile, relative to microencapsulation techniques. A major challenge

Encapsulation is a promising technology to deliver cell-based therapies to patients safely and with reduced need for immunosuppression. Macroencapsulation devices are advantageous due to their ease of retrieval, and thus enhanced safety profile, relative to microencapsulation techniques. A major challenge in macroencapsulation device design is ensuring sufficient oxygen transport to encapsulated cells, requiring high surface area-to-volume device geometries. In this work, a hydrogel injection molding biofabrication method was modified to design and generate complex three-dimensional macroencapsulation devices that have greater complexity in the z-axis. The rheological properties of diverse hydrogels were evaluated and used to perform computational flow modeling within injection mold devices to evaluate pressure regimes suitable for cell viability. 3D printed device designs were evaluated for the reproducibility of hydrogel filling and extraction. This work demonstrated that injection molding biofabrication to construct complex three-dimensional geometries is feasible in pressure regimes consistent with preserving cell viability. Future work will evaluate encapsulated cell viability after injection molding.
Date Created
2022
Agent

Mutant P53-specific Differential Regulation of Yap1/Taz Proteins in Promoting Invasion of Mammary Epithelial Cells

168657-Thumbnail Image.png
Description
The TP53 tumor suppressor gene is the most frequently mutated gene in human cancers. In the highly aggressive triple negative breast cancer (TNBC), TP53 is mutated in 80% of cases. TNBC lacks viable drug targets, resulting in a low prognosis

The TP53 tumor suppressor gene is the most frequently mutated gene in human cancers. In the highly aggressive triple negative breast cancer (TNBC), TP53 is mutated in 80% of cases. TNBC lacks viable drug targets, resulting in a low prognosis (12.2% 5 year survivability rate). As such, the discovery of druggable targets in TNBC would be beneficial. Mutated p53 protein typically occurs as a missense mutation and often endows cancer cells with gain of function (GOF) properties by dysregulating metabolic pathways. One of these frequently dysregulated pathways is the Hippo/Yes-associated protein-1 (YAP1)/WW Domain Containing Transcription Regulator 1 (TAZ) tumor suppressor pathway. This study therefore analyzed the involvement of the Hippo/YAP1/TAZ pathway in p53-mediated breast cancer cell invasion. From an RNA-seq screen in MCF10A cell lines harboring different TP53 missense mutations, each with a differing invasive phenotype, components of the Hippo pathway were found to correlate with cell invasion. To this end, the active and inactive forms of YAP1 and TAZ were studied. Phosphorylated (inactive) YAP1 and TAZ are retained in the cytoplasm and eventually degraded. Unphosphorylated (active) YAP1 and TAZ translocate to the nucleus to activate TEAD-family transcription factors, inducing cell survival and proliferation genes leading to increased cell invasion. Using quantitative western blot analysis, it was found that inactive TAZ expression was lower in the most invasive cell lines and higher in the least invasive cell lines (p = 0.003). Moreover, the ratio of inactive TAZ protein to total TAZ protein was also shown to be predominantly lower in the invasive cell lines compared to the non-invasive lines (p = 0.04). Finally, active TAZ expression was primarily higher in p53-mutant invasive cell lines and lower in non-invasive p53 mutant cells. Additionally, although YAP1 and TAZ are thought to be functionally redundant, the pattern seen in TAZ was not seen in the YAP1 protein. Taken together, the results demonstrated here suggest that TAZ holds a more dominant role in governing TNBC cell invasion compared to YAP1 and further highlights TAZ as a potential therapeutic target in TNBC.
Date Created
2022
Agent

Development of an On-Chip Microfluidic Model of Human Cardiac Tissue as a Platform for the Study of Cardiovascular Diseases

Description
Cardiovascular disease (CVD) remains the leading cause of mortality, resulting in 1 out of 4 deaths in the United States at the alarming rate of 1 death every 36 seconds, despite great efforts in ongoing research. In vitro research to

