chip assay
Recently Published Documents


TOTAL DOCUMENTS

94
(FIVE YEARS 25)

H-INDEX

13
(FIVE YEARS 4)

2021 ◽  
Author(s):  
Ada Hang-Heng Wong ◽  
Semih Can Akincilar ◽  
Joelle Yi Heng Chua ◽  
Dhakshayini d/o K. Chanthira Morgan ◽  
Dorcas Hei ◽  
...  

Droplet microfluidics provides a miniaturized platform to conduct biological assays. We previously developed a droplet microfluidic chip assay for screening cancer cells against chemical drugs and chimeric antigen receptor T (CAR-T) cells, respectively. In this study, we investigated chip application on a cytokine expression assay using MCF7 breast cancer reporter cells engineered by fusing green fluorescent protein (GFP) to the C-terminus of endogenous interleukin-6 (IL6) gene. Combined tumor necrosis factor alpha (TNFalpha) treatment and serum-free medium starvation stimulated IL6-GFP expression and enhanced GFP fluorescence. Our data showed that on-chip assay recapitulates the cellular response in vitro, although absolute quantification of IL6 induction could not be accomplished. The demonstration of multi-timepoint IL6 expression analysis paves the way for our future study on tumor response to immune attack via cytokine signaling.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2667-2667
Author(s):  
Chandraditya Chakraborty ◽  
Srikanth Talluri ◽  
Eugenio Morelli ◽  
Sanika Derebail ◽  
Yao Yao ◽  
...  

Abstract Integrated genomic analysis including whole genome and RNA sequencing in primary multiple myeloma (MM) cells have reported a universal loss of BCL7A gene in MM patients compared to normal plasma cells (PC). Our Genetic modulation in in-vitro and in-vivo MM models have also validated the loss of BCL7A as an oncogenic event responsible for acquisition of a more proliferative phenotype in MM cells. We performed a comparative mass spectrometric analysis confirming BCL7A as a member of the canonical m-SWI/SNF chromatin remodeling complex. We therefore performed the Assay for Transposase Accessible Chromatin with high-throughput sequencing (ATAC-seq) to assess genome-wide changes in DNA accessibility upon BCL7A gain or loss. We found that loss of BCL7A in wild-type KMS12BM and NCI-H929 cells resulted in enrichment of important transcription factor motifs in the transcriptionally active sites of chromatin. We identified 36 de novo accessible and 1079 de novo inaccessible regions across the genome after BCL7A shRNA KD (knock-down). These genomic regions with altered accessibility were associated with genes involved in protein binding (FDR < 0.001), GTPase activation (FDR = 0.016), and GTPase regulation (FDR = 0.030). Candidate transcription factors for the genomic regions with altered accessibility were identified by querying a database of human ChiP-seq experiments (ReMap 2020). IRF4 was identified to be enriched in regions of accessible chromatin upon BCL7A loss. IRF4 is an oncoprotein transcription factor and is a direct target of myc, generating a feedback loop in MM cells. IRF4 dependency is central to myeloma cell proliferation. Most importantly, we found that in addition to functions within the m-SWI/SNF complex, BCL7A forms a protein complex with IRF4. Altogether these data suggest a role for BCL7A in driving IRF4 oncogenic activities in MM. To understand how BCL7A-dependent changes may influence IRF4 activity, we performed CHiP assay using IRF4 antibody in BCL7A KO (knock-out) and AB (add-back) KMS12BM and NCI-H929 cells. We observed increased binding of IRF4 to the promoter of its target genes like PRDM1, CDK6, STAG2, PIM2, SQLE, Myc, CANX, IRF4, SCD, ELL2, CASP3 in BCL7A KO cells, while the binding of these target genes was significantly decreased in AB cells. While, CHiP assay using IRF4 antibody in control and ectopically expressed BCL7A in AMO1 and KMS11 cells showed significantly low binding of IRF4 to the promoter of most of its target genes in BCL7A overexpressed cells compared to control. Integrated transcriptomic analysis following BCL7A KD and overexpression revealed the existence of a set of genes transcriptionally regulated by IRF4 to be significantly upregulated following BCL7A depletion and downregulated following ectopic expression of BCL7A. To investigate whether these genes are involved in the phenotypic and functional effects observed in MM after BCL7A depletion, we performed LOF studies (si-RNA screen) in scrambled and BCL7A KD MM cells. Among others, we observed that MM cells are highly sensitive to the inhibition of EEF1B2, RPS3A, SOX2, DCC and NDUFA1 only in the context of BCL7A loss, implicating a role as critical effector molecules downstream of the IRF4-BCL7A transcriptional network. In conclusion, we observe that BCL7A binds to IRF4, functionally restricting its activity. The universally observed down regulation of BCL7A in MM provides the necessary molecular change to allow IRF4 to exert its required transcriptional activity to induce MM cell growth. Our results now provide the basis to understand the mechanism of development of IRF4 dependency in MM. Disclosures Anderson: Pfizer: Membership on an entity's Board of Directors or advisory committees; Millenium-Takeda: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Sanofi-Aventis: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Membership on an entity's Board of Directors or advisory committees; Scientific Founder of Oncopep and C4 Therapeutics: Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company; Mana Therapeutics: Membership on an entity's Board of Directors or advisory committees. Munshi: Janssen: Consultancy; Karyopharm: Consultancy; Adaptive Biotechnology: Consultancy; Novartis: Consultancy; Legend: Consultancy; Bristol-Myers Squibb: Consultancy; Amgen: Consultancy; Abbvie: Consultancy; Takeda: Consultancy; Oncopep: Consultancy, Current equity holder in publicly-traded company, Other: scientific founder, Patents & Royalties; Celgene: Consultancy; Pfizer: Consultancy.


