Molecular fingerprinting by single cell clone analysis in adverse drug reaction (ADR) assessment

2021 ◽  
Vol 16 ◽  
Author(s):  
Anjan K Banerjee

: Causality assessment for idiosyncratic ADRs mainly relies on epidemiology, signal detection and less often on proven or plausible mechanistic evidence of the drug at a cellular or organ level. Distinct clones of cells can exist within organs of individual patients, some conferring susceptibility to well recognised adverse drug reactions (ADRs). Recent advances in molecular biology has allowed the development of single cell clonal techniques, including single cell RNA sequencing (scRNA-seq) to molecularly fingerprint ADRs and distinguish between distinct clones of cells within organs in individuals, which may confer differing susceptibilities to ADRs. ScRNA-seq permits molecular fingerprinting of some serious ADRs, mainly in the skin, through identification of directly expressed genes (DEG) of interest within specific clones. Overexpressed DEGs provides an opportunity for targeted treatment strategies to be developed. scRNA-seq could be applied to a number of other ADRs involving tissues that can be biopsied/sampled (including skin, liver, kidney, blood, stem cells) as well as providing a molecular basis for rapid screening of potential therapeutic candidates, which may not otherwise be predictable from class of toxicity/organ involvement. . A framework for putative assessment for ADRs using scRNA-seq is proposed as well as speculating on potential regulatory implications for pharmacovigilance and drug development. Molecular fingerprinting of ADRs using scRNA-seq may allow better targeting for enhanced pharmacovigilance and risk minimisation measures for medicines with appropriate benefit risk profiles, although cost-effectiveness and other factors, such as frequency/severity of individual ADRs and population differences will still be relevant.

2010 ◽  
Vol 205 (1) ◽  
pp. 69-78 ◽  
Author(s):  
Maria J Chen ◽  
Pinwen Peter Chiou ◽  
Yu-Hsian Liao ◽  
Chun-Mean Lin ◽  
Thomas T Chen

Five single-cell clone lines (mRTP1B, mRTP1E, mRTP1F, mRTP1K, and mRTP2A) have been developed from adult rainbow trout pituitary glands. These cell lines have been maintained in a CO2-independent medium supplemented with 10% fetal bovine serum (FBS) for more than 150 passages. At about 150 passages, the doubling time of each single-cell clone in a CO2-independent medium supplemented with 10% FBS at 20 °C was 3.6±0.7, 2.8±0.7, 3.2±0.8, 5.5±0.6, and 6.6±0.6 days respectively. Each single-cell clone contains 60±2 chromosomes, which is within the range of the 2N chromosome numbers reported for rainbow trout. Reverse transcription-PCR analysis revealed that in addition to expressing gh, prolactin (prl), and estradiol (E2) receptor α (e2rα or esr1) genes, each single-cell clone line also expressed other pituitary-specific genes such as tsh, gonadotropin 1 (gth-1 or fshb), gonadotropin 2 (gth-2 or lhb), somatolactin (sl or smtl), proopiomelanocortin-B (pomcb), and corticosteroid receptor (cr or nr3c1). Immunocytochemical analysis showed that all the five single-cell clones produced both Gh and Prl. Furthermore, the expression of gh and prl genes in the single-cell clone lines is responsive to induction by E2, dexamethasone, and o,p′-dichlorodiphenyltrichloroethane. All together, these results confirm that each of the single-cell clones was derived from rainbow trout pituitary glands. These single-cell clone lines not only can be used to study factors that regulate the expression of pituitary hormone genes, but can also be developed as a rapid screening system for identifying environmental endocrine disruptors.


Author(s):  
Nattaphong Rattanavirotkul ◽  
Kristina Kirschner ◽  
Tamir Chandra

Abstract Senescence is a cellular stress response triggered by diverse stressors, including oncogene activation, where it serves as a bona-fide tumour suppressor mechanism. Senescence can be transmitted to neighbouring cells, known as paracrine secondary senescence. Secondary senescence was initially described as a paracrine mechanism, but recent evidence suggests a more complex scenario involving juxtacrine communication between cells. In addition, single-cell studies described differences between primary and secondary senescent end-points, which have thus far not been considered functionally distinct. Here we discuss emerging concepts in senescence transmission and heterogeneity in primary and secondary senescence on a cellular and organ level.


Nature ◽  
2019 ◽  
Vol 572 (7767) ◽  
pp. 120-124 ◽  
Author(s):  
T. Yvanka de Soysa ◽  
Sanjeev S. Ranade ◽  
Satoshi Okawa ◽  
Srikanth Ravichandran ◽  
Yu Huang ◽  
...  

