scholarly journals Single Cell Gene Transcriptome Analysis of Ovarian Mature Teratomas

2021 ◽  
Vol 27 ◽  
Author(s):  
Sun Shin ◽  
Youn Jin Choi ◽  
Seung-Hyun Jung ◽  
Yeun-Jun Chung ◽  
Sug Hyung Lee

Teratoma is a type of germ cell tumor that originates from totipotential germ cells that are present in gonads, which can differentiate into any of the cell types found in adult tissues. Ovarian teratomas are usually mature cystic teratomas (OMCTs, also known as dermoid cysts). Chromosome studies in OMCTs show that the chromosomes are uniformly homozygous with karyotype of 46, XX, indicating that they may be parthenogenic tumors that arise from a single ovum after thefirst meiotic division. However, the tissues in OMCTs have been known to be morphologically and immunophenotypically identical to the orthotopic tissues. Currently, expression profiles of tissue components in OMCTs are not known. To identify whether OMCT tissues are expressionally similar to or different from the orthotopic tissues, we adopted single-cell RNA-sequencing (scRNA-seq), and analyzed transcriptomes of individual cells in heterogenous tissues of two OMCTs. We found that transcriptome profiles of the OMCTs at single cell level were not significantly different from those of normal cells in orthotopic locations. The present data suggest that parthenogeneticlly altered OMCTs may not alter expression profiles of inrivirual tissue components in OMCTs.

2021 ◽  
Author(s):  
Qiang Li ◽  
Zuwan Lin ◽  
Ren Liu ◽  
Xin Tang ◽  
Jiahao Huang ◽  
...  

AbstractPairwise mapping of single-cell gene expression and electrophysiology in intact three-dimensional (3D) tissues is crucial for studying electrogenic organs (e.g., brain and heart)1–5. Here, we introducein situelectro-sequencing (electro-seq), combining soft bioelectronics within situRNA sequencing to stably map millisecond-timescale cellular electrophysiology and simultaneously profile a large number of genes at single-cell level across 3D tissues. We appliedin situelectro-seq to 3D human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) patches, precisely registering the CM gene expression with electrophysiology at single-cell level, enabling multimodalin situanalysis. Such multimodal data integration substantially improved the dissection of cell types and the reconstruction of developmental trajectory from spatially heterogeneous tissues. Using machine learning (ML)-based cross-modal analysis,in situelectro-seq identified the gene-to-electrophysiology relationship over the time course of cardiac maturation. Further leveraging such a relationship to train a coupled autoencoder, we demonstrated the prediction of single-cell gene expression profile evolution using long-term electrical measurement from the same cardiac patch or 3D millimeter-scale cardiac organoids. As exemplified by cardiac tissue maturation,in situelectro-seq will be broadly applicable to create spatiotemporal multimodal maps and predictive models in electrogenic organs, allowing discovery of cell types and gene programs responsible for electrophysiological function and dysfunction.


GigaScience ◽  
2020 ◽  
Vol 9 (11) ◽  
Author(s):  
Fatemeh Behjati Ardakani ◽  
Kathrin Kattler ◽  
Tobias Heinen ◽  
Florian Schmidt ◽  
David Feuerborn ◽  
...  

Abstract Background Single-cell RNA sequencing is a powerful technology to discover new cell types and study biological processes in complex biological samples. A current challenge is to predict transcription factor (TF) regulation from single-cell RNA data. Results Here, we propose a novel approach for predicting gene expression at the single-cell level using cis-regulatory motifs, as well as epigenetic features. We designed a tree-guided multi-task learning framework that considers each cell as a task. Through this framework we were able to explain the single-cell gene expression values using either TF binding affinities or TF ChIP-seq data measured at specific genomic regions. TFs identified using these models could be validated by the literature. Conclusion Our proposed method allows us to identify distinct TFs that show cell type–specific regulation. This approach is not limited to TFs but can use any type of data that can potentially be used in explaining gene expression at the single-cell level to study factors that drive differentiation or show abnormal regulation in disease. The implementation of our workflow can be accessed under an MIT license via https://github.com/SchulzLab/Triangulate.