Epigenomics ◽  
2021 ◽  
Author(s):  
Wen-Na Liu ◽  
Kai-Xuan Wu ◽  
Xiao-Tong Wang ◽  
Li-Rong Lin ◽  
Man-Li Tong ◽  
...  

Aim: Neurosyphilis patients exhibited significant expression of long noncoding RNA (lncRNA) in peripheral blood T lymphocytes. In this study, we further clarified the role of lncRNA- ENST00000421645 in the pathogenic mechanism of neurosyphilis. Methods: lncRNA- ENST00000421645 was transfected into Jurkat-E6-1 cells, namely lentivirus (Lv)-1645 cells. RNA pull-down assay, flow cytometry, RT-qPCR, ELISA (Neobioscience Technology Co Ltd, Shenzhen, China) and RNA immunoprecipitation chip assay were used to analyze the function of lncRNA- ENST00000421645. Results: The expression of IFN-γ in Lv-1645 cells was significantly increased compared to that in Jurkat-E6-1 cells stimulated by phorbol-12-myristate-13-acetate (PMA). Then, it was suggested that lncRNA- ENST00000421645 interacts with PCM1 protein. Silencing PCM1 significantly increased the level of IFN-γ in Lv-1645 cells stimulated by PMA. Conclusion: This study revealed that lncRNA- ENST00000421645 mediates the production of IFN-γ by sponging PCM1 protein after PMA stimulation.


2021 ◽  
Author(s):  
Li Gao ◽  
Miao-Miao Yang ◽  
Ming-Juan Gu ◽  
Yun-Peng Liu ◽  
Cai-Hong Bu ◽  
...  

