Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.070
Filter
1.
Mol Biol Rep ; 51(1): 1028, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39349848

ABSTRACT

BACKGROUND: Normalization with respect to stable housekeeping genes is important to facilitate gene transcription regulation research and acquire more accurate quantitative polymerase chain reaction (qPCR) data. In the current study, five candidates housekeeping genes of the cotton leafworm, Spodoptera littoralis encoding for Actin (Actin), elongation factor 1-alpha (EF1α), ribosomal protein S3 (RPS3), ribosomal protein 49 (RP49), and Ubiquitin (Ubi), were evaluated as normalization housekeeping genes under Spodoptera littoralis nucleopolyhedrovirus (SpliNPV) viral infection. METHODS AND RESULTS: The qPCR results confirmed the expression of all five housekeeping genes in S. littoralis viral infected larvae. The expression profiles of the housekeeping genes showed that the EF1α, Actin, and RP49 had the minimum average Ct values of 18.41 ± 0.66, 18.84 ± 0.90 and 19.01 ± 0.87 in all infected samples, respectively. While RPS3 and Ubi showed the maximum average Ct of 21.61 ± 0.51 and 21.11 ± 0.82, respectively. According to the results of ΔCt and geNorm analysis, EF1α was ranked as the most stable housekeeping gene during infection time-course. While by using BestKeeper, geNorm and NormFinder, the Ubi, RP49, and RPS3 showed the most genes transcription stability. The obtained results were also validated using the Cytochrome c oxidase (COX) gene transcripts in response to SpliNPV infection. CONCLUSIONS: The results revealed that EF1α and Ubi were the most stable housekeeping genes to be used for normalizing S. littoralis gene transcription regulation under SpliNPV infection. These findings, provide a significant addition for gene transcription regulation studies of S. littoralis upon infection using SpliNPV as a bio-agent.


Subject(s)
Genes, Essential , Nucleopolyhedroviruses , Spodoptera , Animals , Spodoptera/genetics , Spodoptera/virology , Genes, Essential/genetics , Nucleopolyhedroviruses/genetics , Gene Expression Regulation , Larva/genetics , Larva/virology , Transcription, Genetic/genetics , Gene Expression Profiling/methods , Insect Proteins/genetics
2.
Genes (Basel) ; 15(9)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39336776

ABSTRACT

SINE-VNTR-Alu (SVA) retrotransposons can regulate expression quantitative trait loci (eQTL) of coding and noncoding genes including transposable elements (TEs) distributed throughout the human genome. Previously, we reported that expressed SVAs and human leucocyte antigen (HLA) class II genotypes on chromosome 6 were associated significantly with Parkinson's disease (PD). Here, our aim was to follow-up our previous study and evaluate the SVA associations and their regulatory effects on the transcription of TEs within the HLA class II genomic region. We reanalyzed the transcriptome data of peripheral blood cells from the Parkinson's Progression Markers Initiative (PPMI) for 1530 subjects for TE and gene RNAs with publicly available computing packages. Four structurally polymorphic SVAs regulate the transcription of 20 distinct clusters of 235 TE loci represented by LINES (37%), SINES (28%), LTR/ERVs (23%), and ancient transposon DNA elements (12%) that are located in close proximity to HLA genes. The transcribed TEs were mostly short length, with an average size of 389 nucleotides. The numbers, types and profiles of positive and negative regulation of TE transcription varied markedly between the four regulatory SVAs. The expressed SVA and TE RNAs in blood cells appear to be enhancer-like elements that are coordinated differentially in the regulation of HLA class II genes. Future work on the mechanisms underlying their regulation and potential impact is essential for elucidating their roles in normal cellular processes and disease pathogenesis.