2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Bo Li ◽  
Naixia Ren ◽  
Lele Yang ◽  
Junhao Liu ◽  
Qilai Huang

AbstractCRISPR/Cas9 technology has been widely used for targeted genome modification both in vivo and in vitro. However, an effective method for evaluating genome editing efficiency and screening single-cell clones for desired modification is still lacking. Here, we developed this real time PCR method based on the sensitivity of Taq DNA polymerase to nucleotide mismatch at primer 3′ end during initiating DNA replication. Applications to CRISPR gRNAs targeting EMX1, DYRK1A and HOXB13 genes in Lenti-X 293 T cells exhibited comprehensive advantages. Just in one-round qPCR analysis using genomic DNA from cells underwent CRISPR/Cas9 or BE4 treatments, the genome editing efficiency could be determined accurately and quickly, for indel, HDR as well as base editing. When applied to single-cell clone screening, the genotype of each cell colony could also be determined accurately. This method defined a rigorous and practical way in quantify genome editing events.


Stem Cells ◽  
1993 ◽  
Vol 11 (S3) ◽  
pp. 43-47
Author(s):  
Ph. R. Hénon ◽  
J. C. Eisenmann ◽  
M. Becker ◽  
G. Beck‐Wirth ◽  
E. Wunder ◽  
...  

2020 ◽  
Author(s):  
Xitiz Chamling ◽  
Alyssa Kallman ◽  
Cindy Berlinicke ◽  
Prajwal Devkota ◽  
Joseph L. Mertz ◽  
...  

ABSTRACTInjury and loss of oligodendrocytes can cause demyelinating diseases such as multiple sclerosis. To improve our understanding of oligodendrocyte development, which could facilitate development of remyelination-based treatment strategies, we performed single-cell-transcriptomic-analysis of developing human oligodendrocyte-precursor-cells (hOPCs). We engineered knock-in hESC-reporter lines in which an Identification-and-Purification tag is expressed under control of the endogenous, OPC-specific, PDGFRα promoter, and performed time-course single-cell-RNA-sequencing of purified hOPCs. Our analysis uncovered marked transcriptional heterogeneity of PDGFRα+ hOPCs and identified regulatory genes and networks that control their differentiation and myelination competence. Pseudotime trajectory analysis revealed two distinct trajectories for the development of oligodendrocytes vs astrocytes from hOPCs. We also identified novel transcription factors and other genes that developing hOPCs potentially use to choose between oligodendrocyte vs astrocyte lineages. In addition, pathway enrichment analysis followed by pharmacological intervention of those pathways confirm that mTOR and cholesterol biosynthesis signaling pathways are involved in maturation of oligodendrocytes from hOPCs.


2021 ◽  
pp. 1-8
Author(s):  
Mengmeng Jiang ◽  
Haide Chen ◽  
Guoji Guo

<b><i>Background:</i></b> The kidney is a highly complex organ that performs diverse functions that are essential for health. Kidney disease occurs when the kidneys are damaged and fail to function properly. Single-cell analysis is a powerful technology that provides unprecedented insights into normal and abnormal kidney cell types and will transform our understanding of the mechanism underlying common kidney diseases. <b><i>Summary:</i></b> Our understanding of kidney disease pathogenesis is limited by the incomplete molecular characterization of cell types responsible for kidney functions. Application of single-cell technologies for the study of the kidney has revealed cellular heterogeneity, gene expression signatures, and molecular dynamics during the onset and development of kidney diseases. Single-cell analyses of kidney organoids and allograft tissues offer new insights into kidney organogenesis, disease mechanisms, and therapeutic outcomes. Collectively, a better understanding of kidney cell heterogeneity and the molecular dynamics of kidney diseases will improve diagnostic accuracy and facilitate the identification of novel treatment strategies in nephrology. <b><i>Key Message:</i></b> In this review article, we summarize recent single-cell studies on kidney diseases and discuss the impact of single-cell technology on both basic and clinical nephrology research.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 320-320
Author(s):  
Yong Shen ◽  
Mir Hossain ◽  
Isaac Knudson ◽  
Shaleen Thakur ◽  
Jorg Bungert