Abstract Background: Myostatin (MSTN), also known as growth/differentiation factor 8, mostly expressed in skeletal muscle and plays negative roles in regulation of muscle development. Previous studies had proved that MSTN have important effect on cell proliferation. Therefore we aimed to investigate the mechanism of MSTN in regulating the proliferation of bovine muscle satellite cells (MSCs).Methods: Bovine MSCs of MSTN mutant (MT) and wild type (WT) were obtained, we detected the cell proliferation and cell cycle by EdU proliferation assay and Flow cytometry. Then we detected the expression of genes associated with cell cycle by Real-time PCR and Western blotting . RNA-seq and Chromatin immunoprecipitation (ChIP)assay were performed to research the mechanism of MSTN in regulating the cell proliferation. Results: In this study, we found that MSTN mutant promoted the proliferation of MSCs. The expression of CyclinA, CyclinD and CyclinE were all increased after MSTN mutant, while the expression of CDKN1C (P57), CDKN2A, CDKN2C and CDKN2D were down-regulated, which were consistent with the promotion of cell proliferation. Among these genes, CDKN1C(P57) down-regulated most significantly. RNA-seq results showed that MSTN mutant affected the SMAD binding, so we performed ChIP-qPCR and demonstrated that the SMAD2/SMAD3 transcription factor combined with the promoter of CDKN1C thus to increase the expression of CDKN1C, this demonstrating that MSTN regulated the expression of CDKN1C through SMAD2/SMAD3 complex. Finally, overexpression of SMAD3 in wild type cells increased the expression of CDKN1C, further suggested that SMAD3 regulated the expression of CDKN1C. Conclusion: MSTN mutant down-regulated the expression of SMAD2/SMAD3, then reduced the promotion of SMAD2/SMAD3 to the expression of CDKN1C, thus to inhibit the expression of CDKN1C, then promoting the cell cycle.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Hai-Tao Jiang ◽  
Rui Deng ◽  
Yan Deng ◽  
Mao Nie ◽  
Yi-Xuan Deng ◽  
...  

Abstract Background Glucocorticoid-induced osteoporosis (GIOP) is the most common secondary osteoporosis. Patients with GIOP are susceptible to fractures and the subsequent delayed bone union or nonunion. Thus, effective drugs and targets need to be explored. In this regard, the present study aims to reveal the possible mechanism of the anti-GIOP effect of all-trans retinoic acid (ATRA). Methods Bone morphogenetic protein 9 (BMP9)-transfected mesenchymal stem cells (MSCs) were used as an in vitro osteogenic model to deduce the relationship between ATRA and dexamethasone (DEX). The osteogenic markers runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), and osteopontin were detected using real-time quantitative polymerase chain reaction, Western blot, and immunofluorescent staining assay. ALP activities and matrix mineralization were evaluated using ALP staining and Alizarin Red S staining assay, respectively. The novel genes associated with ATRA and DEX were detected using RNA sequencing (RNA-seq). The binding of the protein–DNA complex was validated using chromatin immunoprecipitation (ChIP) assay. Rat GIOP models were constructed using intraperitoneal injection of dexamethasone at a dose of 1 mg/kg, while ATRA intragastric administration was applied to prevent and treat GIOP. These effects were evaluated based on the serum detection of the osteogenic markers osteocalcin and tartrate-resistant acid phosphatase 5b, histological staining, and micro-computed tomography analysis. Results ATRA enhanced BMP9-induced ALP, RUNX2 expressions, ALP activities, and matrix mineralization in mouse embryonic fibroblasts as well as C3H10T1/2 and C2C12 cells, while a high concentration of DEX attenuated these markers. When DEX was combined with ATRA, the latter reversed DEX-inhibited ALP activities and osteogenic markers. In vivo analysis showed that ATRA reversed DEX-inhibited bone volume, bone trabecular number, and thickness. During the reversal process of ATRA, the expression of retinoic acid receptor beta (RARβ) was elevated. RARβ inhibitor Le135 partly blocked the reversal effect of ATRA. Meanwhile, RNA-seq demonstrated that serine protease inhibitor, clade A, member 3N (Serpina3n) was remarkably upregulated by DEX but downregulated when combined with ATRA. Overexpression of Serpina3n attenuated ATRA-promoted osteogenic differentiation, whereas knockdown of Serpina3n blocked DEX-inhibited osteogenic differentiation. Furthermore, ChIP assay revealed that RARβ can regulate the expression of Serpina3n. Conclusion ATRA can reverse DEX-inhibited osteogenic differentiation both in vitro and in vivo, which may be closely related to the downregulation of DEX-promoted Serpina3n. Hence, ATRA may be viewed as a novel therapeutic agent, and Serpina3n may act as a new target for GIOP.