Subject(s)
Parkinson Disease , Quantitative Trait Loci , Humans , Parkinson Disease/genetics , Parkinson Disease/pathology , DNA Transposable Elements/genetics , Alu Elements/genetics , Short Interspersed Nucleotide Elements/genetics , Transcription, Genetic/genetics , Gene Expression Regulation/genetics , Disease Progression , Histocompatibility Antigens Class II/genetics
3.
STAR Protoc ; 5(3): 103284, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39217608

ABSTRACT

Quantifying the number of proteins that interact with mRNAs, in particular with poly(A) tails of mRNAs, is crucial for understanding gene regulation. Biochemical assays offer significant advantages for this purpose. Here, we present a protocol for synthesizing mRNAs with accurate, length-specific poly(A) tails through a PCR-based approach. We also describe steps for an in vitro (i.e., cell-free) approach for visualizing the sequential binding of Cytoplasmic Poly(A)-Binding Proteins (PABPCs) to these poly(A) tails. We detail quality control steps throughout the procedure. For complete details on the use and execution of this protocol, please refer to Grandi et al.1.


Subject(s)
Biochemistry , Poly A , Poly(A)-Binding Proteins , RNA, Messenger , Humans , Poly A/metabolism , Poly(A)-Binding Proteins/metabolism , Poly(A)-Binding Proteins/genetics , Polymerase Chain Reaction/methods , Protein Binding , RNA, Messenger/metabolism , RNA, Messenger/genetics , Transcription, Genetic/genetics , Biochemistry/methods
4.
Cell Syst ; 15(9): 808-823.e6, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39243757

ABSTRACT

The regulation of genes can be mathematically described by input-output functions that are typically assumed to be time invariant. This fundamental assumption underpins the design of synthetic gene circuits and the quantitative understanding of natural gene regulatory networks. Here, we found that this assumption is challenged in mammalian cells. We observed that a synthetic reporter gene can exhibit unexpected transcriptional memory, leading to a shift in the dose-response curve upon a second induction. Mechanistically, we investigated the cis-dependency of transcriptional memory, revealing the necessity of promoter DNA methylation in establishing memory. Furthermore, we showed that the synthetic transcription factor's effective DNA binding affinity underlies trans-dependency, which is associated with its capacity to undergo biomolecular condensation. These principles enabled modulating memory by perturbing either cis- or trans-regulation of genes. Together, our findings suggest the potential pervasiveness of transcriptional memory and implicate the need to model mammalian gene regulation with time-varying input-output functions. A record of this paper's transparent peer review process is included in the supplemental information.


Subject(s)
DNA Methylation , Gene Expression Regulation , Promoter Regions, Genetic , Transcription Factors , Transcription, Genetic , DNA Methylation/genetics , Promoter Regions, Genetic/genetics , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation/genetics , Animals , Transcription, Genetic/genetics , Gene Regulatory Networks/genetics , Mammals/genetics
5.
STAR Protoc ; 5(3): 103277, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39196783

ABSTRACT

The repetitive C-terminal domain (CTD) of the largest subunit of RNA polymerase II (RNAPII) becomes differentially phosphorylated throughout the transcription cycle. Here, we present a protocol to site-specifically phosphorylate the CTD of RNAPII by leveraging the specificity of well-characterized CTD kinases. We describe the steps for optimal phosphorylation of the CTD and the preparation of nuclear protein extract. This protocol can be used to identify the interactome of a phospho-CTD and has the potential to identify novel RNAPII-binding proteins. For complete details on the use and execution of this protocol, please refer to Moreno et al.1.


Subject(s)
RNA Polymerase II , RNA Polymerase II/metabolism , RNA Polymerase II/chemistry , Phosphorylation , Humans , Transcription, Genetic/genetics
6.
Cell Syst ; 15(8): 694-708.e12, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39121860

ABSTRACT

Single-cell transcriptomics reveals significant variations in transcriptional activity across cells. Yet, it remains challenging to identify mechanisms of transcription dynamics from static snapshots. It is thus still unknown what drives global transcription dynamics in single cells. We present a stochastic model of gene expression with cell size- and cell cycle-dependent rates in growing and dividing cells that harnesses temporal dimensions of single-cell RNA sequencing through metabolic labeling protocols and cel lcycle reporters. We develop a parallel and highly scalable approximate Bayesian computation method that corrects for technical variation and accurately quantifies absolute burst frequency, burst size, and degradation rate along the cell cycle at a transcriptome-wide scale. Using Bayesian model selection, we reveal scaling between transcription rates and cell size and unveil waves of gene regulation across the cell cycle-dependent transcriptome. Our study shows that stochastic modeling of dynamical correlations identifies global mechanisms of transcription regulation. A record of this paper's transparent peer review process is included in the supplemental information.