2017 ◽  
Author(s):  
Wenfa Ng

Single cell studies increasing reveal myriad cellular subtypes beyond those postulated or observed through optical and fluorescence microscopy as well as DNA sequencing studies. While gene sequencing at the single cell level offer a path towards illuminating, in totality, the different subtypes of cells present, the technique nevertheless does not offer answers concerning the functional repertoire of the cell, which is defined by the collection of RNA transcribed from the genome. Known as the transcriptome, transcribed RNA defines the function of the cell as proteins or effector RNA molecules, while the genome is the collection of all information endowed in the cell type, expressed or not. Thus, a particular cell state, lineage, cell fate or cellular differentiation is more fully depicted by transcriptomic analysis compared to delineating the genomic context at the single cell level. While conceptually sound and could be analysed by contemporary single cell RNA sequencing technology and data analysis pipelines, the relative instability of RNA in view of RNase in the environment would make sample preparation particularly challenging, where degradation of cellular RNA by extraneous factors could provide a misinterpretation of specific functions available to a cell type. Hence, RNA as the de facto functional molecule of the cell defining the proteomics landscape as well as effector RNA repertoire, meant that RNA transcriptomics at the single cell level is the way forward if the goal is to understand all available cell types, lineage, cell fate and cellular differentiation. Given that a cell state is defined by the functions encoded by functional molecules such as proteins and RNA, single cell RNA sequencing offers a larger contextual basis for understanding cellular decision making and functions, for example, proteins are increasingly known to work in concert with RNA effector molecules in enabling a function. Hence, providing a view of the diverse cell types and lineages present in a body, single cell RNA sequencing is only hampered by the high sensitivity required to analyse the small amount of RNA available in single cells, as well as the perennial problem of RNA studies: how to prevent or reduce RNA degradation by environmental RNase enzymes. Ability to reduce RNA degradation would provide the cell biologist a unique view of the functional landscape of different cells in the body through the language of RNA.


2017 ◽  
Author(s):  
Wenfa Ng

Single cell studies increasing reveal myriad cellular subtypes beyond those postulated or observed through optical and fluorescence microscopy as well as DNA sequencing studies. While gene sequencing at the single cell level offer a path towards illuminating, in totality, the different subtypes of cells present, the technique nevertheless does not offer answers concerning the functional repertoire of the cell, which is defined by the collection of RNA transcribed from the genome. Known as the transcriptome, transcribed RNA defines the function of the cell as proteins or effector RNA molecules, while the genome is the collection of all information endowed in the cell type, expressed or not. Thus, a particular cell state, lineage, cell fate or cellular differentiation is more fully depicted by transcriptomic analysis compared to delineating the genomic context at the single cell level. While conceptually sound and could be analysed by contemporary single cell RNA sequencing technology and data analysis pipelines, the relative instability of RNA in view of RNase in the environment would make sample preparation particularly challenging, where degradation of cellular RNA by extraneous factors could provide a misinterpretation of specific functions available to a cell type. Hence, RNA as the de facto functional molecule of the cell defining the proteomics landscape as well as effector RNA repertoire, meant that RNA transcriptomics at the single cell level is the way forward if the goal is to understand all available cell types, lineage, cell fate and cellular differentiation. Given that a cell state is defined by the functions encoded by functional molecules such as proteins and RNA, single cell RNA sequencing offers a larger contextual basis for understanding cellular decision making and functions, for example, proteins are increasingly known to work in concert with RNA effector molecules in enabling a function. Hence, providing a view of the diverse cell types and lineages present in a body, single cell RNA sequencing is only hampered by the high sensitivity required to analyse the small amount of RNA available in single cells, as well as the perennial problem of RNA studies: how to prevent or reduce RNA degradation by environmental RNase enzymes. Ability to reduce RNA degradation would provide the cell biologist a unique view of the functional landscape of different cells in the body through the language of RNA.


Author(s):  
Zilong Zhang ◽  
Feifei Cui ◽  
Chen Lin ◽  
Lingling Zhao ◽  
Chunyu Wang ◽  
...  

Abstract Single-cell RNA sequencing (scRNA-seq) has enabled us to study biological questions at the single-cell level. Currently, many analysis tools are available to better utilize these relatively noisy data. In this review, we summarize the most widely used methods for critical downstream analysis steps (i.e. clustering, trajectory inference, cell-type annotation and integrating datasets). The advantages and limitations are comprehensively discussed, and we provide suggestions for choosing proper methods in different situations. We hope this paper will be useful for scRNA-seq data analysts and bioinformatics tool developers.


2021 ◽  
Author(s):  
Stella Belonwu ◽  
Yaqiao Li ◽  
Daniel Bunis ◽  
Arjun Arkal Rao ◽  
Caroline Warly Solsberg ◽  
...  

Abstract Alzheimer’s Disease (AD) is a complex neurodegenerative disease that gravely affects patients and imposes an immense burden on caregivers. Apolipoprotein E4 (APOE4) has been identified as the most common genetic risk factor for AD, yet the molecular mechanisms connecting APOE4 to AD are not well understood. Past transcriptomic analyses in AD have revealed APOE genotype-specific transcriptomic differences; however, these differences have not been explored at a single-cell level. Here, we leverage the first two single-nucleus RNA sequencing AD datasets from human brain samples, including nearly 55,000 cells from the prefrontal and entorhinal cortices. We observed more global transcriptomic changes in APOE4 positive AD cells and identified differences across APOE genotypes primarily in glial cell types. Our findings highlight the differential transcriptomic perturbations of APOE isoforms at a single-cell level in AD pathogenesis and have implications for precision medicine development in the diagnosis and treatment of AD.