2020 ◽  
Author(s):  
Yining Zhang ◽  
Tingting Meng ◽  
Jianan Chen ◽  
Ying Zhang ◽  
Jianning Kang ◽  
...  

Abstract Background Reactive astrocytes play an important role in Traumatic Spinal Cord Injury (TSCI). Interestingly, naive astrocytes can easily transform into neurotoxic reactive astrocytes(A1s) when inflammatory stimulation occurs. Previous researches have reported that miR-21a-5p is involved in the regulation of various stages of Spinal Cord Injury (SCI). However, it is not clear whether miR-21a-5p affected the polarization of reactive astrocytes. The purpose of our study was to detect the effects and mechanism of miR-21a-5p in the induction of neurotoxic reactive astrocytes (A1s) formation. Methods Gene chip assay and qRT-PCR were used to detect the expression of Cntfr α in TSCI models or sham operation. Bioinformatics analysis was used to speculate the potential targeting of miR-21a-5p, which was further confirmed by qRT-PCR, western blotting, a dual-luciferase reporter assay, and RNA pulldown assay. In vivo, the TSCI model was performed by a 68099Ⅱ precision percussion device, and the A1s phenotype was identified by immunofluorescence staining. In vitro, A1s were induced by IL-1 α, TNF-α, and C1q. A1s and neuroprotective reactive astrocytes (A2s) markers were confirmed by qRT-PCR, western blotting, and immunofluorescence. ChIP assay was used to explore the targeting gene of STAT3, the downstream of Cntfr α. Results The expression of miR-21a-5p was significantly increased while Cntfr α was decreased since naive astrocytes transformed into A1s after 3 days post-TSCI. In addition, the mRNA and protein of Cntfr α were decreased while miR-21a-5p was overexpressed. The binding site between miR-21a-5p and Cntfr α was further confirmed by the dual-luciferase reporter and RNA pulldown assay. We also discovered that A1s markers were decreased while markers of A2s were increased with the pretreatment of CNTF. Chromatin immunoprecipitation (ChIP) assay was used to prove that CNTF inhibited A1s induction by activating the expression of Nkrf via the CNTF/STAT3 pathway. Downregulation of miR-21a-5p enhanced the inhibitory effect of CNTF in A1s in vitro. In vivo, the expression of A1s markers significantly decreased with the treatment of antagomir-21, while Cntfr α siRNA treatment was just the opposite. Conclusion We observed that increased miR-21a-5p down-regulated Cntfr α in A1s induced by TSCI, promoting the inflammatory process. In addition, we also identified the effect and potential mechanism of CNTF, a specific ligand of CNTFR α, on inhibiting naive astrocytes transformed into A1s for the first time. Collectively, our studies demonstrated that targeting miR-21a-5p is a prospective therapy for curing TSCI.


2020 ◽  
Vol 11 ◽  
Author(s):  
Sidong Cai ◽  
Rong Liu ◽  
Panxia Wang ◽  
Jingyan Li ◽  
Tingting Xie ◽  
...  

The present study reveals a link between protein arginine methyltransferase 5 (PRMT5) and Homebox A9 (HoxA9) in the regulation of cardiomyocyte hypertrophy. In cardiomyocyte hypertrophy induced by β-adrenergic receptor agonist isoprenaline (ISO), PRMT5 expression was decreased while HoxA9 was upregulated. Silencing of PRMT5 or inhibition of PRMT5 by its pharmacological inhibitor EPZ augmented the expressions of cardiomyocyte hypertrophic genes brain natriuretic peptide (BNP) and β-Myosin Heavy Chain (β-MHC), whereas overexpression of PRMT5 inhibited ISO-induced cardiomyocyte hypertrophy, suggesting that PRMT5 ameliorates cardiomyocyte hypertrophy. On the contrary, HoxA9 promoted cardiomyocyte hypertrophy, as implied by the gain-of-function and loss-of-function experiments. HoxA9 was involved in the regulation of PRMT5 in cardiomyocyte hypertrophy, since HoxA9 knockdown prevented si-RPMT5-induced cardiomyocyte hypertrophy, and HoxA9 expression impaired the anti-hypertrophic effect of PRMT5. Co-immunoprecipitation experiments revealed that there were physical interactions between PRMT5 and HoxA9. The symmetric dimethylation level of HoxA9 was decreased by ISO or EPZ treatment, suggesting that HoxA9 is methylated by PRMT5. Additionally, PRMT5 repressed the expression of HoxA9. Chromatin immunoprecipitation (ChIP) assay demonstrated that HoxA9 could bind to the promoter of BNP, and that this binding affinity was further enhanced by ISO or EPZ. In conclusion, this study suggests that PRMT5 symmetric dimethylates HoxA9 and represses HoxA9 expression, thus impairing its binding to BNP promoter and ultimately protecting against cardiomyocyte hypertrophy. These findings provide a novel insight of the mechanism underlying the cardiac protective effect of PRMT5, and suggest potential therapeutic strategies of PRMT5 activation or HoxA9 inhibition in treatment of cardiac hypertrophy.