Subject(s)
Cell Cycle , Gene Expression Regulation , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic , Single-Cell Analysis/methods , Sequence Analysis, RNA/methods , Transcription, Genetic/genetics , Gene Expression Regulation/genetics , Cell Cycle/genetics , Humans , Bayes Theorem , Transcriptome/genetics , Stochastic Processes
7.
Hum Cell ; 37(5): 1446-1461, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39014290

ABSTRACT

Cervical cancer poses a significant health burden for women globally, and the rapid proliferation of cervical cancer cells greatly worsens patient prognosis. Long non-coding RNAs (lncRNAs) play a crucial role in regulating tumor cell proliferation. However, the involvement of lncRNAs in cervical cancer cell proliferation remains unclear. In this study, we investigated the lncRNA SIX1-1, which was found to be upregulated in cervical cancer tissues and cell lines. Functional assays revealed that knockdown of SIX1-1 inhibited cell proliferation in vitro and reduced tumor growth in vivo. Mechanistically, SIX1-1 was predominantly localized in the nucleus and could bind with DNMT1 protein. The expression of SIX1-1 enhanced the interaction of DNMT1 with RASD1 promoter, leading to the methylation of the promoter and decreased mRNA transcription. Then RASD1 downregulation activated the cAMP/PKA/CREB signaling pathway, promoting cell proliferation. Rescue experiments showed that knockdown of RASD1 restored the inhibited cell proliferation caused by decreased expression of SIX1-1, indicating that RASD1 acted as the functional mediator of SIX1-1. In conclusion, SIX1-1 promoted cervical cancer cell proliferation by modulating RASD1 expression. This suggests that targeting the SIX1-1/RASD1 axis could be a potential antitumor strategy for cervical cancer.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding , Uterine Cervical Neoplasms , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Humans , Cell Proliferation/genetics , Female , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , RNA, Long Noncoding/metabolism , Gene Expression Regulation, Neoplastic/genetics , Cell Line, Tumor , Transcription, Genetic/genetics , Signal Transduction/genetics , ras Proteins/genetics , ras Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Gene Expression/genetics
8.
Hum Cell ; 37(5): 1505-1521, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39073525

ABSTRACT

The electrospun nanofiber system is correlated with high efficacy of drug delivery. This study aims to investigate the effect of nanofiber-based delivery of evodiamine, an indole alkaloid derived from Rutaceae plants Evodia rutaecarpa (Juss.) Benth, on intrahepatic cholangiocarcinoma (ICC), as well as to explore the molecular mechanisms. An electrospun nanofiber system carrying evodiamine was generated. Compared to evodiamine treatment alone, the nano-evodiamine exhibited more pronounced effects on suppressing proliferation, colony formation, invasiveness, migration, apoptosis resistance, cell cycle progression, and in vivo tumorigenesis of two ICC cell lines (HUCC-T1 and RBE). ICC cells exhibited increased expression of histone deacetylase 4 (HDAC4) while decreased tropomyosin 1 (TPM1). HDAC4 suppressed TPM1 expression by removing H3K9ac modifications from its promoter. Nano-evodiamine reduced HDAC4 protein levels in ICC cells, thus promoting transcription and expression of TPM1. Either overexpression of HDAC4 or downregulation of TPM1 negated the tumor-suppressive effects of nano-evodiamine. Collectively, this study demonstrates that the electrospun nanofiber system enhances the efficiency of evodiamine. Additionally, evodiamine suppresses the malignant properties of ICC cells. The findings may provide fresh insights into the application of electrospun nanofiber system for drug delivery and the effects of evodiamine on tumor suppression.