Abstract The human β-globin genes are expressed in a developmental stage-specific manner and regulated by many cis- and trans-regulatory components including a locus control region (LCR) and proximal promoter and enhancer elements. The two γ-globin genes, Gγ and Aγ, are expressed in the fetal period. Persistent expression of γ-globin in the adult ameliorates the symptoms associated with mutations of the adult β-globin gene, such as sickle cell disease or β-thalassemia. Thus, understanding the mechanisms by which the fetal globin genes are activated and silenced during development may lead to new avenues for the treatment of hemoglobinopathies. Using data from the human ENCODE project we identified a DNase I hypersensitive site located 4 Kbp upstream of the Gγ-globin gene (Gγ -4Kb DHS) in K562 cell line. Gγ -4Kb DHS is characterized by the presence of histone modifications typical for enhancer elements (H3K4 monomethylation and H3K27 acetylation) and binding of ubiquitous (USF, E2F, YY1, c-Myc, Egr1, and MafK) or tissue-restricted (NF-E2) transcription factors in K562 cells which express high levels of the γ-globin genes (Figure 1). The presence of USF and NF-E2 is interesting as both proteins have been implicated in the recruitment of transcription complexes to the β-globin gene locus (Crusselle-Davis et al., 2006, Mol. Cell. Biol.; Liang et al., 2009 Mol. Cell. Biol.; Zhou et al., 2010, J. Biol. Chem.; Stee & Hossain et al., 2015, Mol. Cell. Biol.). We generated and expressed in K562 cells a synthetic Zinc Finger DNA-Binding Domain (ZF-DBD) designed to specifically target the Gγ -4Kb DHS and interfere its activities (ZF@Gγ-4KbDHS). The target site for the ZF-DBD overlaps with a CCCAC Egr1 motif and is close to an E-box sequence, which is predicted to bind USF and c-Myc (Figure 1). Figure 2A and C shows the Western blot results for cells transfected with empty vector or cells expressing the ZF@Gγ-4KbDHS in cell pools or a single cell clone selected from the pool of transfected cells (Figure 2A and C, respectively). We analyzed the binding of ZF@Gγ-4KbDHS at the globin locus in K562 cells using Chromatin Immunoprecipitation (ChIP). The data demonstrate that the ZF@Gγ-4KbDHS efficiently interacted with the Gγ -4Kb DHS and less efficiently with the γ-globin promoters (Figure 2B). Expression of the ZF@Gγ-4KbDHS led to a significant reduction in expression of the γ-globin genes but had no effect on expression of the GATA-1 gene (Figure 2D). The data suggest that the Gγ -4Kb DHS contributes to high-level γ-globin gene expression in K562 cells. Additionally, a SNP (rs11036496, Figure 1) within the Gγ -4Kb DHS has been reported to be associated with a disorder of γ-globin gene expression in African Americans and Chromatin Conformation Capture (3C) experiments showed that the Gγ-globin upstream region participates in interactions with the LCR and γ-globin genes (Shriner et al., 2015, BMC Medical Genetics; Kiefer et al., 2011, Blood). Therefore, further characterization of the Gγ -4Kb DHS will enhance understanding molecular mechanism(s) regulating hemoglobin switching. Figure 1 Epigenetic signatures and transcription factor binding at the Gγ -4Kb DHS. Shown on top are the two γ-globin genes and the relative position of the Gγ -4Kb DHS. Shown on the bottom is an enlarged view of the Gγ -4Kb DHS and binding peaks for several transcription factors as indicated. Figure 1. Epigenetic signatures and transcription factor binding at the Gγ -4Kb DHS. Shown on top are the two γ-globin genes and the relative position of the Gγ -4Kb DHS. Shown on the bottom is an enlarged view of the Gγ -4Kb DHS and binding peaks for several transcription factors as indicated. Figure 2 Reduced expression of γ-globin in K562 cells expressing the ZF@Gγ-4KbDHS. K562 cells were transfected with a plasmid expressing the ZF@Gγ-4KbDHS or with an empty vector. The K562 cells (pool) were subjected to Western blotting (A) and to Chromatin Immunoprecipitation (ChIP) using antibodies specific for the FLAG-tag, which is linked to the ZF@Gγ-4KbDHS, or negative control antibodies IgG (B). Single clonal K562 cells were subjected to Western blotting using antibodies specific for the ZF-DBD backbone (αZF) or for CTCF or GATA-1 as indicated (C). The single cell clone expressing the ZF@Gγ-4KbDHS was subjected to expression analysis by RT-qPCR using primers specific for γ-globin or GATA-1 as indicated (D). Figure 2. Reduced expression of γ-globin in K562 cells expressing the ZF@Gγ-4KbDHS. K562 cells were transfected with a plasmid expressing the ZF@Gγ-4KbDHS or with an empty vector. The K562 cells (pool) were subjected to Western blotting (A) and to Chromatin Immunoprecipitation (ChIP) using antibodies specific for the FLAG-tag, which is linked to the ZF@Gγ-4KbDHS, or negative control antibodies IgG (B). Single clonal K562 cells were subjected to Western blotting using antibodies specific for the ZF-DBD backbone (αZF) or for CTCF or GATA-1 as indicated (C). The single cell clone expressing the ZF@Gγ-4KbDHS was subjected to expression analysis by RT-qPCR using primers specific for γ-globin or GATA-1 as indicated (D). Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. SCI-2-SCI-2
Author(s):  
Bertie Gottgens

Abstract Transcription factor proteins have long been recognized as key regulators of blood stem cell function. They are thought to act as components of wider regulatory networks, with combinatorial interactions responsible for directing blood stem cell fate choices. However, the complexity of these networks coupled with the infrequency of stem cells have presented formidable challenges for past research efforts aiming to define blood stem cell regulatory networks. Importantly, recent innovations in single cell genomics and computational network inference technologies offer new opportunities to better understand the regulatory network control of blood cell development. The Göttgens group uses a combination of experimental and computational approaches to study how transcription factor networks control the function of blood stem cells and how mutations that perturb such networks cause leukemia. This integrated approach has resulted in the discovery of new combinatorial interactions between key blood stem cell regulators, as well as experimentally validated computational models for blood stem cells. Current research focuses on single cell genomics of early blood development and the development of computer models to chart the transcriptional landscape of blood stem and progenitor cell differentiation. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document