Biosensors ◽  
2020 ◽  
Vol 10 (12) ◽  
pp. 202
Author(s):  
Rosario Iemmolo ◽  
Valentina La Cognata ◽  
Giovanna Morello ◽  
Maria Guarnaccia ◽  
Mariamena Arbitrio ◽  
...  

Background: Antineoplastic agents represent the most common class of drugs causing Adverse Drug Reactions (ADRs). Mutant alleles of genes coding for drug-metabolizing enzymes are the best studied individual risk factors for these ADRs. Although the correlation between genetic polymorphisms and ADRs is well-known, pharmacogenetic tests are limited to centralized laboratories with expensive or dedicated instrumentation used by specialized personnel. Nowadays, DNA chips have overcome the major limitations in terms of sensibility, specificity or small molecular detection, allowing the simultaneous detection of several genetic polymorphisms with time and costs-effective advantages. In this work, we describe the design of a novel silicon-based lab-on-chip assay able to perform low-density and high-resolution multi-assay analysis (amplification and hybridization reactions) on the In-Check platform. Methods: The novel lab-on-chip was used to screen 17 allelic variants of three genes associated with adverse reactions to common chemotherapeutic agents: DPYD (Dihydropyrimidine dehydrogenase), MTHFR (5,10-Methylenetetrahydrofolate reductase) and TPMT (Thiopurine S-methyltransferase). Results: Inter- and intra assay variability were performed to assess the specificity and sensibility of the chip. Linear regression was used to assess the optimal hybridization temperature set at 52 °C (R2 ≈ 0.97). Limit of detection was 50 nM. Conclusions: The high performance in terms of sensibility and specificity of this lab-on-chip supports its further translation to clinical diagnostics, where it may effectively promote precision medicine.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 11-12
Author(s):  
Orit Uziel ◽  
Lian Lipshtein ◽  
Zinab Sarsor-Amer ◽  
Einat Beery ◽  
Meir Lahav ◽  
...  