Subject(s)
Cholangiocarcinoma , Drug Delivery Systems , Histone Deacetylases , Nanofibers , Tropomyosin , Tropomyosin/genetics , Tropomyosin/metabolism , Humans , Cholangiocarcinoma/pathology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Cell Line, Tumor , Quinazolines/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Gene Expression/drug effects , Gene Expression/genetics , Molecular Targeted Therapy , Apoptosis/drug effects , Apoptosis/genetics , Repressor Proteins
10.
ACS Synth Biol ; 13(8): 2402-2411, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39023433

ABSTRACT

Plastid engineering offers the potential to carry multigene traits in plants; however, it requires reliable genetic parts to balance expression. The difficulty of chloroplast transformation and slow plant growth makes it challenging to build plants just to characterize genetic parts. To address these limitations, we developed a high-yield cell-free system from Nicotiana tabacum chloroplast extracts for prototyping genetic parts. Our cell-free system uses combined transcription and translation driven by T7 RNA polymerase and works with plasmid or linear template DNA. To develop our system, we optimized lysis, extract preparation procedures (e.g., runoff reaction, centrifugation, and dialysis), and the physiochemical reaction conditions. Our cell-free system can synthesize 34 ± 1 µg/mL luciferase in batch reactions and 60 ± 4 µg/mL in semicontinuous reactions. We apply our batch reaction system to test a library of 103 ribosome binding site (RBS) variants and rank them based on cell-free gene expression. We observe a 1300-fold dynamic range of luciferase expression when normalized by maximum mRNA expression, as assessed by the malachite green aptamer. We also find that the observed normalized gene expression in chloroplast extracts and the predictions made by the RBS Calculator are correlated. We anticipate that chloroplast cell-free systems will increase the speed and reliability of building genetic systems in plant chloroplasts for diverse applications.


Subject(s)
Cell-Free System , Chloroplasts , Nicotiana , Chloroplasts/genetics , Chloroplasts/metabolism , Nicotiana/genetics , Nicotiana/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Genetic Engineering/methods , Luciferases/genetics , Luciferases/metabolism , Plasmids/genetics , Plasmids/metabolism , Ribosomes/metabolism , Ribosomes/genetics , Binding Sites , Transcription, Genetic/genetics , Viral Proteins
11.
Genes Dev ; 38(11-12): 504-527, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38986581

ABSTRACT

Genome integrity relies on the accuracy of DNA metabolism, but as appreciated for more than four decades, transcription enhances mutation and recombination frequencies. More recent research provided evidence for a previously unforeseen link between RNA and DNA metabolism, which is often related to the accumulation of DNA-RNA hybrids and R-loops. In addition to physiological roles, R-loops interfere with DNA replication and repair, providing a molecular scenario for the origin of genome instability. Here, we review current knowledge on the multiple RNA factors that prevent or resolve R-loops and consequent transcription-replication conflicts and thus act as modulators of genome dynamics.


Subject(s)
Genomic Instability , R-Loop Structures , RNA , Genomic Instability/genetics , RNA/metabolism , RNA/genetics , DNA Replication/genetics , Animals , Humans , Transcription, Genetic/genetics
12.
Nature ; 632(8027): 1073-1081, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39020177

ABSTRACT

Measurements of gene expression or signal transduction activity are conventionally performed using methods that require either the destruction or live imaging of a biological sample within the timeframe of interest. Here we demonstrate an alternative paradigm in which such biological activities are stably recorded to the genome. Enhancer-driven genomic recording of transcriptional activity in multiplex (ENGRAM) is based on the signal-dependent production of prime editing guide RNAs that mediate the insertion of signal-specific barcodes (symbols) into a genomically encoded recording unit. We show how this strategy can be used for multiplex recording of the cell-type-specific activities of dozens to hundreds of cis-regulatory elements with high fidelity, sensitivity and reproducibility. Leveraging signal transduction pathway-responsive cis-regulatory elements, we also demonstrate time- and concentration-dependent genomic recording of WNT, NF-κB and Tet-On activities. By coupling ENGRAM to sequential genome editing via DNA Typewriter1, we stably record information about the temporal dynamics of two orthogonal signalling pathways to genomic DNA. Finally we apply ENGRAM to integratively record the transient activity of nearly 100 transcription factor consensus motifs across daily windows spanning the differentiation of mouse embryonic stem cells into gastruloids, an in vitro model of early mammalian development. Although these are proof-of-concept experiments and much work remains to fully realize the possibilities, the symbolic recording of biological signals or states within cells, to the genome and over time, has broad potential to complement contemporary paradigms for how we make measurements in biological systems.


