Matching Items (50)
155356-Thumbnail Image.png
Description
The past decade has seen a drastic increase in collaboration between Computer Science (CS) and Molecular Biology (MB). Current foci in CS such as deep learning require very large amounts of data, and MB research can often be rapidly advanced by analysis and models from CS. One of the places

The past decade has seen a drastic increase in collaboration between Computer Science (CS) and Molecular Biology (MB). Current foci in CS such as deep learning require very large amounts of data, and MB research can often be rapidly advanced by analysis and models from CS. One of the places where CS could aid MB is during analysis of sequences to find binding sites, prediction of folding patterns of proteins. Maintenance and replication of stem-like cells is possible for long terms as well as differentiation of these cells into various tissue types. These behaviors are possible by controlling the expression of specific genes. These genes then cascade into a network effect by either promoting or repressing downstream gene expression. The expression level of all gene transcripts within a single cell can be analyzed using single cell RNA sequencing (scRNA-seq). A significant portion of noise in scRNA-seq data are results of extrinsic factors and could only be removed by customized scRNA-seq analysis pipeline. scRNA-seq experiments utilize next-gen sequencing to measure genome scale gene expression levels with single cell resolution.

Almost every step during analysis and quantification requires the use of an often empirically determined threshold, which makes quantification of noise less accurate. In addition, each research group often develops their own data analysis pipeline making it impossible to compare data from different groups. To remedy this problem a streamlined and standardized scRNA-seq data analysis and normalization protocol was designed and developed. After analyzing multiple experiments we identified the possible pipeline stages, and tools needed. Our pipeline is capable of handling data with adapters and barcodes, which was not the case with pipelines from some experiments. Our pipeline can be used to analyze single experiment scRNA-seq data and also to compare scRNA-seq data across experiments. Various processes like data gathering, file conversion, and data merging were automated in the pipeline. The main focus was to standardize and normalize single-cell RNA-seq data to minimize technical noise introduced by disparate platforms.
ContributorsBalachandran, Parithi (Author) / Wang, Xiao (Thesis advisor) / Brafman, David (Committee member) / Lockhart, Thurmon (Committee member) / Arizona State University (Publisher)
Created2017
155320-Thumbnail Image.png
Description
Alzheimer’s disease (AD), despite over a century of research, does not have a clearly defined pathogenesis for the sporadic form that makes up the majority of disease incidence. A variety of correlative risk factors have been identified, including the three isoforms of apolipoprotein E (ApoE), a cholesterol transport protein in

Alzheimer’s disease (AD), despite over a century of research, does not have a clearly defined pathogenesis for the sporadic form that makes up the majority of disease incidence. A variety of correlative risk factors have been identified, including the three isoforms of apolipoprotein E (ApoE), a cholesterol transport protein in the central nervous system. ApoE ε3 is the wild-type variant with no effect on risk. ApoE ε2, the protective and most rare variant, reduces risk of developing AD by 40%. ApoE ε4, the risk variant, increases risk by 3.2-fold and 14.9-fold for heterozygous and homozygous representation respectively. Study of these isoforms has been historically complex, but the advent of human induced pluripotent stem cells (hiPSC) provides the means for highly controlled, longitudinal in vitro study. The effect of ApoE variants can be further elucidated using this platform by generating isogenic hiPSC lines through precise genetic modification, the objective of this research. As the difference between alleles is determined by two cytosine-thymine polymorphisms, a specialized CRISPR/Cas9 system for direct base conversion was able to be successfully employed. The base conversion method for transitioning from the ε3 to ε2 allele was first verified using the HEK293 cell line as a model with delivery via electroporation. Following this verification, the transfection method was optimized using two hiPSC lines derived from ε4/ε4 patients, with a lipofection technique ultimately resulting in successful base conversion at the same site verified in the HEK293 model. Additional research performed included characterization of the pre-modification genotype with respect to likely off-target sites and methods of isolating clonal variants.
ContributorsLakers, Mary Frances (Author) / Brafman, David (Thesis advisor) / Haynes, Karmella (Committee member) / Wang, Xiao (Committee member) / Arizona State University (Publisher)
Created2017
158125-Thumbnail Image.png
Description
Alzheimer’s disease (AD) affects over 5 million individuals each year in the United States. Furthermore, most cases of AD are sporadic, making it extremely difficult to model and study in vitro. CRISPR/Cas9 and base editing technologies have been of recent interest because of their ability to create single nucleotide edits

Alzheimer’s disease (AD) affects over 5 million individuals each year in the United States. Furthermore, most cases of AD are sporadic, making it extremely difficult to model and study in vitro. CRISPR/Cas9 and base editing technologies have been of recent interest because of their ability to create single nucleotide edits at nearly any genomic sequence using a Cas9 protein and a guide RNA (sgRNA). Currently, there is no available phenotype to differentiate edited cells from unedited cells. Past research has employed fluorescent proteins bound to Cas9 proteins to attempt to enrich for edited cells, however, these methods are only reporters of transfection (RoT) and are no indicative of actual base-editing occurring. Thus, this study proposes a transient reporter for editing enrichment (TREE) and Cas9-mediated adenosine TREE (CasMasTREE) which use plasmids to co-transfect with CRISPR/Cas9 technologies to serve as an indicator of base-editing. Specifically, TREE features a blue fluorescent protein (BFP) mutant that, upon a C-T conversion, changes the emission spectrum to a green fluorescent protein (GFP). CasMasTREE features a mCherry and GFP protein separated by a stop codon which can be negated using an A-G conversion. By employing a sgRNA that targets one of the TREE plasmids and at least one genomic site, cells can be sorted for GFP(+) cells. Using these methods, base-edited isogenic hiPSC line generation using TREE (BIG-TREE) was created to generate isogenic hiPSC lines with AD-relevant edits. For example, BIG-TREE demonstrates the capability of converting Apolipoprotein E (APOE), a gene associated with AD-risk development, wildtype (3/3) into another isoform, APOE2/2, to create isogenic hiPSC lines. The capabilities of TREE are vast and can be applied to generate various models of diseases with specific genomic edits.
ContributorsNguyen, Toan Thai Tran (Author) / Brafman, David (Thesis advisor) / Wang, Xiao (Committee member) / Tian, Xiaojun (Committee member) / Arizona State University (Publisher)
Created2020
158747-Thumbnail Image.png
Description
Gene circuit engineering facilitates the discovery and understanding of fundamental biology and has been widely used in various biological applications. In synthetic biology, gene circuits are often constructed by two main strategies: either monocistronic or polycistronic constructions. The Latter architecture can be commonly found in prokaryotes, eukaryotes, and viruses and

Gene circuit engineering facilitates the discovery and understanding of fundamental biology and has been widely used in various biological applications. In synthetic biology, gene circuits are often constructed by two main strategies: either monocistronic or polycistronic constructions. The Latter architecture can be commonly found in prokaryotes, eukaryotes, and viruses and has been largely applied in gene circuit engineering. In this work, the effect of adjacent genes and noncoding regions are systematically investigated through the construction of batteries of gene circuits in diverse scenarios. Data-driven analysis yields a protein expression metric that strongly correlates with the features of adjacent transcriptional regions (ATRs). This novel mathematical tool helps the guide for circuit construction and has the implication for the design of synthetic ATRs to tune gene expression, illustrating its potential to facilitate engineering complex gene networks. The ability to tune RNA dynamics is greatly needed for biotech applications, including therapeutics and diagnostics. Diverse methods have been developed to tune gene expression through transcriptional or translational manipulation. Control of RNA stability/degradation is often overlooked and can be the lightweight alternative to regulate protein yields. To further extend the utility of engineered ATRs to regulate gene expression, a library of RNA modules named degradation-tuning RNAs (dtRNAs) are designed with the ability to form specific 5’ secondary structures prior to RBS. These modules can modulate transcript stability while having a minimal interference on translation initiation. Optimization of their functional structural features enables gene expression level to be tuned over a wide dynamic range. These engineered dtRNAs are capable of regulating gene circuit dynamics as well as noncoding RNA levels and can be further expanded into cell-free system for gene expression control in vitro. Finally, integrating dtRNA with synthetic toehold sensor enables improved paper-based viral diagnostics, illustrating the potential of using synthetic dtRNAs for biomedical applications.
ContributorsZhang, Qi (Author) / Wang, Xiao (Thesis advisor) / Green, Alexander (Committee member) / Brafman, David (Committee member) / Tian, Xiaojun (Committee member) / Plaisier, Christopher (Committee member) / Arizona State University (Publisher)
Created2020
158301-Thumbnail Image.png
Description
The human transcriptional regulatory machine utilizes hundreds of transcription factors which bind to specific genic sites resulting in either activation or repression of targeted genes. Networks comprised of nodes and edges can be constructed to model the relationships of regulators and their targets. Within these biological networks small enriched structural

The human transcriptional regulatory machine utilizes hundreds of transcription factors which bind to specific genic sites resulting in either activation or repression of targeted genes. Networks comprised of nodes and edges can be constructed to model the relationships of regulators and their targets. Within these biological networks small enriched structural patterns containing at least three nodes can be identified as potential building blocks from which a network is organized. A first iteration computational pipeline was designed to generate a disease specific gene regulatory network for motif detection using established computational tools. The first goal was to identify motifs that can express themselves in a state that results in differential patient survival in one of the 32 different cancer types studied. This study identified issues for detecting strongly correlated motifs that also effect patient survival, yielding preliminary results for possible driving cancer etiology. Second, a comparison was performed for the topology of network motifs across multiple different data types to identify possible divergence from a conserved enrichment pattern in network perturbing diseases. The topology of enriched motifs across all the datasets converged upon a single conserved pattern reported in a previous study which did not appear to diverge dependent upon the type of disease. This report highlights possible methods to improve detection of disease driving motifs that can aid in identifying possible treatment targets in cancer. Finally, networks where only minimally perturbed, suggesting that regulatory programs were run from evolved circuits into a cancer context.
ContributorsStriker, Shawn Scott (Author) / Plaisier, Christopher (Thesis advisor) / Brafman, David (Committee member) / Wang, Xiao (Committee member) / Arizona State University (Publisher)
Created2020
161295-Thumbnail Image.png
Description
Genome wide association studies (GWAS) have identified polymorphism in the Apolipoprotein E (APOE) gene to be the most prominent risk factor for Alzheimer’s disease (AD). Compared to individuals homozygous for the APOE3 variant, individuals with the APOE4 variant have a significantly elevated risk of AD. On the other hand, longitudinal

Genome wide association studies (GWAS) have identified polymorphism in the Apolipoprotein E (APOE) gene to be the most prominent risk factor for Alzheimer’s disease (AD). Compared to individuals homozygous for the APOE3 variant, individuals with the APOE4 variant have a significantly elevated risk of AD. On the other hand, longitudinal studies have shown that the presence of the APOE2 variant reduces lifetime risk of developing AD by 40 percent. While there has been significant research that has identified the risk-inducing effects of APOE4, the underlying mechanisms by which APOE2 influences AD onset and progression have not been extensively explored. The hallmarks of AD pathology manifest in human neurons in the form of extracellular amyloid deposits and intracellular neurofibrillary tangles, whereas astrocytes are the primary source of the APOE protein in the brain. In this study, an isogenic human induced pluripotent stem cell (hiPSC)-based system is utilized to demonstrate that conversion of APOE3 to APOE2 greatly reduced the production of amyloid-beta (Aβ) peptides in hiPSC-derived neural cultures. Mechanistically, analysis of pure populations of neurons and astrocytes derived from these neural cultures revealed that mitigating effects of APOE2 is mediated by cell autonomous and non-autonomous effects. In particular, it was demonstrated the reduction in Aβ and pathogenic β-C-terminal fragments (APP-βCTF) is potentially driven by a mechanism related to non-amyloidogenic processing of amyloid precursor protein (APP), suggesting a gain of protective function of the APOE2 variant. Together, this study provides insights into the risk-modifying effects associated with the APOE2 allele and establishes a platform to probe the mechanisms by which APOE2 enhances neuroprotection against AD.
ContributorsRaman, Sreedevi (Author) / Brafman, David (Thesis advisor) / Smith, Barbara (Committee member) / Plaiser, Christopher (Committee member) / Wang, Xiao (Committee member) / Tian, Xiaojun (Committee member) / Arizona State University (Publisher)
Created2021
Description

Neural progenitor cells (NPCs) derived from human pluripotent stem cells (hPSCs) are a multipotent cell population that is capable of nearly indefinite expansion and subsequent differentiation into the various neuronal and supporting cell types that comprise the CNS. However, current protocols for differentiating NPCs toward neuronal lineages result in a

Neural progenitor cells (NPCs) derived from human pluripotent stem cells (hPSCs) are a multipotent cell population that is capable of nearly indefinite expansion and subsequent differentiation into the various neuronal and supporting cell types that comprise the CNS. However, current protocols for differentiating NPCs toward neuronal lineages result in a mixture of neurons from various regions of the CNS. In this study, we determined that endogenous WNT signaling is a primary contributor to the heterogeneity observed in NPC cultures and neuronal differentiation. Furthermore, exogenous manipulation of WNT signaling during neural differentiation, through either activation or inhibition, reduces this heterogeneity in NPC cultures, thereby promoting the formation of regionally homogeneous NPC and neuronal cultures. The ability to manipulate WNT signaling to generate regionally specific NPCs and neurons will be useful for studying human neural development and will greatly enhance the translational potential of hPSCs for neural-related therapies.

ContributorsMoya, Noel (Author) / Cutts, Joshua (Author) / Gaasterland, Terry (Author) / Willert, Karl (Author) / Brafman, David (Author) / Ira A. Fulton Schools of Engineering (Contributor)
Created2014-12-09
128416-Thumbnail Image.png
Description

Adult and pluripotent stem cells represent a ready supply of cellular raw materials that can be used to generate the functionally mature cells needed to replace damaged or diseased heart tissue. However, the use of stem cells for cardiac regenerative therapies is limited by the low efficiency by which stem

Adult and pluripotent stem cells represent a ready supply of cellular raw materials that can be used to generate the functionally mature cells needed to replace damaged or diseased heart tissue. However, the use of stem cells for cardiac regenerative therapies is limited by the low efficiency by which stem cells are differentiated in vitro to cardiac lineages as well as the inability to effectively deliver stem cells and their derivatives to regions of damaged myocardium. In this review, we discuss the various biomaterial-based approaches that are being implemented to direct stem cell fate both in vitro and in vivo. First, we discuss the stem cell types available for cardiac repair and the engineering of naturally and synthetically derived biomaterials to direct their in vitro differentiation to the cell types that comprise heart tissue. Next, we describe biomaterial-based approaches that are being implemented to enhance the in vivo integration and differentiation of stem cells delivered to areas of cardiac damage. Finally, we present emerging trends of using stem cell-based biomaterial approaches to deliver pro-survival factors and fully vascularized tissue to the damaged and diseased cardiac tissue.

ContributorsCutts, Joshua (Author) / Nikkhah, Mehdi (Author) / Brafman, David (Author) / Ira A. Fulton Schools of Engineering (Contributor)
Created2015-06-01
127946-Thumbnail Image.png
Description

Due to the limitation of current pharmacological therapeutic strategies, stem cell therapies have emerged as a viable option for treating many incurable neurological disorders. Specifically, human pluripotent stem cell (hPSC)-derived neural progenitor cells (hNPCs), a multipotent cell population that is capable of near indefinite expansion and subsequent differentiation into the

Due to the limitation of current pharmacological therapeutic strategies, stem cell therapies have emerged as a viable option for treating many incurable neurological disorders. Specifically, human pluripotent stem cell (hPSC)-derived neural progenitor cells (hNPCs), a multipotent cell population that is capable of near indefinite expansion and subsequent differentiation into the various cell types that comprise the central nervous system (CNS), could provide an unlimited source of cells for such cell-based therapies. However the clinical application of these cells will require (i) defined, xeno-free conditions for their expansion and neuronal differentiation and (ii) scalable culture systems that enable their expansion and neuronal differentiation in numbers sufficient for regenerative medicine and drug screening purposes. Current extracellular matrix protein (ECMP)-based substrates for the culture of hNPCs are expensive, difficult to isolate, subject to batch-to-batch variations, and, therefore, unsuitable for clinical application of hNPCs. Using a high-throughput array-based screening approach, we identified a synthetic polymer, poly(4-vinyl phenol) (P4VP), that supported the long-term proliferation and self-renewal of hNPCs. The hNPCs cultured on P4VP maintained their characteristic morphology, expressed high levels of markers of multipotency, and retained their ability to differentiate into neurons. Such chemically defined substrates will eliminate critical roadblocks for the utilization of hNPCs for human neural regenerative repair, disease modeling, and drug discovery.

ContributorsTsai, Yihuan (Author) / Cutts, Joshua (Author) / Kimura, Azuma (Author) / Varun, Divya (Author) / Brafman, David (Author) / Ira A. Fulton Schools of Engineering (Contributor)
Created2015-05-13
127902-Thumbnail Image.png
Description

Although the majority of late-onset Alzheimer's disease (AD) patients are labeled sporadic, multiple genetic risk variants have been identified, the most powerful and prevalent of which is the e4 variant of the Apolipoprotein E (APOE) gene. Here, we generated human induced pluripotent stem cell (hiPSC) lines from the peripheral blood

Although the majority of late-onset Alzheimer's disease (AD) patients are labeled sporadic, multiple genetic risk variants have been identified, the most powerful and prevalent of which is the e4 variant of the Apolipoprotein E (APOE) gene. Here, we generated human induced pluripotent stem cell (hiPSC) lines from the peripheral blood mononuclear cells (PBMCs) of a clinically diagnosed AD patient [ASUi003-A] and a non-demented control (NDC) patient [ASUi004-A] homozygous for the APOE4 risk allele. These hiPSCs maintained their original genotype, expressed pluripotency markers, exhibited a normal karyotype, and retained the ability to differentiate into cells representative of the three germ layers.

ContributorsBrookhouser, Nicholas (Author) / Zhang, Ping (Author) / Caselli, Richard (Author) / Kim, Jean J. (Author) / Brafman, David (Author) / Ira A. Fulton Schools of Engineering (Contributor)
Created2017-07-10