Background: CLL is characterized by a gradual accumulation of mature appearing long-lived lymphocytes. Several pro-survival pathways that protect CLL cells from apoptosis are activated in these cells. For example, IL-6 dependent phosphorylation of the signal transducer and activator of transcription 3 (STAT3) provides CLL cells with a survival and a proliferative advantages. What is the source of IL-6 is currently unknown. Within lymphoid organs CLL cells engage in complex molecular interactions. In these sites, CLL cells are not simply the seeds that grow on the supportive soil of the microenvironment but play an active role in shaping its surrounding. Secreted by all types of cells, exosomes are nano-scaled particles that travel in blood and function as stable intercellular transport vehicles that deliver their cargo to cells that engulf them. For example, CLL-derived exosomes (herein CLL-exosomes) are taken up by mesenchymal stromal cells, transforming them to cancer associated fibroblasts. Given the appropriate stimulation, endothelial cells produce IL-6. Therefore, we hypothesized that CLL-exosomes actively recruit endothelial cells to become IL-6-secreting cells.Methods: CLL cells from 37 patients were included in this study. CLL-exosomes were isolated by ultracentrifugation. Electron microscopy, NanoSight tracking analysis, flow cytometry and Western immunoblotting were used to characterize CLL exosomes. Exosomal uptake by HUVECs was assessed by flow cytometry and fluorescent microscopy. The phosphor-protein profiling of exposed HUVECs was analyzed by mass spectrometry. RT-PCR and Western immunoblotting were used to determine the expression profile of HUVEC-exposed cells. HUVECs were transfected with β-catenin containing plasmid using DNA transfection reagent. Cytokine levels were determined by ELISA and CHIP assay was used to identify activated transcription factors .Results: First, we isolated CLL-exosomes from 37 treatment naïve patients. For that purpose we grew CLL cells and collected the secreted exosomes after 72 hours by ultracentrifugation. By NanoSight tracking system we identified large amount of exosomes and verified the presence of the typical cap shaped vesicles by electron microscopy. Western immunoblotting confirmed the presence of CD63 and CD81 exosomal markers and by flow we detected CD19/CD5 on these particles. Next, we exposed HUVECs to an increasing amount of CLL-exosomes and verified by two distinct methods that these particles are up taken by HUVECs in a dose- and time- dependent manner. By mass spectrometry we found 53 phosphorproteins that were at least 2 folds upregulated and none that were downregulated in HUVEC-exposed cells. Pathway analysis unraveled the central position of β-catenin. Immunoprecipitation studies verified that levels of phosphor-β-catenin are upregulated while levels of total β-catenin remained unchanged, suggesting that CLL-exosomes induced phosphorylation rather than the generation of newly formed β-catenin and leaving us wondering whether upregulation of phosphor β-catenin is somehow beneficial to their parental neoplastic cells. Because the IL-6 promoter binds 3 transcription factors that are activated by β-catenin we assumed that by activating the β-catenin pathway, endothelial cells become "micro factories" for the production of humoral IL-6. To verify the role of β-catenin in promoting production of IL-6 molecules in endothelial cells we transfected HUVECs with β-catenin containing plasmid. By Western immunoblotting we verified that the protein levels of β-catenin were upregulated. Then, by ChIP assay we found that 3 different transcription factors, namely LEF/TCF, CEBP and NFkB increased their binding to the IL-6 promoter region by 1.5 to 12 folds in HUVECs that were transfected with β-catenin ORF. Finally we show that intracellular STAT3 of CLL cells that were grown on this IL-6-rich medium are phosphorylated on tyrosine residues and that the rate of CLL cells in active apoptosis was markedly decreased. Conclusions: CLL cells shape their own fate. They do so by sending exosomes that activate the β-catenin axes. In this way CLL cells reprogram endothelial cells to become IL-6 producing cells and IL-6 contributes to CLL cells' survival. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 11 (10) ◽  
Author(s):  
Zhange Li ◽  
Honglin Xu ◽  
Xin Liu ◽  
Yang Hong ◽  
Han Lou ◽  
...  

Abstract NLRP3 (Nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3) inflammasome-mediated cardiomyocytes pyroptosis plays a crucial part in progression of acute myocardial infarction (MI). GDF11 (Growth Differentiation Factor 11) has been reported to generate cytoprotective effects in phylogenesis and multiple diseases, but the mechanism that GDF11 contributes to cardioprotection of MI and cardiomyocytes pyroptosis remains poorly understood. In our study, we first determined that GDF11 was abnormally downregulated in the heart tissue of MI mice and hypoxic cardiomyocytes. Moreover, AAV9-GDF11 markedly alleviated heart function in MI mice. Meanwhile, GDF11 overexpression also decreased the pyroptosis of hypoxic cardiomyocytes. PROMO and JASPAR prediction software found that transcription factor HOXA3 was predicted as an important regulator of NLRP3, and was confirmed by ChIP assay. Further analysis identifying GDF11 promoted the Smad2/3 pathway resulted in HOXA3 overexpression. Taken together, our study implies that GDF11 prevents cardiomyocytes pyroptosis via HOXA3/NLRP3 signaling pathway in MI mice.


Sign in / Sign up

Export Citation Format

Share Document