Subject(s)
DNA , Gene Editing , Signal Transduction , Transcription, Genetic , Animals , Mice , Cell Differentiation/genetics , DNA/genetics , DNA/metabolism , Enhancer Elements, Genetic/genetics , Gene Editing/methods , Genomics , Mouse Embryonic Stem Cells/cytology , NF-kappa B/metabolism , Reproducibility of Results , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Signal Transduction/genetics , Time Factors , Transcription Factors/metabolism , Transcription, Genetic/genetics , Wnt Signaling Pathway/genetics , Nucleotide Motifs , Consensus Sequence/genetics , Developmental Biology , Proof of Concept Study
13.
PLoS Comput Biol ; 20(6): e1012194, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38857275

ABSTRACT

Transcription factors (TFs) regulate the process of transcription through the modulation of different kinetic steps. Although models can often describe the observed transcriptional output of a measured gene, predicting a TFs role on a given promoter requires an understanding of how the TF alters each step of the transcription process. In this work, we use a simple model of transcription to assess the role of promoter identity, and the degree to which TFs alter binding of RNAP (stabilization) and initiation of transcription (acceleration) on three primary characteristics: the range of steady-state regulation, cell-to-cell variability in expression, and the dynamic response time of a regulated gene. We find that steady state regulation and the response time of a gene behave uniquely for TFs that regulate incoherently, i.e that speed up one step but slow the other. We also find that incoherent TFs have dynamic implications, with one type of incoherent mode configuring the promoter to respond more slowly at intermediate TF concentrations. We also demonstrate that the noise of gene expression for these TFs is sensitive to promoter strength, with a distinct non-monotonic profile that is apparent under stronger promoters. Taken together, our work uncovers the coupling between promoters and TF regulatory modes with implications for understanding natural promoters and engineering synthetic gene circuits with desired expression properties.


Subject(s)
Promoter Regions, Genetic , Transcription Factors , Transcription Factors/metabolism , Transcription Factors/genetics , Promoter Regions, Genetic/genetics , Computational Biology , Gene Expression Regulation/genetics , Models, Genetic , Transcription, Genetic/genetics , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , Kinetics
14.
Adv Sci (Weinh) ; 11(29): e2307804, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837887

ABSTRACT

RNA splicing is crucial in the multilayer regulatory networks for gene expression, making functional interactions with DNA- and other RNA-processing machineries in the nucleus. However, these established couplings are all major spliceosome-related; whether the minor spliceosome is involved remains unclear. Here, through affinity purification using Drosophila lysates, an interaction is identified between the minor spliceosomal 65K/RNPC3 and ANKRD11, a cofactor of histone deacetylase 3 (HDAC3). Using a CRISPR/Cas9 system, Deletion strains are constructed and found that both Dm65KΔ/Δ and Dmankrd11Δ/Δ mutants have reduced histone deacetylation at Lys9 of histone H3 (H3K9) and Lys5 of histone H4 (H4K5) in their heads, exhibiting various neural-related defects. The 65K-ANKRD11 interaction is also conserved in human cells, and the HsANKRD11 middle-uncharacterized domain mediates Hs65K association with HDAC3. Cleavage under targets and tagmentation (CUT&Tag) assays revealed that HsANKRD11 is a bridging factor, which facilitates the synergistic common chromatin-binding of HDAC3 and Hs65K. Knockdown (KD) of HsANKRD11 simultaneously decreased their common binding, resulting in reduced deacetylation of nearby H3K9. Ultimately, this study demonstrates that expression changes of many genes caused by HsANKRD11-KD are due to the decreased common chromatin-binding of HDAC3 and Hs65K and subsequently reduced deacetylation of H3K9, illustrating a novel and conserved coupling mechanism that links the histone deacetylation with minor spliceosome for the regulation of gene expression.