Cephalalgia ◽  
2018 ◽  
Vol 38 (13) ◽  
pp. 1976-1983 ◽  
Author(s):  
William Renthal

Background Migraine is a debilitating disorder characterized by severe headaches and associated neurological symptoms. A key challenge to understanding migraine has been the cellular complexity of the human brain and the multiple cell types implicated in its pathophysiology. The present study leverages recent advances in single-cell transcriptomics to localize the specific human brain cell types in which putative migraine susceptibility genes are expressed. Methods The cell-type specific expression of both familial and common migraine-associated genes was determined bioinformatically using data from 2,039 individual human brain cells across two published single-cell RNA sequencing datasets. Enrichment of migraine-associated genes was determined for each brain cell type. Results Analysis of single-brain cell RNA sequencing data from five major subtypes of cells in the human cortex (neurons, oligodendrocytes, astrocytes, microglia, and endothelial cells) indicates that over 40% of known migraine-associated genes are enriched in the expression profiles of a specific brain cell type. Further analysis of neuronal migraine-associated genes demonstrated that approximately 70% were significantly enriched in inhibitory neurons and 30% in excitatory neurons. Conclusions This study takes the next step in understanding the human brain cell types in which putative migraine susceptibility genes are expressed. Both familial and common migraine may arise from dysfunction of discrete cell types within the neurovascular unit, and localization of the affected cell type(s) in an individual patient may provide insight into to their susceptibility to migraine.


2017 ◽  
Author(s):  
Shilo Rosenwasser ◽  
Miguel J. Frada ◽  
David Pilzer ◽  
Ron Rotkopf ◽  
Assaf Vardi

AbstractMarine viruses are major evolutionary and biogeochemical drivers of microbial life in the ocean. Host response to viral infection typically includes virus-induced rewiring of metabolic network to supply essential building blocks for viral assembly, as opposed to activation of anti-viral host defense. Nevertheless, there is a major bottleneck to accurately discern between viral hijacking strategies and host defense responses when averaging bulk population response. Here we use Emiliania huxleyi, a bloom-forming alga and its specific virus (EhV), as one of the most ecologically important host-virus model system in the ocean. Using automatic microfluidic setup to capture individual algal cells, we quantified host and virus gene expression on a single-cell resolution during the course of infection. We revealed high heterogeneity in viral gene expression among individual cells. Simultaneous measurements of expression profiles of host and virus genes at a single-cell level allowed mapping of infected cells into newly defined infection states and uncover a yet unrecognized early phase in host response that occurs prior to viral expression. Intriguingly, resistant cells emerged during viral infection, showed unique expression profiles of metabolic genes which can provide the basis for discerning between viral resistant and sensitive cells within heterogeneous populations in the marine environment. We propose that resolving host-virus arms race at a single-cell level will provide important mechanistic insights into viral life cycles and will uncover host defense strategies.


2019 ◽  
Vol 15 (4) ◽  
pp. e1007708 ◽  
Author(s):  
Shilo Rosenwasser ◽  
Uri Sheyn ◽  
Miguel J. Frada ◽  
David Pilzer ◽  
Ron Rotkopf ◽  
...  

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 10539-10539 ◽  
Author(s):  
Yu-Chieh Wang ◽  
Daniel Ramskold ◽  
Shujun Luo ◽  
Robin Li ◽  
Qiaolin Deng ◽  
...  

10539 Background: Melanoma is the most aggressive type of skin cancer. Late-stage melanoma is highly metastatic and currently lacks effective treatment. This discouraging clinical observation highlights the need for a better understanding of the molecular mechanisms underlying melanoma initiation and progression and for developing new therapeutic approaches based on novel targets. Although genome-wide transcriptome analyses have been frequently used to study molecular alterations in clinical samples, it has been technically challenging to obtain the transcriptomic profiles at single-cell level. Methods: Using antibody-mediated magnetic activated cell separation (MACS), we isolated and individualized putative circulating melanoma cells (CMCs) from the blood samples of the melanoma patients at advance stages. The transcriptomic analysis based on a novel and robust mRNA-Seq protocol (Smart-Seq) was established and applied to the putative CMCs for single-cell profiling. Results: We have discovered distinct gene expression patterns, including new putative markers for CMCs. Meanwhile, the gene expression profiles derived of the CMC candidates isolated from the patient’s blood samples are closely-related to the expression profiles of other cells originated from human melanocytes, including normal melanocytes in primary culture and melanoma cell lines. Compared with existing methods, Smart-Seq has improved read coverage across transcripts, which provides advantage for better analyzing transcript isoforms and SNPs. Conclusions: Our results suggest that the techniques developed in this research for cell isolation and transcriptomic analyses can potentially be used for addressing many biological and clinical questions requiring genomewide transcriptome profiling in rare cells.


Sign in / Sign up

Export Citation Format

Share Document