Matching Items (13)
Filtering by

Clear all filters

Description
Myocardial infarction (MI) remains the leading cause of mortality and morbidity in the U.S., accounting for nearly 140,000 deaths per year. Heart transplantation and implantation of mechanical assist devices are the options of last resort for intractable heart failure, but these are limited by lack of organ donors and potential

Myocardial infarction (MI) remains the leading cause of mortality and morbidity in the U.S., accounting for nearly 140,000 deaths per year. Heart transplantation and implantation of mechanical assist devices are the options of last resort for intractable heart failure, but these are limited by lack of organ donors and potential surgical complications. In this regard, there is an urgent need for developing new effective therapeutic strategies to induce regeneration and restore the loss contractility of infarcted myocardium. Over the past decades, regenerative medicine has emerged as a promising strategy to develop scaffold-free cell therapies and scaffold-based cardiac patches as potential approaches for MI treatment. Despite the progress, there are still critical shortcomings associated with these approaches regarding low cell retention, lack of global cardiomyocytes (CMs) synchronicity, as well as poor maturation and engraftment of the transplanted cells within the native myocardium. The overarching objective of this dissertation was to develop two classes of nanoengineered cardiac patches and scaffold-free microtissues with superior electrical, structural, and biological characteristics to address the limitations of previously developed tissue models. An integrated strategy, based on micro- and nanoscale technologies, was utilized to fabricate the proposed tissue models using functionalized gold nanomaterials (GNMs). Furthermore, comprehensive mechanistic studies were carried out to assess the influence of conductive GNMs on the electrophysiology and maturity of the engineered cardiac tissues. Specifically, the role of mechanical stiffness and nano-scale topographies of the scaffold, due to the incorporation of GNMs, on cardiac cells phenotype, contractility, and excitability were dissected from the scaffold’s electrical conductivity. In addition, the influence of GNMs on conduction velocity of CMs was investigated in both coupled and uncoupled gap junctions using microelectrode array technology. Overall, the key contributions of this work were to generate new classes of electrically conductive cardiac patches and scaffold-free microtissues and to mechanistically investigate the influence of conductive GNMs on maturation and electrophysiology of the engineered tissues.
ContributorsNavaei, Ali (Author) / Nikkhah, Mehdi (Thesis advisor) / Brafman, David (Committee member) / Migrino, Raymond Q. (Committee member) / Stabenfeldt, Sarah (Committee member) / Vernon, Brent (Committee member) / Arizona State University (Publisher)
Created2018
154988-Thumbnail Image.png
Description
Stromal cell-derived factor-1α (SDF-1α) and its key receptor, CXCR4 are ubiquitously expressed in systems across the body (e.g. liver, skin, lung, etc.). This signaling axis regulates a myriad of physiological processes that range from maintaining of organ homeostasis in adults to, chemotaxis of stem/progenitor and immune cell types after injury.

Stromal cell-derived factor-1α (SDF-1α) and its key receptor, CXCR4 are ubiquitously expressed in systems across the body (e.g. liver, skin, lung, etc.). This signaling axis regulates a myriad of physiological processes that range from maintaining of organ homeostasis in adults to, chemotaxis of stem/progenitor and immune cell types after injury. Given its potential role as a therapeutic target for diverse applications, surprisingly little is known about how SDF-1α mediated signaling propagates through native tissues. This limitation ultimately constrains rational design of interventional biomaterials that aim to target the SDF-1α/CXCR4 signaling axis. One application of particular interest is traumatic brain injury (TBI) for which, there are currently no means of targeting the underlying biochemical pathology to improve prognosis.

Growing evidence suggests a relationship between SDF-1α/CXCR4 signaling and endogenous neural progenitor/stem cells (NPSC)-mediated regeneration after neural injury. Long-term modulation of the SDF-1α/CXCR4 signaling axis is thus hypothesized as a possible avenue for harnessing and amplifying endogenous regenerative mechanisms after TBI. In order to understand how the SDF-1α/CXCR4 signaling can be modulated in vivo, we first developed and characterized a sustained protein delivery platform in vitro. We were the first, to our knowledge, to demonstrate that protein release profiles from poly(D,L,-lactic-co-glycolic) acid (PLGA) particles can be tuned independent of particle fabrication parameters via centrifugal fractioning. This process of physically separating the particles altered the average diameter of a particle population, which is in turn was correlated to critical release characteristics. Secondly, we demonstrated sustained release of SDF-1α from PLGA/fibrin composites (particles embedded in fibrin) with tunable burst release as a function of fibrin concentration. Finally, we contrasted the spatiotemporal localization of endogenous SDF-1α and CXCR4 expression in response to either bolus or sustained release of exogenous SDF-1α. Sustained release of exogenous SDF-1α induced spatially diffuse endogenous SDF-1/CXCR4 expression relative to bolus SDF-1 administration; however, the observed effects were transient in both cases, persisting only to a maximum of 3 days post injection. These studies will inform future systematic evaluations of strategies that exploit SDF-1α/CXCR4 signaling for diverse applications.
ContributorsDutta, Dipankar (Author) / Stabenfeldt, Sarah E (Thesis advisor) / Kleim, Jeffrey (Committee member) / Nikkhah, Mehdi (Committee member) / Sirianni, Rachael (Committee member) / Vernon, Brent (Committee member) / Arizona State University (Publisher)
Created2016
155581-Thumbnail Image.png
Description
A tumor is a heterogeneous combination of proliferating tumor cells, infiltrating immune cells and stromal components along with a variety of associated host tissue cells, collectively termed the tumor microenvironment (TME). The constituents of the TME and their interaction with the host organ shape and define the properties of tumors

A tumor is a heterogeneous combination of proliferating tumor cells, infiltrating immune cells and stromal components along with a variety of associated host tissue cells, collectively termed the tumor microenvironment (TME). The constituents of the TME and their interaction with the host organ shape and define the properties of tumors and contribute towards the acquisition of hallmark traits such as hypoxia. Hypoxia imparts resistance to cancer from chemotherapy and radiotherapy due to the decreased production of reactive oxygen species and also promotes angiogenesis, malignant progression and metastasis. It also provides a powerful physiological stimulus that can be exploited as a tumor-specific condition, allowing for the rational design of anticancer hypoxia-activated pro-drugs (HAP). Accurate evaluation of tumor oxygenation in response to therapeutics interventions at various stages of growth should provide a better understanding of tumor response to therapy, potentially allowing therapy to be tailored to individual characteristics. The primary goal of this research was to investigate the utility of prospective identification of hypoxic tumors, by two different Magnetic Resonance Imaging (MRI) based oximetry approaches, in successful treatment with hypoxia activated therapy. In the present study, I report the utility of these two techniques 1) PISTOL (Proton Imaging of Siloxanes to map Tissue Oxygenation Levels) and 2) use of a hypoxia binding T1 contrast agent GdDO3NI in reporting the modulations of hypoxia pre and post hypoxia activated therapies in pre-clinical models of cancer. I have performed these studies in non-small cell lung cancer (NSCLC) and epidermoid carcinoma (NCI-H1975 and A431 cell lines, respectively) as well as in patient derived xenograft models of NSCLC. Both the oximetry techniques have the potential to differentiate between normoxic and hypoxic regions of the tumor and reveal both baseline heterogeneity and differential response to therapeutic intervention. The response of the tumor models to therapeutic interventions indicates that, in conjunction with pO2, other factors such as tumor perfusion (essential for delivering HAPs) and relative expression of nitroreductases (essential for activating HAPs) may play an important role. The long term goal of the proposed research is the clinical translation of both the MRI techniques and aiding the design and development of personalized therapy (e.g. patient stratification for novel hypoxia activated pro-drugs) particularly for cancer.
ContributorsAgarwal, Shubhangi (Author) / Kodibagkar, Vikram D (Thesis advisor) / Inge, Landon J (Committee member) / Nikkhah, Mehdi (Committee member) / Pagel, Mark D. (Committee member) / Sadleir, Rosalind J (Committee member) / Arizona State University (Publisher)
Created2017
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 study CVDs has had limited success, due to lack of

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.
ContributorsVeldhuizen, Jaimeson (Author) / Nikkhah, Mehdi (Thesis advisor) / Brafman, David (Committee member) / Ebrahimkhani, Mo (Committee member) / Migrino, Raymond Q (Committee member) / Plaisier, Christopher (Committee member) / Arizona State University (Publisher)
Created2021
Description
Solid tumors advance from benign stage to a deadly metastatic state due to the complex interaction between cancer cells and tumor microenvironment (TME) including stromal cells and extracellular matrix (ECM). Multiple studies have demonstrated that ECM dysregulation is one of the critical hallmarks of cancer progression leading to formation of

Solid tumors advance from benign stage to a deadly metastatic state due to the complex interaction between cancer cells and tumor microenvironment (TME) including stromal cells and extracellular matrix (ECM). Multiple studies have demonstrated that ECM dysregulation is one of the critical hallmarks of cancer progression leading to formation of a desmoplastic microenvironment that participates in tumor progression. Cancer associated fibroblasts (CAFs) are the predominant stromal cell type that participates in desmoplasia by depositing matrix proteins and increasing ECM stiffness. Although the influence of matrix stiffness on enhanced tumorigenicity has been well studied, the biological understanding about the dynamic changes in ECM architecture and the role of cancer-stromal cell interaction on ECM remodeling is still limited.

In this dissertation, the primary goal was to develop a comprehensive cellular and molecular level understanding of ECM remodeling due to the interaction of breast tumor cells and CAFs. To that end, a novel three-dimensional (3D) high-density tumor-stroma model was fabricated in which breast tumor cells (MDA-MB-231 and MCF7) were spatially organized surrounded by CAF-embedded collagen-I hydrogel (Aim 1). Further the platform was integrated with atomic force microscopy to assess the dynamic changes in ECM composition and stiffness during active tumor invasion. The results established an essential role of crosstalk between breast tumor cells and CAFs in ECM remodeling. The studies were further extended by dissecting the mode of interaction between tumor cells and CAFs followed by characterization of the role of various tumor secreted factors on ECM remodeling (Aim 2). The results for the first time established a critical role of paracrine signaling between breast tumor cells and CAFs in modulating biophysical properties of ECM. More in-depth analysis highlighted the role of tumor secreted cytokines, specifically PDGF-AA/BB, on CAF-induced desmoplasia. In aim 3, the platform was further utilized to test the synergistic influence of anti-fibrotic drug (tranilast) in conjugation with chemotherapeutic drug (Doxorubicin) on desmoplasia and tumor progression in the presence of CAFs. Overall this dissertation provided an in-depth understanding on the impact of breast cancer-stromal cell interaction in modulating biophysical properties of the ECM and identified the crucial role of tumor secreted cytokines including PDGF-AA/BB on desmoplasia.
ContributorsSaini, Harpinder (Author) / Nikkhah, Mehdi (Thesis advisor) / Ros, Robert (Committee member) / LaBaer, Joshua (Committee member) / Kodibagkar, Vikram (Committee member) / Ebrahimkhani, Mohammad (Committee member) / Arizona State University (Publisher)
Created2019
155660-Thumbnail Image.png
Description
This work describes the development of a device for measuring CO2 in breath, which has applications in monitoring a variety of health issues, such as Chronic Obstructive Pulmonary Disease (COPD), asthma, and cardiovascular disease. The device takes advantage of colorimetric sensing technology in order to maintain a low cost and

This work describes the development of a device for measuring CO2 in breath, which has applications in monitoring a variety of health issues, such as Chronic Obstructive Pulmonary Disease (COPD), asthma, and cardiovascular disease. The device takes advantage of colorimetric sensing technology in order to maintain a low cost and high user-friendliness. The sensor consists of a pH dye, reactive element, and base coated on a highly porous Teflon membrane. The transmittance of the sensor is measured in the device via a simple LED/photodiode system, along with the flow rate, ambient relative humidity, and barometric pressure. The flow is measured by a newly developed flow meter described in this work, the Confined Pitot Tube (CPT) flow meter, which provides a high accuracy with reduced flow-resistance with a standard differential pressure transducer. I demonstrate in this work that the system has a high sensitivity, high specificity, fast time-response, high reproducibility, and good stability. The sensor has a simple calibration method which requires no action by the user, and utilizes a sophisticated, yet lightweight, model in order to predict temperature changes on the sensor during breathing and track changes in water content. It is shown to be effective for measuring CO2 waveform parameters on a breath-by-breath basis, such as End-Tidal CO2, Alveolar Plateau Slope, and Beginning Exhalation Slope.
ContributorsBridgeman, Devon (Author) / Forzani, Erica S (Thesis advisor) / Nikkhah, Mehdi (Committee member) / Holloway, Julianne (Committee member) / Raupp, Gregory (Committee member) / Emady, Heather (Committee member) / Arizona State University (Publisher)
Created2017
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 study genome-wide methylome profiles of the cells require a long

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.
ContributorsRidha, Inam (Author) / LaBaer, Joshua (Thesis advisor) / Murugan, Vel (Thesis advisor) / Plaisier, Christopher (Committee member) / Nikkhah, Mehdi (Committee member) / Vernon, Brent (Committee member) / Arizona State University (Publisher)
Created2022
157806-Thumbnail Image.png
Description
The WNT signaling pathway plays numerous roles in development and maintenance of adult homeostasis. In concordance with it’s numerous roles, dysfunction of WNT signaling leads to a variety of human diseases ranging from developmental disorders to cancer. WNT signaling is composed of a family of 19 WNT soluble secreted glycoproteins,

The WNT signaling pathway plays numerous roles in development and maintenance of adult homeostasis. In concordance with it’s numerous roles, dysfunction of WNT signaling leads to a variety of human diseases ranging from developmental disorders to cancer. WNT signaling is composed of a family of 19 WNT soluble secreted glycoproteins, which are evolutionarily conserved across all phyla of the animal kingdom. WNT ligands interact most commonly with a family of receptors known as frizzled (FZ) receptors, composed of 10 independent genes. Specific interactions between WNT proteins and FZ receptors are not well characterized and are known to be promiscuous, Traditionally canonical WNT signaling is described as a binary system in which WNT signaling is either off or on. In the ‘off’ state, in the absence of a WNT ligand, cytoplasmic β-catenin is continuously degraded by the action of the APC/Axin/GSK-3β destruction complex. In the ‘on’ state, when WNT binds to its Frizzled (Fz) receptor and LRP coreceptor, this protein destruction complex is disrupted, allowing β-catenin to translocate into the nucleus where it interacts with the DNA-bound T cell factor/lymphoid factor (TCF/LEF) family of proteins to regulate target gene expression. However in a variety of systems in development and disease canonical WNT signaling acts in a gradient fashion, suggesting more complex regulation of β-catenin transcriptional activity. As such, the traditional ‘binary’ view of WNT signaling does not clearly explain how this graded signal is transmitted intracellularly to control concentration-dependent changes in gene expression and cell identity. I have developed an in vitro human pluripotent stem cell (hPSC)-based model that recapitulates the same in vivo developmental effects of the WNT signaling gradient on the anterior-posterior (A/P) patterning of the neural tube observed during early development. Using RNA-seq and ChIP-seq I have characterized β-catenin binding at different levels of WNT signaling and identified different classes of β-catenin peaks that bind cis-regulatory elements to influence neural cell fate. This work expands the traditional binary view of canonical WNT signaling and illuminates WNT/β-catenin activity in other developmental and diseased contexts.
ContributorsCutts, Joshua Patrick (Author) / Brafman, David A (Thesis advisor) / Stabenfeldt, Sarah (Committee member) / Nikkhah, Mehdi (Committee member) / Wang, Xiao (Committee member) / Plaisier, Christopher (Committee member) / Arizona State University (Publisher)
Created2019
Description
According to the World Health Organization, cancer is one of the leading causes of death around the world. Although early diagnostics using biomarkers and improved treatments with targeted therapy have reduced the rate of cancer related mortalities, there remain many unknowns regarding the contributions of the tumor microenvironment to cancer

According to the World Health Organization, cancer is one of the leading causes of death around the world. Although early diagnostics using biomarkers and improved treatments with targeted therapy have reduced the rate of cancer related mortalities, there remain many unknowns regarding the contributions of the tumor microenvironment to cancer progression and therapeutic resistance. The tumor microenvironment plays a significant role by manipulating the progression of cancer cells through biochemical and biophysical signals from the surrounding stromal cells along with the extracellular matrix. As such, there is a critical need to understand how the tumor microenvironment influences the molecular mechanisms underlying cancer metastasis to facilitate the discovery of better therapies. This thesis described the development of microfluidic technologies to study the interplay of cancer cells with their surrounding microenvironment. The microfluidic model was used to assess how exposure to chemoattractant, epidermal growth factor (EGF), impacted 3D breast cancer cell invasion and enhanced cell motility speed was noted in the presence of EGF validating physiological cell behavior. Additionally, breast cancer and patient-derived cancer-associated fibroblast (CAF) cells were co-cultured to study cell-cell crosstalk and how it affected cancer invasion. GPNMB was identified as a novel gene of interest and it was shown that CAFs enhanced breast cancer invasion by up-regulating the expression of GPNMB on breast cancer cells resulting in increased migration speed. Lastly, this thesis described the design, biological validation, and use of this microfluidic platform as a new in vitro 3D organotypic model to study mechanisms of glioma stem cell (GSC) invasion in the context of a vascular niche. It was confirmed that CXCL12-CXCR4 signaling is involved in promoting GSC invasion in a 3D vascular microenvironment, while also demonstrating the effectiveness of the microfluidic as a drug screening assay. Taken together, the broader impacts of the microfluidic model developed in this dissertation include, a possible alternative platform to animal testing that is focused on mimicking human physiology, a potential ex vivo platform using patient-derived cells for studying the interplay of cancer cells with its surrounding microenvironment, and development of future therapeutic strategies tailored toward disrupting key molecular pathways involved in regulatory mechanisms of cancer invasion.
ContributorsTruong, Danh, Ph.D (Author) / Nikkhah, Mehdi (Thesis advisor) / LaBaer, Joshua (Committee member) / Smith, Barbara (Committee member) / Mouneimne, Ghassan (Committee member) / Vernon, Brent (Committee member) / Arizona State University (Publisher)
Created2018
157729-Thumbnail Image.png
Description
Extracellular Vesicles (EVs), particularly exosomes, are of considerable interest as tumor biomarkers since tumor-derived EVs contain a broad array of information about tumor pathophysiology including its metabolic and metastatic status. However, current EV based assays cannot distinguish between EV biomarker changes by altered secretion of EVs during diseased conditions like

Extracellular Vesicles (EVs), particularly exosomes, are of considerable interest as tumor biomarkers since tumor-derived EVs contain a broad array of information about tumor pathophysiology including its metabolic and metastatic status. However, current EV based assays cannot distinguish between EV biomarker changes by altered secretion of EVs during diseased conditions like cancer, inflammation, etc. that express a constant level of a given biomarker, stable secretion of EVs with altered biomarker expression, or a combination of these two factors. This issue was addressed by developing a nanoparticle and dye-based fluorescent immunoassay that can distinguish among these possibilities by normalizing EV biomarker level(s) to EV abundance, revealing average expression levels of EV biomarker under observation. In this approach, EVs are captured from complex samples (e.g. serum), stained with a lipophilic dye and hybridized with antibody-conjugated quantum dot probes for specific EV surface biomarkers. EV dye signal is used to quantify EV abundance and normalize EV surface biomarker expression levels. EVs from malignant (PANC-1) and nonmalignant pancreatic cell lines (HPNE) exhibited similar staining, and probe-to-dye ratios did not change with EV abundance, allowing direct analysis of normalized EV biomarker expression without a separate EV quantification step. This EV biomarker normalization approach markedly improved the ability of serum levels of two pancreatic cancer biomarkers, EV EpCAM, and EV EphA2, to discriminate pancreatic cancer patients from nonmalignant control subjects. The streamlined workflow and robust results of this assay are suitable for rapid translation to clinical applications and its flexible design permits it to be rapidly adapted to quantitate other EV biomarkers by the simple swapping of the antibody-conjugated quantum dot probes for those that recognize a different disease-specific EV biomarker utilizing a workflow that is suitable for rapid clinical translation.
ContributorsRodrigues, Meryl (Author) / Hu, Tony (Thesis advisor) / Nikkhah, Mehdi (Committee member) / Kiani, Samira (Committee member) / Smith, Barbara (Committee member) / Han, Haiyong (Committee member) / Arizona State University (Publisher)
Created2019