Cardiovascular disease (CVD) remains the leading cause of mortality, resulting in 1 out of 4 deaths in the United States at the alarming rate of 1 death every 36 seconds, despite great efforts in ongoing research. In vitro research to study CVDs has had limited success, due to lack of biomimicry and structural complexity of 2D models. As such, there is a critical need to develop a 3D, biomimetic human cardiac tissue within precisely engineered in vitro platforms. This PhD dissertation involved development of an innovative anisotropic 3D human stem cell-derived cardiac tissue on-a-chip model (i.e., heart on-a-chip), with an enhanced maturation tissue state, as demonstrated through extensive biological assessments. To demonstrate the potential of the platform to study cardiac-specific diseases, the developed heart on-a-chip was used to model myocardial infarction (MI) due to exposure to hypoxia. The successful induction of MI on-a-chip (heart attack-on-a-chip) was evidenced through fibrotic tissue response, contractile dysregulation, and transcriptomic regulation of key pathways.This dissertation also described incorporation of CRISPR/Cas9 gene-editing to create a human induced pluripotent stem cell line (hiPSC) with a mutation in KCNH2, the gene implicated in Long QT Syndrome Type 2 (LQTS2). This novel stem cell line, combined with the developed heart on-a-chip technology, led to creation of a 3D human cardiac on-chip tissue model of LQTS2 disease.. Extensive mechanistic biological and electrophysiological characterizations were performed to elucidate the mechanism of R531W mutation in KCNH2, significantly adding to existing knowledge about LQTS2. In summary, this thesis described creation of a LQTS2 cardiac on-a-chip model, incorporated with gene-edited hiPSC-cardiomyocytes and hiPSC-cardiac fibroblasts, to study mechanisms of LQTS2. Overall, this dissertation provides broad impact for fundamental studies toward cardiac biological studies as well as drug screening applications. Specifically, the developed heart on-a-chip from this dissertation provides a unique alternative platform to animal testing and 2D studies that recapitulates the human myocardium, with capabilities to model critical CVDs to study disease mechanisms, and/or ultimately lead to development of future therapeutic strategies.
Date Created
2021
Agent

Magnetic Needle Steering for Medical Applications

161936-Thumbnail Image.png
Description
Many medical procedures, like surgeries, deal with the physical manipulation of sensitive internal tissues. Over time, new medical tools and techniques have been developed to improve the safety and efficacy of these procedures. Despite the leaps and bounds of progress

Many medical procedures, like surgeries, deal with the physical manipulation of sensitive internal tissues. Over time, new medical tools and techniques have been developed to improve the safety and efficacy of these procedures. Despite the leaps and bounds of progress made up to the present day, three major obstacles (among others) persist, bleeding, pain, and the risk of infection. Advances in minimally invasive treatments have transformed many formerly risky surgical procedures into very safe and highly successful routines. Minimally invasive surgeries are characterized by small incision profiles compared to the large incisions in open surgeries, minimizing the aforementioned issues. Minimally invasive procedures lead to several benefits, such as shorter recovery time, fewer complications, and less postoperative pain. In minimally invasive surgery, doctors use various techniques to operate with less damage to the body than open surgery. Today, these procedures have an established, successful history and promising future. Steerable needles are one of the tools proposed for minimally invasive operations. Needle steering is a method for guiding a long, flexible needle through curved paths to reach targets deep in the body, eliminating the need for large incisions. In this dissertation, we present a new needle steering technology: magnetic needle steering. This technology is proposed to address the limitations of conventional needle steering that hindered its clinical applications. Magnetic needle steering eliminates excessive tissue damage, restrictions of the minimum radius of curvature, and the need for a complex nonlinear model, to name a few. It also allows fabricating the needle shaft out of soft and tissue-compliant materials. This is achieved by first developing an electromagnetic coil system capable of producing desired magnetic fields and gradients; then, a magnetically actuated needle is designed, and its effectiveness is experimentally evaluated. Afterward, the scalability of this technique was tested using permanent magnets controlled with a robotic arm. Furthermore, different configurations of permanent magnets and their influence on the magnetic field are investigated, enabling the possibility of designing a desired magnetic field for a specific surgical procedure and operation on a particular organ. Finally, potential future directions towards animal studies and clinical trials are discussed.
Date Created
2021
Agent