Subject(s)
Histone Deacetylases , Histones , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histones/metabolism , Histones/genetics , Humans , Animals , Spliceosomes/metabolism , Spliceosomes/genetics , Acetylation , Drosophila/genetics , Drosophila/metabolism , Transcription, Genetic/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Repressor Proteins
15.
Mol Genet Genomics ; 299(1): 59, 2024 May 26.
Article in English | MEDLINE | ID: mdl-38796829

ABSTRACT

RECQL5 is a member of the conserved RecQ family of DNA helicases involved in the maintenance of genome stability that is specifically found in higher eukaryotes and associates with the elongating RNA polymerase II. To expand our understanding of its function we expressed human RECQL5 in the yeast Saccharomyces cerevisiae, which does not have a RECQL5 ortholog. We found that RECQL5 expression leads to cell growth inhibition, increased genotoxic sensitivity and transcription-associated hyperrecombination. Chromatin immunoprecipitation and transcriptomic analysis of yeast cells expressing human RECQL5 shows that this is recruited to transcribed genes and although it causes only a weak impact on gene expression, in particular at G + C-rich genes, it leads to a transcription termination defect detected as readthrough transcription. The data indicate that the interaction between RNAPII and RECQL5 is conserved from yeast to humans. Unexpectedly, however, the RECQL5-ID mutant, previously shown to have reduced the association with RNAPII in vitro, associates with the transcribing polymerase in cells. As a result, expression of RECQL5-ID leads to similar although weaker phenotypes than wild-type RECQL5 that could be transcription-mediated. Altogether, the data suggests that RECQL5 has the intrinsic ability to function in transcription-dependent and independent genome dynamics in S. cerevisiae.


Subject(s)
Genomic Instability , RecQ Helicases , Saccharomyces cerevisiae , Transcription, Genetic , Saccharomyces cerevisiae/genetics , Genomic Instability/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism , Humans , Transcription, Genetic/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
17.
STAR Protoc ; 5(2): 103071, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38768029

ABSTRACT

The elucidation of the role of microorganisms in human infections has been hindered by difficulties using conventional culture-based techniques. Here, we present a protocol for the investigation of transcriptionally active microbes (TAMs) using an RNA sequencing (RNA-seq)-based approach. We describe the steps for RNA isolation, viral genome sequencing, RNA-seq library preparation, and metatranscriptomic and transcriptomic analysis. This protocol permits a comprehensive evaluation of TAMs' contributions to the differential severity of infectious diseases, with a particular focus on diseases such as COVID-19. For complete details on the use and execution of this protocol, please refer to Devi et al.1.


Subject(s)
COVID-19 , RNA, Viral , RNA-Seq , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/virology , COVID-19/genetics , RNA-Seq/methods , RNA, Viral/genetics , Genome, Viral/genetics , Sequence Analysis, RNA/methods , Transcription, Genetic/genetics , Gene Expression Profiling/methods , Transcriptome/genetics
18.
Cell Cycle ; 23(5): 573-587, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38701194

ABSTRACT

Myogenic differentiation (MyoD) 1, which is known as a pivotal transcription factor during myogenesis, has been proven dysregulated in several cancers. However, litter is known about the precise role and downstream genes of MyoD1 in gastric cancer (GC) cells. Here, we report that MyoD1 is lowly expressed in primary GC tissues and cells. In our experiments, overexpression of MyoD1 inhibited cell proliferation. Downstream genes of MyoD1 regulation were investigated using RNA-Seq. As a result, 138 up-regulated genes and 20 down-regulated genes and 27 up-regulated lncRNAs and 20 down-regulated lncRNAs were identified in MyoD1 overexpressed MKN-45 cells, which participated in epithelial cell signaling in Helicobacter pylori infection, glycosaminoglycan biosynthesis (keratan sulfate), notch signaling pathway, and others. Among these genes, BIK was directly regulated by MyoD1 in GC cells and inhibited cancer cell proliferation. The BIK knockdown rescued the effects of MyoD1 overexpression on GC cells. In conclusion, MyoD1 inhibited cell proliferation via 158 genes and 47 lncRNAs downstream directly or indirectly that participated in multiple signaling pathways in GC, and among these, MyoD1 promotes BIK transcription by binding to its promoter, then promotes BIK-Bcl2-caspase 3 axis and regulates GC cell apoptosis.


Subject(s)
Apoptosis , Cell Proliferation , Gene Expression Regulation, Neoplastic , MyoD Protein , RNA, Long Noncoding , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Humans , Apoptosis/genetics , MyoD Protein/metabolism , MyoD Protein/genetics , Cell Proliferation/genetics , Cell Line, Tumor , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Transcription, Genetic/genetics
19.
PeerJ ; 12: e17234, 2024.
Article in English | MEDLINE | ID: mdl-38666079

ABSTRACT

Background: Post-translational modification by Small Ubiquitin-like MOdifier (SUMO) is an important mechanism to regulate protein activity, protein stability, and localization of substrates. Zbtb21 is a zinc finger and BTB (Broad-complex, Tram-track and Bric à brac) domain-containing transcription factor. Bioinformatic prediction suggests several putative SUMOylated sites in Zbtb21 protein. Methods: Two evolutionarily conserved lysine residues in Zbtb21 protein were mutated alone or in combination to disrupt the binding with SUMO molecules. Western blot and co-immunoprecipitation analyses were performed to detect the SUMOylation state of wild type and mutant Zbtb21 proteins, respectively. Luciferase reporter assays were conducted to evaluate their transcription activities. Meanwhile, immunofluorescence staining was carried out to show their sub-nuclear localizations. Finally, co-immunoprecipitation was performed to detect the interaction between Zbtb21 and its partners. Results: Phylogenetically conserved lysines 419 and 845 of zebrafish Zbtb21 protein can be conjugated with SUMO molecules. SUMOylation does not affect the subcellular localization and protein stability of Zbtb21, as well as the interaction with Zbtb14 or Zbtb21. Nevertheless, luciferase reporter assays revealed that Zbtb21 is a dual-function transcription factor which exerts activation or repression effect on different promoters, and SUMOylation can modulate the transcriptional activity of Zbtb21 in regulating downstream target genes. Hence, Zbtb21 is identified as a novel substrate of SUMOylation, which would be important for its function. Conclusions: Zebrafish Zbtb21 protein can be SUMOylated on lysines 419 and 845, which is evolutionary conserved. SUMOylation affects the dual role of Zbtb21 on transcription.


Subject(s)
Sumoylation , Zebrafish Proteins , Zebrafish , Sumoylation/genetics , Animals , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription, Genetic/genetics , Humans
20.
PLoS Comput Biol ; 20(4): e1012029, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38648221

ABSTRACT

The circadian clock is an evolutionarily-conserved molecular oscillator that enables species to anticipate rhythmic changes in their environment. At a molecular level, the core clock genes induce circadian oscillations in thousands of genes in a tissue-specific manner, orchestrating myriad biological processes. While previous studies have investigated how the core clock circuit responds to environmental perturbations such as temperature, the downstream effects of such perturbations on circadian regulation remain poorly understood. By analyzing bulk-RNA sequencing of Drosophila fat bodies harvested from flies subjected to different environmental conditions, we demonstrate a highly condition-specific circadian transcriptome: genes are cycling in a temperature-specific manner, and the distributions of their phases also differ between the two conditions. Further employing a reference-based gene regulatory network (Reactome), we find evidence of increased gene-gene coordination at low temperatures and synchronization of rhythmic genes that are network neighbors. We report that the phase differences between cycling genes increase as a function of geodesic distance in the low temperature condition, suggesting increased coordination of cycling on the gene regulatory network. Our results suggest a potential mechanism whereby the circadian clock mediates the fly's response to seasonal changes in temperature.


Subject(s)
Circadian Clocks , Circadian Rhythm , Gene Expression Regulation , Gene Regulatory Networks , Temperature , Animals , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Gene Regulatory Networks/genetics , Circadian Clocks/genetics , Circadian Clocks/physiology , Gene Expression Regulation/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Drosophila/genetics , Drosophila/physiology , Transcriptome/genetics , Computational Biology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL