Next Article in Journal
Microbial Synthesis of Lactic Acid from Cotton Stalk for Polylactic Acid Production
Next Article in Special Issue
Effects of Environmental Stresses on Synthesis of 2-Phenylethanol and IAA by Enterobacter sp. CGMCC 5087
Previous Article in Journal
Oral Microbiota, Bacterial Infections, Antibiotic Prescriptions, and Antimicrobial Resistance in Children
Previous Article in Special Issue
Extracellular Expression of Feruloyl Esterase and Xylanase in Escherichia coli for Ferulic Acid Production from Agricultural Residues
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Time-Course Transcriptome Analysis of Bacillus subtilis DB104 during Growth

Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University, Goyang-si 10326, Republic of Korea
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(8), 1928; https://doi.org/10.3390/microorganisms11081928
Submission received: 8 June 2023 / Revised: 27 July 2023 / Accepted: 28 July 2023 / Published: 28 July 2023
(This article belongs to the Special Issue Bacterial Engineering and Metabolism Regulation)

Abstract

:
Bacillus subtilis DB104, an extracellular protease-deficient derivative of B. subtilis 168, is widely used for recombinant protein expression. An understanding of the changes in gene expression during growth is essential for the commercial use of bacterial strains. Transcriptome and proteome analyses are ideal methods to study the genomic response of microorganisms. In this study, transcriptome analysis was performed to monitor changes in the gene expression level of B. subtilis DB104 while growing on a complete medium. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, K-mean cluster analysis, gene ontology (GO) enrichment analysis, and the function of sigma factors were used to divide 2122 differentially expressed genes (DEGs) into 10 clusters and identified gene functions according to expression patterns. The results of KEGG pathway analysis indicated that ABC transporter is down-regulated during exponential growth and metabolic changes occur at the transition point where sporulation starts. At this point, several stress response genes were also turned on. The genes involved in the lipid catabolic process were up-regulated briefly at 15 h as an outcome of the programmed cell death that postpones sporulation. The results suggest that changes in the gene expression of B. subtilis DB104 were dependent on the initiation of sporulation. However, the expression timing of the spore coat gene was only affected by the relevant sigma factor. This study can help to understand gene expression and regulatory mechanisms in B. subtilis species by providing an overall view of transcriptional changes during the growth of B. subtilis DB104.

1. Introduction

Bacillus subtilis is a Gram-positive endospore-forming bacterium and is not considered pathogenic or toxigenic to humans and animals [1,2]. Because of its well-known genetic background and easy genetic manipulation, it is an ideal strain as a host for metabolic engineering or industrial enzyme. B. subtilis DB104, a protease-deficient derivative of B. subtilis 168, is widely used for recombinant protein expression [3].
Understanding bacterial gene expression is essential for the development of a strain or vector for industrial use. To date, microarray technology has been used to monitor bacterial gene expression. Patten et al. [4] described alternative sigma factor RpoS-dependent gene expression in Escherichia coli K-12 using a microarray, while Kitichalermkiat et al. [5] explored the effect of epigallocatechin gallate on Staphylococcus aureus gene expression using this technology. During growth in specific conditions, such as with glucose [6] and batch culture [7], transcript profiling was also conducted on B. subtilis using microarrays. Because the technique relies on hybridization, several disadvantages exist, such as background hybridization that could limit the accuracy and difficulty of designing a large number of probes. RNA-sequencing (RNA-seq) is another technique that provides information about gene expression [8]. RNA-seq based on next-generation sequencing is a widely used transcriptome analysis technology. However, it has limitations of information loss due to the short read length that must be assembled [8,9]. Direct RNA-seq using nanopore is a recently developed technology that can avoid the limitation of the classical RNA-seq [10,11].
RNA-seq and proteomics have become the favored technology platforms for large-scale expression analysis due to the advantages of sensitive and precise detection using technological tools. To date, various omics studies have been conducted on B. subtilis. For example, transcriptome or proteome analysis was conducted during specific growth stages, such as early to middle sporulation [12] and germination [13]. Zhang et al. [14] revealed the transcriptional response to cell envelope stress in B. subtilis. Nicolas et al. [15] explored the regulatory architecture of B. subtilis using transcriptome analysis in a wide range of conditions. Proteome and transcriptome analyses were applied to define the response to salicylic acid by Van Duy et al. [16] and the response to salt stress by Hahne et al. [17]. Meanwhile, Xu et al. [18] suggested that tmRNA, which liberate ribosomes arrested at mRNA lacking a stop codon, controls biofilm formation using RNA-seq analysis.
However, as it can be seen, most of the omics-based studies on B. subtilis have been conducted on a defined medium or at a single time point or specific growth stage. For the immediate application of gene expression data, both in academic research and industrial applications, studies on gene expression and gene function according to their expression patterns are demanded throughout the growth phase of B. subtilis in a rich medium.
This study uses transcriptome analysis to monitor changes in the gene expression of B. subtilis DB104 while growing in Luria–Bertani (LB) medium. First, the growth curve of B. subtilis DB104 was prepared, and the total number of bacteria and spores was counted. Then, the growth stage was divided based on optical density and sporulation rate, and changes in RNA expression according to phase were confirmed. In addition, the expression patterns of genes were analyzed, and their functions were investigated. The results of a comprehensive gene expression and functional analysis are expected to increase knowledge about the B. subtilis DB104 life cycle.

2. Materials and Methods

2.1. Bacterial Strain and Growth Conditions

B. subtilis DB104 (Δnpr Δapr) was routinely grown in LB broth (10 g/L tryptone, 10 g/L NaCl, 5 g/L yeast extract, and pH 7.5). A single colony of B. subtilis DB104, grown in LB agar, was inoculated into 5 mL of LB broth and incubated at 30 °C overnight with shaking at 250 rpm. Afterward, the overnight culture was transferred into 50 mL of fresh LB broth in 500 mL baffled flasks at an optical density of 0.1 (OD600) and allowed to grow under the same conditions. The cell density was measured by reading the OD600 on a spectrometer at various time points. The number of total cells and endospores was also determined. To count the endospores, an aliquot of the culture was centrifugated, resuspended in an equal volume of 1 mg/mL lysozyme, and incubated at 4 °C for 30 min to kill the vegetative cells.

2.2. RNA Isolation, Library Construction, and RNA-Sequencing

RNA was isolated essentially as described [19,20]. The 1.5 mL of B. subtilis DB104 culture samples were pelleted by centrifugation at the indicated time points (8, 10, 12, 15, 18, and 24 h). The pellets were treated with 1.5 mg/mL lysozyme in TE buffer [21,22]. Subsequently, 200 μL of TRIzol® Max Bacterial Enhancement Reagent (Life Technologies, Grand Island, NY, USA) was added and the samples were incubated at 95 °C for 4 min. The cell suspensions were mixed with 1 mL of RNAiso Plus (Takara, Otsu, Japan) and total RNA was isolated following the manufacturer’s instructions. Libraries were constructed using a NEBNext Bacteria rRNA Depletion Kit (New England Biolabs, Ipswich, MA, USA) and TruSeq Stranded Total RNA Library Prep Gold Kit (Illumina, San Diego, CA, USA), and 101-bp paired-end reads were generated. Sequencing quality control was conducted using FastQC v0.11.7 (http://www.bioinformatics.babraham.ac.uk/projects/fastqc/, accessed on 15 February 2022). After the library was constructed, clean data were obtained from the raw data by removing the adapter and low-quality reads using Trimmomatic 0.38 (http://www.usadellab.org/cms/?page=trimmomatic, accessed on 15 February 2022). The transcriptome sequences were mapped to the B. subtilis subsp. subtilis str. 168 genome (GenBank accession number: AL009126.3) using HTseq version 0.10.0 and Bowtie 1.1.2 software. All raw transcriptome data are publicly available in the NCBI Sequence Read Archive (https://ncbi.nlm.nih.gov/sra, accessed on 13 February 2023) database under the accession number PRJNA934277.

2.3. Analysis of DEGs

The expression levels of each gene were calculated using the fragments per kilobase of transcript per million fragments mapped (FPKM) value, based on the mapped genes and using Bowtie software. Next, the differentially expressed genes (DEGs) were extracted using edgeR software with a p-value < 0.05 and |fold change| ≥ 2. DEGs were obtained from five compared groups (10 h/8 h, 12 h/10 h, 15 h/12 h, 18 h/15 h, and 24 h/18 h). Afterward, all DEGs were grouped into 10 K-mean clusters based on the Z-score.

2.4. Functional Analysis of DEGs

All DEGs were enriched through the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (https://www.genome.jp/kegg, accessed on 20 December 2022). Meanwhile, the DEGs from each cluster that exhibited distinct expression patterns were analyzed with the Biological Networks Gene Ontology (BiNGO) tool to identify their function. The DEGs were annotated using then Gene Ontology (GO) database according to p-value < 0.03 and Benjamini–Hochberg-corrected p-value < 0.03.

2.5. Reverse Transcription-Quantitative Polymerase Chain Reaction (RT-qPCR) Confirmation

The primers used for RT-qPCR are listed in Table S2 (Supplementary File S2). A total of 8 genes (cotD, spoIIID, yqfX, spmH, gsiB, frlO, cheY, and gbsB) that were found to be most up- and down-regulated DEGs according to fold change were selected and validated by RT-qPCR. One μg of RNA was used to synthesize cDNA using random hexamer (Roche, Base, Switzerland) and M-MLV reverse transcriptase (Promega, Madison, SI, USA), following the manufacturer’s protocols. RT-qPCR was conducted at 95 °C for 10 s, followed by 40 cycles of 95 °C for 5 s and 60 °C for 30 s using Taq Pro Universal SYBR qPCR Master Mix (Vazyme, Nanjing, China) on a CFX Connect Real-Time System (Bio-Rad, Hercules, CA, USA). The RT-qPCR analysis was conducted in triplicate independently. The target genes were normalized against rpsJ gene as an internal control. The relative expression levels of the target genes were calculated with the 2−ΔΔCt method [23].

3. Results

3.1. Cell Growth and Sporulation

Growth was monitored while cultured in an LB medium to investigate time-related transcriptome changes in B. subtilis DB104. First, the growth curve was monitored by optical density measurements at OD600 (Figure 1). After 12 h, cell density declined for a while and then from 15 h increased again. Next, sporulation was calculated at various times by plate counting (Figure 1) and observed using an optical microscope (Supplementary File S1: Figure S1). Sporulation was approximately 1% until 12 h, and then rapidly progressed to 18% at 15 h. After 15 h, the sporulation rate constantly increased to 90% at 24 h. Therefore, B. subtilis DB104 growth could be divided into two parts: exponential growth and sporulation with a transition point at 12 h.

3.2. RNA Sequencing and Analysis of Differentially Expressed Genes (DEGs)

Cell samples from six-time points were obtained based on the growth curve: the middle of exponential growth (8 h), the end of exponential growth (10 h), the beginning of sporulation (12 h), the former part of sporulation (15 h), the latter part of sporulation (18 h), and the end of sporulation (24 h). At each time point, the total RNAs were extracted from three independent cultures and used for RNA-seq. The transcriptome sequences were mapped against the B. subtilis subsp. subtilis str. 168 reference genome (GenBank accession number: AL009126.3). A summary of the resulting sequence data is shown in Table S1 (Supplementary File S2). The filtered clean reads ranged from 21,238,576 to 35,756,134. The Q20 values were greater than 98.19%, Q30 values were greater than 94.16%, and the GC contents were greater than 44.58% in each sample.
To ensure the reliability of the transcriptome analysis, multidimensional scaling and the Pearson correlation coefficient were used to confirm the differences among all samples (Figure 2a,b). According to the results, the 10 h and 12 h groups had similar RNA expression patterns. The 15 h, 18 h, and 24 h groups were similar, whereas the 8 h group was relatively separated from the other groups. This suggests that the transcription profile of B. subtilis DB104 was altered, depending on the growth stage.
With a p-value < 0.05 and |fold change| ≥ 2 as the criteria, a total of 2122 DEGs were identified in all the comparison groups. The numbers of DEGs identified between the various time points during the growth of B. subtilis DB104 were analyzed using a Venn diagram (Figure 2c). The number of genes that were up- or down-regulated in each compared group is shown in Figure 2d. The results show that more transcriptome changes occurred during exponential growth to the former part of sporulation than the latter part and end of sporulation. In general, the number of up-regulated DEGs was higher than that of down-regulated DEGs.

3.3. DEGs with the Highest Expression Levels and Fold Changes

To identify the most highly expressed genes, expression levels at all time points were calculated based on transcripts per millions mapped reads (TPM). The 10 most highly expressed DEGs are listed in Table 1. Genes encoding various spore coat proteins, such as cotX, cotY, and cotZ, showed high TPM values. Other extreme TPM values were observed for genes related to the spore of B. subtilis, such as gerE, safA, cgeA, and cgeB. Most of these genes were strongly expressed after 12 h. Among the genes listed in Table 1, only sdpC and skfA were highly expressed during exponential growth, and their expressions were down-regulated after 12 h. The DEGs with the highest fold-change values were also confirmed, and the top 10 of them are listed in Table 2. The most up-regulated genes were mostly related to sporulation and germination, such as the spore coat proteins cotD and cotG and the germination proteins yqfX and yqzD. In contrast, the most down-regulated genes were associated with a higher variety, such as response to stress (gsiB and gabT), chemotaxis (cheY), and metabolism (gabT, gbsB, gbsA, and xpt). Furthermore, the down-regulated genes showed a much smaller fold-change value than the up-regulated genes. The highest expression changes did not appear after 18 h. Altogether, these results show that sporulation-related genes are highly expressed and regulated, and sharp changes in gene expression mainly occur up to the former part of sporulation (15 h) during B. subtilis DB104 growth.

3.4. KEGG Pathway Enrichment Analysis

KEGG pathway analysis was performed to elucidate changes in pathways. DEGs in each compared group were divided into two groups consisting of up- or down-regulated cases and were analyzed separately. The total number of DEGs involved in each pathway was calculated. Table 3 shows the top 10 KEGG pathways with the number of DEGs involved. The DEGs were related to metabolic pathways (bsu01100), microbial metabolism in diverse environments (bsu01120), biosynthesis of amino acids (bsu01230), and butanoate metabolism (bsu00650) in both cases, which suggests that metabolic pathway changes are significant throughout growth. In addition, it was noticed that the genes involved in ABC transporters (bsu02010) were down-regulated up to the beginning of sporulation (12 h) and up-regulated from the latter part of sporulation (18 h).

3.5. Cluster Analysis

Based on the Z-score, a K-mean cluster analysis was performed to further analyze the DEG expression patterns further. Ten distinct clusters were obtained, and their expression levels across growth were presented by a heatmap (Figure 3). Each cluster exhibits various expression patterns over time; in cluster 1, gene expression was decreased from 8 to 12 h, while it was increased thereafter until being equal to 8 h (n = 111 genes); in cluster 2, expression sharply decreased at 10 h and remained at that level (n = 177 genes); in cluster 3, gene expression increased slightly at 10 h and decreased with a nadir at 15 h, while it was slightly recovered thereafter (n = 133 genes); in cluster 4, the expression of the included genes was down-regulated to 12 h and then remained at a similar level (n = 265 genes); in cluster 5, the expression increased in a time-dependent manner with the zenith at 15 h (n = 336 genes); in cluster 6, 7, and 9, gene expression was up-regulated from 8 to 12 h, and then decreased at 15 h (n = 221, 330, and 116, respectively); in cluster 8, expression levels increased in a time-dependent manner with maximum expression at 18 h (n = 116 genes); and in cluster 10, genes were expressed in a zig-zag manner with the deepest parts at 12 and 18 h and the highest point at 15 h (n = 86 genes). The genes belonging to each cluster are listed in Supplementary File S3.

3.6. Functional Enrichment Analysis of Each Cluster

Gene ontology (GO) enrichment analysis was conducted to confirm the biological function of genes belonging to each cluster that showed different expression patterns over time. The genes in each cluster were classified into biological process (BP), molecular function (MF), and cellular component (CC). The GO terms that were significantly enriched and their relationships are shown in Tables S3–S11 (Supplementary File S2) and Figures S2–S10 (Supplementary File S1).

3.6.1. Genes That Are Down-Regulated at the End of Exponential Growth: Cluster 2

The 177 genes in cluster 2 showed the high expression levels in the middle of exponential growth (8 h) but decreased expression from 10 h at the end of exponential growth (Figure 3). DEGs belonging to this cluster were classified into 130 GO terms. The enriched GO terms are listed in Table S4 (Supplementary File S2), and their relationship is shown in Figure S2 (Supplementary File S1). Among them, the terms related to BP were the most common with 96, and mostly consisted of various metabolic processes (GO:0008152), such as ribonucleotide metabolic process (GO:0009259), amine catabolic process (GO:0009310), and organic acid metabolic process (GO:0006082). Terms, such as signaling (GO:0023052), secretion (GO:0046903), locomotion (GO:0040011), and localization (GO:0051179), were also found to be significantly enriched terms. Sixteen GO terms were related to CC and associated with the flagellum (GO:0019861).

3.6.2. Genes That Are Down-Regulated from the Beginning of Sporulation: Clusters 3 and 4

Genes belonging to clusters 3 and 4 were expressed at a low level from 12 h when sporulation begins to 24 h when sporulation ends (Figure 3). Among them, 133 DEGs belonging to cluster 3 were classified into 90 GO terms. The enriched classified GO terms are listed in Table S5 (Supplementary File S2), and their relationship is shown in Figure S4 (Supplementary File S1). The 79 BP terms include subterms of metabolic processes, such as cellular amino acid biosynthetic process (GO:0008652) and cellular metabolic process (GO:0044237), and subterms of developmental process, such as sporulation (GO:0043934) and cell differentiation (GO:0030154). In addition, GO terms, such as urea metabolic process (GO:0019627), secretion (GO:0046903), response to stimulus (GO:0050896), and cell communication (GO:0007154), were also revealed as significantly enriched terms. The 256 DEGs of cluster 4 were significantly enriched in 69 BP, 25 MF, and 15 CC terms. The enriched terms are listed in Table S6 (Supplementary File S2), and their relationship is shown in Figure S5 (Supplementary File S1). The DEGs enriched in BP were related to tricarboxylic acid cycle (GO:0006099), chemical homeostasis (GO:0048878), chemotaxis (GO:0006935), response to stimulus (GO:0050896), and localization (GO:0051179). In addition to protein complex (GO:0043234) and extracellular region (GO:0005576), the DEGs enriched in CC were especially related to flagellum (GO:0019861).

3.6.3. Genes That Are Turned on at the Beginning of Sporulation and then Turned off in the Middle: Clusters 5, 6, 7, and 9

Four out of ten clusters showed a pattern where expression decreased from 15 h or 18 h (middle of sporulation). Among them, clusters 6 and 7 showed similar expression patterns, with expression at a low level in the exponential phase and then increased until sporulation began; however, differences in expression level were observed after 15 h. The 221 DEGs belonging to cluster 6 were classified into only 21 BP terms. The 330 DEGs belonging to cluster 7 were classified into 30 BP, 8 MF, and 2 CC terms. The enriched terms in each cluster are listed in Tables S8 and S9 (Supplementary File S2), and their relationships are shown in Figures S7 and S8 (Supplementary File S1), respectively. The BP-enriched DEGs of clusters 6 and 7 were mainly related to developmental process (GO:0032502), such as sporulation (GO:0043934) and cell differentiation (GO:0030154), and some other metabolic processes. The expression pattern of cluster 9 differed in that it had a high expression level from exponential growth, although it decreased from 15 h. As a result of annotating 116 DEGs belonging to cluster 9 into the GO database, there was no significant term that satisfied the p-value < 0.03, and the Benjamini–Hochberg-corrected p-value < 0.03. Unlike other clusters, DEGs of cluster 5 increased expression up to 15 h and then decreased from 18 h. The 336 DEGs belonging to cluster 5 were classified into nine BP terms and one CC term. The enriched terms are listed in Table S7 (Supplementary File S2), and their relationship is shown in Figure S6 (Supplementary File S1). BP terms were focused on developmental processes (GO:0032502), such as sporulation (GO:0043934) and cell differentiation (GO:0030154).

3.6.4. Genes That Are Up-Regulated in the Middle of Sporulation: Clusters 1 and 8

Genes belonging to cluster 1 were down-regulated at 12 h, but their expression increased from 15 h, the former part of the sporulation (Figure 3). The 111 DEGs of cluster 1 were classified into 53 GO terms. The enriched terms are listed in Table S3 (Supplementary File S2), and their relationship is shown in Figure S2 (Supplementary File S1). In cluster 1, most DEGs were enriched in BP terms, including metabolic processes such as tryptophan metabolic process (GO:0006568) and lipoteichoic acid biosynthetic process (GO:0070395) in addition to cell wall organization or biogenesis (GO:0071554), localization (GO:0051179), and small molecule metabolic process (GO:0044281). Of the seven MF terms, terms related to transporter activity (GO:0005215) were the most common, appearing in four. Terms enriched in this cluster were classified with relatively high p-values. In contrast, terms classified as enriched in cluster 8 had low p-values and were concentrated in developmental processes (GO:0032502). DEGs belonging to cluster 8 steadily increased expression from the beginning to the latter part of sporulation. The 347 genes belonging to this cluster were enriched in 15 BP terms. The enriched terms are listed in Table S10 (Supplementary File S2), and their relationship is expressed in Figure S9 (Supplementary File S1). In addition to the subterm of the developmental process (GO:0032502), these terms were also found to be significant in terms related to the organic cation transport (GO:0015695).

3.6.5. Genes That Are Transiently Turned on at the Former Part of Sporulation: Cluster 10

The genes belonging to cluster 10 were specifically turned on at 15 h (the former part of sporulation) and then down-regulated. The enriched terms are listed in Table S11 (Supplementary File S2), and their relationship is shown in Figure S10 (Supplementary File S1). The 86 DEGs in this cluster were enriched mainly for BP terms related to metabolic process, such as lipid catabolic process (GO:0016042) and organic acid catabolic process (GO:0016054), or monosaccharide transport (GO:0015749).

3.7. Sigma Factors

Various alternative sigma factors exist in B. subtilis and are transcribed by recognizing the promoter of the specialized gene set. By analyzing the expression patterns of genes regulated by each alternative sigma factor, their expression patterns were confirmed with clusters revealed through K-mean cluster analysis. The genes regulated by each sigma factor were classified using the DBTBS database [24]. σA is a housekeeping sigma factor that regulates gene expression during the exponential phase and early sporulation in B. subtilis. The gene that encodes σA, sigA, was expressed highly during whole growth, especially at 12 h, where sporulation started (Table 4). Genes controlled by σA were found to be distributed across all clusters. In B. subtilis, σB is a sigma factor that regulates various general stress genes. Clusters 6, 7, and 9, which have the highest expression at the transition point of 12 h, commonly contain many genes controlled by this sigma factor. However, the gene that encodes σB, sigB, was expressed constantly during growth (Table 4). σD is a sigma factor that regulates genes related to flagellar gene expression, chemotaxis, autolysis, and motility in B. subtilis. Cluster 2, whose expression was suppressed at the end of exponential growth, contained many genes controlled by σD, including sigD. sigD (the gene for σD) is a component of the fla-che operon, which consists of more than 30 genes and can self-activate. Many genes comprising this operon were included in cluster 2, including the genes encoding the rod protein flgC and the hook proteins flgE, cheA, cheB, and cheW, which comprise the flagellar of B. subtilis. In addition, flhO and flhP, which do not belong to this operon but encode the proteins that make up the flagellar rod, were also included in this cluster. Cluster 4, which exhibits a pattern where expression decreases as sporulation starts, also contained many genes controlled by σD, similar to cluster 2. Among the fla-che operon genes, genes in cluster 4 included the genes encoding the hook proteins flgL and flgK and the rod protein flgB. In addition, the genes encoding flagellar assembly protein genes fliS and fliT, the flagellin protein hag, and the flagellar stators motA and motB are not part of the fla-che operon and are controlled by σD, belonging to this cluster. Among the genes controlled by σD, genes related to autolysis, such as lytB, lytC, and lytF, were included in cluster 4. Genes regulated by the sigma factor associated with sporulation were included in the cluster with the highest expression at 15 h or 18 h. The genes controlled by σE and σF, which are early sporulation-related sigma factors, were included in cluster 7. The DEGs controlled by σE that regulate early sporulation genes in mother cells were also included in cluster 6. In contrast, genes controlled by σK, a sigma factor related to late sporulation gene expression in the forespore, and σG, sigma factor related to late sporulation gene expression in the mother cell, were predominantly included in clusters 8 and 5, respectively. The genes that encode early sporulation-specific sigma factors, sigE and sigF, were up-regulated at 10 h (Table 4). However, the genes that encode late sporulation-specific sigma factor, sigG, were up-regulated from 10 h. The other late sporulation-specific sigma factor σK, which consists of two truncated genes, sigKc and sigKn [25], are highly expressed from 12 h. General stress protein genes, such as yceD, yceE, yceF, and yceG, are controlled by σW, an extra-cytoplasmic function sigma factor and were included in clusters 4 or 9. The gene for σW, sigW, was also included in cluster 9, and its expression was down-regulated from 15 h (Table 4). Among the regulons of σL, known to be involved in degradative enzyme expression, only the acetoin dehydrogenase operon genes (acoABCL) were classified as cluster 8, and the rest did not belong to any cluster. The gene that encodes σL, sigL, was up-regulated until 10 h and then down-regulated.
Several other factors, such as σI, σM, σX, σY, and YlaA, along with the sigma factors described above, regulate gene expression in B. subtilis. The dhb operon genes (dhbACEBF and mbtH) are controlled by σI and were included in cluster 9. Some flavodoxin genes (fldN, ykuO, and fldP), which are controlled by the same sigma factor, were included in cluster 10. σX regulon has dlt operon genes, such as dltABCDE, belonging to cluster 1 and the efe operon genes (efeUBO) and yce operon genes (yceDEFGH) belonging to cluster 4. In addition, some of the genes controlled by σX belong to cluster 7 or cluster 6. YlaC, known to regulate genes associated with oxidative stress, regulates the yla operon to which it belongs, and genes belonging to this operon (ylaACD) were included in cluster 7 (the ylaB gene in yla operon was not mapped in this study).

3.8. Flagella

Among the movement of bacteria, swimming and swarming are the most representative. B. subtilis, a flagellated bacterium, can swim in liquids and swarm on solid surfaces through flagella. Therefore, the analysis of the flagellar gene could help to understand the mobility of this strain. The expression values of some flagellar structure genes, such as fliO, fliP, flgB, fliD, and flgD, are shown in Figure 4a–c and Table S12 (Supplementary File S2), which were classified as cluster 2 or 4 through K-mean clustering analysis. These genes remained down-regulated from the end of exponential growth (10 h) or the transition point (12 h). On the other hand, yvzB showed a different expression pattern from other flagellar genes, maintaining its expression level until 12 h, when sporulation began, and then down-regulated from the former part of sporulation (15 h).

3.9. Sporulation and Spore Coat

The expression patterns of the gene involved in each sporulation stage are shown in Figure 5. In stage 0, cannibalism delays entering the sporulation process (in the next section). Most genes related to sporulation stage 0 and I were up-regulated at the exponential growth (8 h and 10 h). When septum is formed, many genes expressed in stage II were mostly up-regulated at 10 h (the end of exponential growth). Most of the genes expressed when stage III is completed also showed similar expression patterns to those of stage II but remained at a highly expressed level until 12 h (the beginning of sporulation). Most genes expressed during stages VI and V, in which the cortex and spore coat are formed and assembled, were up-regulated after 12 h, and they remained consistently high until 24 h when sporulation was completed.
After spore engulfment was completed, σE first induced the expression of the spore coat proteins in the mother cell. The expression value of representative spore coat proteins of B. subtilis is shown in Figure 6 and Table S12 (Supplementary File S2). The σE-induced spore coat protein genes (cotE, cotJA, cotJB, and cotJC) and spore morphogenic genes (spoIVA and spoVID) became up-regulated from 10 h at the end of exponential growth (Figure 6; the cotE gene was not mapped in this study). Afterwards, most of the spore coat protein genes, such as cotA, cotD, cotF, cotH, cotM, cotT, cotV, cotW, cotY, cotZ, cotB, cotG, cotS, and cotX, and the spore coat gene transcription regulator gene gerE were up-regulated by σK from 15 h, the former part of sporulation (Figure 6).

3.10. Cannibalism

At the beginning of sporulation in B. subtilis (stage 0), cannibalistic behavior occurs to delay entering this energy-consuming process. The skf (sporulation killing factor) and sdp (sporulation delay protein) operons become involved in cannibalism. Among the skf operon genes, skfA, which encodes the sporulation killing factor itself, was strongly expressed in 10 h at the end of exponential growth and 12 h at the beginning of sporulation then decreased (Figure 4d). However, except for skfA, the remaining skf operon genes showed a different pattern in which expression decreased from 12 h. In addition, the expression of those genes was significantly lower than that of skfA (Supplementary File S2: Table S13). Among the sdpABC operon, only the sdpC gene belonged to DEGs. Its expression was highest at 10 h and decreased sharply from 12 h when sporulation began (Figure 4e). Conversely, the expression of the sdpRI immunity operon continued to be up-regulated from 12 h to 24 h. The sdpA and sdpB genes of the sdpABC operon and the sdpI and sdpR genes of the sdpRI operon showed very-low expression values compared to sdpC (Supplementary File S2: Table S13).

3.11. RNA-Seq Data Validation by RT-qPCR

To validate the accuracy of transcriptome data, RT-qPCR analysis was conducted. Table 2 shows the top 10 up- and down-regulated DEGs according to fold change. Therefore, eight genes included in Table 2 (cotD, spoIIID, yqfX, spmH, gsiB, frlO, and cheY) were selected for validation by RT-qPCR. RNA samples from six-time points were used as templates. The results are shown in Figure 7, indicating that the data from RNA-seq and RT-qPCR analysis were consistent.

4. Discussion

This study investigated gene expression changes in B. subtilis DB104 during growth in a LB complete medium through RNA-seq. The simultaneous application of several analysis methods (DEG analysis, KEGG analysis, K-mean cluster analysis, GO analysis, and sigma factor function analysis) allowed the expression patterns of genes and their function to be revealed. We described gene expression changes using RNA-seq analysis, providing genome-wider information compared to using microarray analysis [6,7].
When bacterial growth becomes limited due to nutrient depletion, exponential growth ceases and the bacteria enter the stationary phase. The period between these two phases is called a transition point [26,27]. This transition point has been also observed in B. subtilis, and various metabolic pathway changes occur here [6,7]. In the present study, 12 h was found to be the transition point at which exponential growth ceases and the stationary phase starts (Figure 1). In addition, most of the highest fold changes in DEGs were found at this time point (Table 2). In addition, KEEG analysis revealed that alterations occurred in metabolic pathways around the transition point. The metabolic pathway (bsu01100) was up-regulated until 12 h, and some of the DEGs involved in this pathway started to become down-regulated from 15 h (Table 3). Previous studies showed that the metabolic rate decreased when the stationary phase started in the growth of B. subtilis [28,29]. The other enriched metabolic pathways, such as microbial metabolism in diverse environments (bsu01110) and the biosynthesis of secondary metabolites (bsu01110), were up-regulated to 12 h.
ABC transporters are responsible for transporting a wide range of substrates, such as amino acids, sugars, and organic acids, across the cell membrane through ATP hydrolysis and can perform various functions. Among the pathways enriched by KEGG analysis, only the DEGs involved in ABC transporters (bsu02010) were down-regulated before the transition point and subsequently up-regulated. A possible reason for this may be that a specific ABC transporter is required to initiate sporulation. The ABC transporter gene ftsEX, whose transcription levels were down-regulated at 10 h in this study (Supplementary File S2: Table S13), plays a role in spore formation initiation [30]. In addition, the other ABC transporter genes oppA, oppB, oppC, oppD, and oppF, which are also involved in the initiation of sporulation [31,32], were down-regulated at 12 h (Supplementary File S2: Table S13). The fact that this system is related to stress responses, such as osmotic stress, could be another reason. When bacteria reach the stationary phase, they undergo various metabolism changes and become resistant to several stresses [7,33]. Corresponding to this, the genes for the osmoprotectant ABC transporters opuBA, opuBB, and opuBC were up-regulated at 18 h but were down-regulated at 12 h.
Diverse transcription regulators exist in B. subtilis. Among them, various alternative sigma factors, which allow RNA polymerase to recognize the promoter sequence rapidly and specifically, are essential for sporulation, stress response, chemotaxis, and motility [34]. Therefore, the sigma factors are a significant tool for understanding the gene expression of B. subtilis. To identify the different expression patterns of DEGs and their functions, K-mean clustering was coupled with GO and sigma factor analysis. In this study, the DEGs were divided into 10 clusters whose expression profiles could be redivided into five patterns in accordance with growth stage. Because gene expression was examined according to six-time points, the 10 clusters were divided again, as described above (Section 3.6).
The first group of clusters, which contains only cluster 2, has a pattern specifically expressed only in the middle of exponential growth. The DEGs belonging to cluster 2 were enriched in most terms. The relationship between them was also complicated, as shown in Figure S3 (Supplementary File S1), indicating that genes for maintaining various biological functions are highly expressed in exponential growth and subsequently turned off. Genes transcribed by σA- or σD-dependent RNA polymerase were the most prevalent among the 177 DEGs in cluster 2. B. subtilis σA, similar to its E. coli counterpart σ70, is the primary sigma factor involved in the transcription of most housekeeping genes [35]. This sigma factor also participates in the transcription of the early sporulation genes [36,37] and stress-inducible genes [38,39]. It has been found that the expression of sigA (gene for σA) remains constantly high during the whole growth of B. subtilis DB104 (Table 4). In addition, the primary sigma factor, σA in B. subtilis, usually transcribes genes with other sigma factors [40,41]. This could be why σA-dependent genes were evenly distributed across all clusters in the transcriptome data. Many genes included in clusters 1 to 10 were transcribed by σA-dependent promoter. However, this result does not correspond with that of a previous study, which found that the expression of σA-dependent genes is turned off in the stationary phase [6]. In contrast, Blom et al. [7] showed results consistent with this research and observed that the SigA regulon is overrepresented in the exponential growth phase but could be expressed after the transition point. Furthermore, results from this study suggest that σA-dependent genes have various expression patterns because σA works both alone and jointly with other sigma factors. B. subtilis σD controls genes related to flagellar [42], autolysis [43], chemotaxis, and motility [44]. σD-dependent genes were included predominantly in cluster 2 in addition to cluster 4 in the second cluster group (genes that are down-regulated from the beginning of sporulation). With corresponding data, the GO term “flagellum (GO:0019861)” was enriched in clusters 2 and 4. As previously stated, yvzB, the flagella structure gene, began to decline after the transition point, but remained highly expressed until sporulation began (Figure 4a–c). This may be because this gene is a putative filament gene, and it is unknown whether it contains the proteins that make up the filament structure. Mukherjee and Kearns [45] suggested that flagella synthesized in a mother cell could function in a granddaughter cell. The results from this study showed that flagellar structural genes are down-regulated during the stationary phase in B. subtilis DB104, but further research is needed to determine whether this weakens the swarming/swimming mobility. Meanwhile, among the genes controlled by σD, autolysis-related genes were included in cluster 4 rather than cluster 2, indicating that autolysin expression continued later than that of flagella proteins. The sigD (gene for σD) was also included in cluster 2, and the expression level was down-regulated from 10 h (Table 4). This result indicates that σD produced in the middle of exponential growth remains and functions afterward. The DEGs belonging to clusters 3 and 4 showed a high expression level in exponential growth, similar to genes belonging to cluster 2. However, the difference is that they maintained a high expression afterward and then showed a low expression from the beginning of sporulation. They were also classified as diverse and complex terms (Tables S5–S7 in Supplementary File S2, Figures S4 and S5 in Supplementary File S1). Therefore, these results show that genes for maintaining various biological functions are highly expressed during exponential growth, and some are up-regulated even after exponential growth is over.
An extracellular RNA polymerase sigma factor of B. subtilis, σW, is involved in the response to cell envelope stress, such as to antimicrobial compounds [46,47]. σB is the general stress sigma factor of B. subtilis and is also involved in the transcription of general stress proteins induced by heat, salt, and oxygen stress [48]. σW-dependent genes were distributed in clusters 4 and 9, and σB-dependent genes were distributed in clusters 6, 7, and 9. Except for cluster 4, these clusters belong to the third group of clusters, which contains clusters 5, 6, 7, and 9 (genes that are turned on at the beginning of sporulation and then turned off in the middle). The genes for these stress-related sigma factor, sigB and sigW, were also up-regulated until 12 h and then down-regulated from 15 h (Table 4). The time point 12 h, where sporulation starts, is also a transition point in this study (Figure 1), and it was found that, if the growth of B. subtilis enters the transition point, stress responses may occur, even if the stresses are not induced from outside factors [7,49]. This study supports this view, showing that the σW- or σB-dependent genes that respond to stress are primarily turned on at the beginning of sporulation. Interestingly, cluster 4, whose expression was decreased from the transition point, also contains σW-dependent genes: iron transporter genes from the efeUOB operon [50], general stress genes from the yceCDEFGH operon [51], and phage shock protein gene and unknown function genes from the pspA-ydjGHI operon [52]. It can be assumed that these genes have different expression patterns among the stress response genes because of the growth conditions used in the study. The results show that a spontaneous stress response may occur at the transition point, but even a general stress gene may not be induced at this time.
For the spore-forming bacteria B. subtilis, nutrition limitation or high population density can trigger the complex process of sporulation [53]. During the growth of B. subtilis in this study, sporulation started from 12 h and progressed rapidly up to 18 h and then proceeded to 90% in 24 h (Figure 1). As expected, the GO term “sporulation (GO:0043934)” was enriched in clusters that belong to the third group, clusters 5, 6, and 7. Furthermore, in the GO analysis of these clusters, sporulation was significantly enriched with a relatively low p-value. Consistent with this, σE, σF, σG, and σK, which are related to gene expression during sporulation, are also included in clusters 6 and 7. The sporulation of this strain could be described as eight stages (stages 0–VII) based on morphological features [54,55]. In this study, we compared these sporulation stages with transcriptomic expression changes and analyzed them with the growth stage (Figure 5). Stages 0 and I are vegetative growth and axial filamentation, respectively. However, they are no longer distinguishable because of any mutant arrests at this stage [55], so we considered them together. In these stages, the cells start the sporulation with cannibalistic behavior (see the discussion below). The master regulator Spo0A is essential in transitioning from vegetative growth to the sporulation stage. The activation via the phosphorylation process from KinA/KinB to Spo0F, Spo0B, and Spo0A of the spo0A gene transcribed by the σH-dependent promoter, the first activated sigma factor, signals the start of sporulation. In addition, phosphatase genes, such as the Rap family (rapA, B, E, and H) [56] and Spo0E [57], the cell division protein gene racA that helps chromosomes properly segregate [58], and the genes like sirA [59] and sda [60] that encode proteins for regulating the appropriate chromosome number, are expressed at these stages. All their expression levels were high at 8 h and/or 10 h, indicating that stages 0–I correspond to those before spores are formed. The activated Spo0A, Spo0A~P, regulates the spoIIA operon, which starts stage II. Genes, such as phosphatase gene spoIIE that dephosphorylates SpoIIAA [61], cell-division protein ftsZ that is required for septum formation [62], mother cell-specific sigma factor gene sigE, and protease gene spoIIGA that activates Pro-SigE [63], are expressed in stage II, including genes of spoIIA operon (anti-anti-sigma factor gene spoIIAA, anti-sigma factor gene spoIIAB, and forespore-specific sigma factor gene sigF). All of these genes expressed highly at 12 h, showing that stage II, at which asymmetric division occurs, matched the end of exponential growth. At stage III, the cell-wall hydrolase genes spoIID and spoIIP that are required for engulfment [64,65] are expressed. The membrane fission protein gene fisB, the spoIIQ gene, and the spoIIIA operon gene (spoIIIAAspoIIIAH) that are required for the activation of σG [66] are also transcripted at this stage, where engulfment is processed. Their up-regulated expressed level from 10 h to 12 h indicates that stage III spans the end of exponential growth and the beginning of sporulation. Cortex and spore coat are produced and assembled at stages VI and V. Genes, such as spore coat morphogenetic protein gene spoIVA, DNA recombinase gene spoIVCA that is required for σK gene, and mother cell-specific sigma factor gene sigKc/sigKn, are related to cortex and spore coat. Most of these genes were highly expressed after 15 h (the formal part of sporulation), but some were up-regulated from 10 h (end of exponential growth). This result shows that the genes involved in stages VI and V could be expressed earlier. So, we decided to discuss spore coat genes in depth.
σE and σF are the sigma factors related to early sporulation genes in the mother cell and forespore, respectively. At the same time, σK and σG are the sigma factors related to late sporulation genes in the mother cell and forespore, respectively. Additionally, many σK-dependent genes were also included in cluster 8 of the fourth cluster group (genes that are up-regulated in the middle of sporulation). Notably, the expression of spore coat genes was clearly distinguished according to which sigma-factor-dependent RNA polymerase transcribes (Figure 6). The B. subtilis spore coat consists of two layers, an inner coat and outer coat. According to the sporulation process, the spore coat proteins are sequentially expressed in the mother cell [67]; however, it is not completely clear how many proteins make up the spore coat and how they are assembled. However, the results show that, no matter where the proteins exist, their transcription is affected only by σE or σK, and there is no difference in the expression time point between them.
Cluster 8, where the GO term “acetoin metabolic process (GO:0045149)” was enriched (Supplementary File S2: Table S10), included some of the genes that are controlled by σL, the acetoin dehydrogenase operon genes acoABCL. σL is known to be involved in degradative enzyme gene expression [68]. The acetoin is one of the extracellular products of B. subtilis [69]. Since it is reused as a carbon source after the cells enter to stationary phase and sporulation [70,71], the operon genes are up-regulated after 15 h. On the other hand, other genes belonging to σL regulon were not classified into any cluster, and even sigL (gene for σL) was down-regulated from 12 h (Table 4). Similar to σL, the regulons of other alternative sigma factors in B. subtilis, such as σI, σM, σX, σY, and YlaA, did not show a consistent expression pattern (Supplementary File S1: Figure S11). Moreover, not only were the TPM values of sigI, sigM, sigX, sigY, and ylaA (gene for σI, σM, σX, σY, and YlaA, respectively) much lower than those of other sigma factors (Table 4), there were also fewer genes belonging to their regulons. These results suggest that these sigma factors are minor compared to the other sigma factors described above (σA, σB, σD, σE, σF, σG, σK, and σW), and the role of other transcription regulators is essential for the regulation of the genes controlled by them.
Of particular interest, GO terms related to the lipid catabolic process were enriched only in cluster 10, which has a unique expression pattern. From these results, the genes related to the lipid catabolism of B. subtilis are expressed specifically throughout the entire sporulation when grown in a rich medium. The programmed cell death (PCD) system could be the reason for this specific expression. In B. subtilis, as in other eukaryotes or prokaryotes, a PCD system exists to delay sporulation, an energy-consuming and irreversible process [72]. The step-by-step expression of the gene involved in this system was well identified in our data. As shown in Figure 4c,d, the expression of skfA and sdpC, which play an essential role in the PCD system, peaked at 12 h and 10 h, respectively. In the Spo0A-active cell, genes encoded in the skf operon are expressed, and killing factors are produced and exported [73,74]. In previous transcriptome analysis conducted in LB with glucose [6], where sporulation was suppressed, there was no skfA expression changes during B. subtilis growth, indicating the PCD system occurs with spore formation. On the other hand, in both the Spo0A-active and -inactive cells, the sdp operon is turned on. SdpC (product of sdpC), a constituent protein of the sdp operon, increases the expression of the fadNAE operon, which encodes lipid catabolism enzymes through SdpR, which delays sporulation [75]. The sdpR and fadNAE operon gene expressions peaked at 12 h and 15 h, respectively (Supplementary File S2: Table S13). Therefore, it can be assumed that regulating the lipid catabolic process through the cascade of this sequential expression prevents sporulation triggers caused by a lack of energy.

5. Conclusions

This transcriptome study provided a novel insight into the dynamic transcriptional response of each gene in B. subtilis DB104 during growth. The highest expressed genes and fold changes were identified around the transition point. Furthermore, the gene expression patterns were identified and DEG functions of were established through K-mean cluster analysis, GO enrichment analysis, the roles of alternative sigma factors, and their simultaneous application. The spore formation process is the most regulated in B. subtilis DB104, and most transcriptomic and metabolic changes occur with the entry of sporulation, named “the transition point”. Because sporulation is an essential program in this organism, many processes during growth, such as metabolism changes, stress-induced responses, and cannibalism, overlap with sporulation. The gene expression involved in each sporulation stage was compared and matched with a growth curve. Additionally, this research showed that the timing of expression of the spore coat gene is determined only by the sigma factor, regardless of its location. Our results also suggest that, among the sigma factors of B. subtilis, σA, σB, σD, σE, σF, σG, σK, and σW play significant roles. Knowledge of transcriptional changes during the growth of B. subtilis DB104 will help to elucidate the expression and regulatory mechanisms of genes under various growth conditions and for the industrial application of this strain.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms11081928/s1. Supplementary File S1. Figure S1: Observation of sporulation by optical microscopy at each time point, Figure S2: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 1, Figure S3: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 2, Figure S4: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 3, Figure S5: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 4, Figure S6: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 5, Figure S7: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 6, Figure S8: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 7, Figure S9: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 8, Figure S10: Gene ontology (GO) term networks of differentially expressed genes involved in cluster 10, Figure S11: Expression patterns of the genes included in each regulon. Supplementary File S2; Table S1: Summary of RNA-seq and the reads mapped to the genome of Bacillus subtilis subsp. subtilis str. 168, Table S2: Primers used for RT-qPCR analysis. Table S3: Gene ontology (GO) enrichment analysis of cluster 1, Table S4: Gene ontology (GO) enrichment analysis of cluster 2, Table S5: Gene ontology (GO) enrichment analysis of cluster 3, Table S6: Gene ontology (GO) enrichment analysis of cluster 4, Table S7: Gene ontology (GO) enrichment analysis of cluster 5, Table S8: Gene ontology (GO) enrichment analysis of cluster 6, Table S9: Gene ontology (GO) enrichment analysis of cluster 7, Table S10: Gene ontology (GO) enrichment analysis of cluster 8, Table S11: Gene ontology (GO) enrichment analysis of cluster 10, Table S12: Transcripts per millions mapped reads (TPM) values of DEGs involved in each structure, Table S13: Transcripts per millions mapped reads (TPM) values of DEGs involved in each operon. Supplementary File S3; List of the differentially expressed genes in clusters 1 to 10.

Author Contributions

Conceptualization, J.-S.J. and K.-W.H.; Data curation, J.-S.J. and K.-W.H.; Formal analysis, J.-S.J. and H.-E.J.; Investigation, J.-S.J., S.-Y.M. and S.-H.S.; Writing—original draft, J.-S.J.; Writing—review and editing, J.-S.J. and K.-W.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financially supported by the National Research Foundation Korea (NRF-2021R1I1A2046284).

Data Availability Statement

The datasets supporting the conclusions of this article are included within the article as well as their additional files. The raw sequence data have been submitted to the Sequence Read Archive (SRA, https://www.ncbi.nlm.nih.gov/sra, accessed on 13 February 2023) under Bioproject accession number PRJNA934277 and reference Biosample accession number SAMN33269663—80.

Conflicts of Interest

The authors declare no conflict to interest.

References

  1. Terpe, K. Overview of bacterial expression systems for heterologous protein production: From molecular and biochemical fundamentals to commercial systems. Appl. Microbiol. Biotechnol. 2006, 72, 211–222. [Google Scholar] [CrossRef] [PubMed]
  2. EPA. Attachment I-Final Risk Assessment of Bacillus subtilis; United States Environmental Protection Agency: Washington, DC, USA, 1997. [Google Scholar]
  3. Kawamura, F.; Doi, R.H. Construction of a Bacillus subtilis double mutant deficient in extracellular alkaline and neutral proteases. J. Bacteriol. 1984, 160, 442–444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Patten, C.; Kirchhof, M.; Schertzberg, M.; Morton, R.; Schellhorn, H. Microarray analysis of RpoS-mediated gene expression in Escherichia coli K-12. Mol. Genet. Genom. 2004, 272, 580–591. [Google Scholar] [CrossRef] [PubMed]
  5. Kitichalermkiat, A.; Katsuki, M.; Sato, J.; Sonoda, T.; Masuda, Y.; Honjoh, K.-I.; Miyamoto, T. Effect of epigallocatechin gallate on gene expression of Staphylococcus aureus. J. Glob. Antimicrob. Resist. 2020, 22, 854–859. [Google Scholar] [CrossRef] [PubMed]
  6. Yang, C.-K.; Tai, P.C.; Lu, C.-D. Time-related transcriptome analysis of B. subtilis 168 during growth with glucose. Curr. Microbiol. 2014, 68, 12–20. [Google Scholar] [CrossRef]
  7. Blom, E.-J.; Ridder, A.N.; Lulko, A.T.; Roerdink, J.B.; Kuipers, O.P. Time-resolved transcriptomics and bioinformatic analyses reveal intrinsic stress responses during batch culture of Bacillus subtilis. PLoS ONE 2011, 6, e27160. [Google Scholar] [CrossRef] [Green Version]
  8. Finotello, F.; Di Camillo, B. Measuring differential gene expression with RNA-seq: Challenges and strategies for data analysis. Brief. Funct. Genom. 2015, 14, 130–142. [Google Scholar] [CrossRef]
  9. Ozsolak, F.; Milos, P.M. RNA sequencing: Advances, challenges and opportunities. Nat. Rev. Genet. 2011, 12, 87–98. [Google Scholar] [CrossRef]
  10. Garalde, D.R.; Snell, E.A.; Jachimowicz, D.; Sipos, B.; Lloyd, J.H.; Bruce, M.; Pantic, N.; Admassu, T.; James, P.; Warland, A. Highly parallel direct RNA sequencing on an array of nanopores. Nat. Methods 2018, 15, 201–206. [Google Scholar] [CrossRef]
  11. Al Kadi, M.; Okuzaki, D. Unfolding the bacterial transcriptome landscape using oxford nanopore technology direct RNA sequencing. In Nanopore Sequencing: Methods and Protocols; Springer: New York, NY, USA, 2023; pp. 269–279. [Google Scholar]
  12. Fawcett, P.; Eichenberger, P.; Losick, R.; Youngman, P. The transcriptional profile of early to middle sporulation in Bacillus subtilis. Proc. Natl. Acad. Sci. USA 2000, 97, 8063–8068. [Google Scholar] [CrossRef]
  13. Swarge, B.; Abhyankar, W.; Jonker, M.; Hoefsloot, H.; Kramer, G.; Setlow, P.; Brul, S.; de Koning, L.J. Integrative analysis of proteome and transcriptome dynamics during Bacillus subtilis spore revival. mSphere 2020, 5, e00463-20. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, Q.; Cornilleau, C.; Muller, R.R.; Meier, D.; Flores, P.; Guerin, C.; Wolf, D.; Fromion, V.; Carballido-Lopez, R.; Mascher, T. Comprehensive and comparative transcriptional profiling of the cell wall stress response in Bacillus subtilis. bioRxiv 2023. [Google Scholar] [CrossRef]
  15. Nicolas, P.; Mäder, U.; Dervyn, E.; Rochat, T.; Leduc, A.; Pigeonneau, N.; Bidnenko, E.; Marchadier, E.; Hoebeke, M.; Aymerich, S. Condition-dependent transcriptome reveals high-level regulatory architecture in Bacillus subtilis. Science 2012, 335, 1103–1106. [Google Scholar] [CrossRef] [PubMed]
  16. Van Duy, N.; Mäder, U.; Tran, N.P.; Cavin, J.F.; Tam, L.T.; Albrecht, D.; Hecker, M.; Antelmann, H. The proteome and transcriptome analysis of Bacillus subtilis in response to salicylic acid. Proteomics 2007, 7, 698–710. [Google Scholar] [CrossRef] [PubMed]
  17. Hahne, H.; Mäder, U.; Otto, A.; Bonn, F.; Steil, L.; Bremer, E.; Hecker, M.; Becher, D. A comprehensive proteomics and transcriptomics analysis of Bacillus subtilis salt stress adaptation. J. Bacteriol. 2010, 192, 870–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Xu, S.; Cao, Q.; Liu, Z.; Chen, J.; Yan, P.; Li, B.; Xu, Y. Transcriptomic analysis reveals the role of tmRNA on biofilm formation in Bacillus subtilis. Microorganisms 2022, 10, 1338. [Google Scholar] [CrossRef] [PubMed]
  19. Sullivan, M.J.; Prince, D.; Goh, K.G.; Katupitiya, L.; Gosling, D.; Crowley, M.R.; Crossman, D.K.; Ulett, G.C. Dual RNA sequencing of group B Streptococcus-infected human monocytes reveals new insights into host–pathogen interactions and bacterial evasion of phagocytosis. Sci. Rep. 2023, 13, 2137. [Google Scholar] [CrossRef]
  20. Han, L.-L.; Shao, H.-H.; Liu, Y.-C.; Liu, G.; Xie, C.-Y.; Cheng, X.-J.; Wang, H.-Y.; Tan, X.-M.; Feng, H. Transcriptome profiling analysis reveals metabolic changes across various growth phases in Bacillus pumilus BA06. BMC Microbiol. 2017, 17, 156. [Google Scholar] [CrossRef] [Green Version]
  21. Villa-Rodríguez, E.; Ibarra-Gámez, C.; de Los Santos-Villalobos, S. Extraction of high-quality RNA from Bacillus subtilis with a lysozyme pre-treatment followed by the Trizol method. J. Microbiol. Methods 2018, 147, 14–16. [Google Scholar] [CrossRef]
  22. Verbeelen, T.; Van Houdt, R.; Leys, N.; Ganigué, R.; Mastroleo, F. Optimization of RNA extraction for bacterial whole transcriptome studies of low-biomass samples. iScience 2022, 25, 105311. [Google Scholar] [CrossRef]
  23. Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef] [PubMed]
  24. Sierro, N.; Makita, Y.; de Hoon, M.; Nakai, K. DBTBS: A database of transcriptional regulation in Bacillus subtilis containing upstream intergenic conservation information. Nucleic Acids Res. 2008, 36, D93–D96. [Google Scholar] [CrossRef] [PubMed]
  25. Stragier, P.; Kunkel, B.; Kroos, L.; Losick, R. Chromosomal rearrangement generating a composite gene for a developmental transcription factor. Science 1989, 243, 507–512. [Google Scholar] [CrossRef]
  26. Strauch, M.A.; Hoch, J.A. Transition-state regulators: Sentinels of Bacillus subtilis post-exponential gene expression. Mol. Microbiol. 1993, 7, 337–342. [Google Scholar] [CrossRef] [PubMed]
  27. Funakoshi, H.; Yamada, A. Transition phenomena in bacterial growth between logarithmic and stationary phases. J. Math. Biol. 1980, 9, 369–387. [Google Scholar] [CrossRef]
  28. Chubukov, V.; Sauer, U. Environmental dependence of stationary-phase metabolism in Bacillus subtilis and Escherichia coli. Appl. Environ. Microbiol. 2014, 80, 2901–2909. [Google Scholar] [CrossRef] [Green Version]
  29. Strauch, M.A. Regulation of Bacillus subtilis gene expression during the transition from exponential growth to stationary phase. Prog. Nucleic Acid Res. Mol. Biol. 1993, 46, 121–153. [Google Scholar] [CrossRef]
  30. Garti-Levi, S.; Hazan, R.; Kain, J.; Fujita, M.; Ben-Yehuda, S. The FtsEX ABC transporter directs cellular differentiation in Bacillus subtilis. Mol. Microbiol. 2008, 69, 1018–1028. [Google Scholar] [CrossRef]
  31. Perego, M.; Higgins, C.; Pearce, S.; Gallagher, M.; Hoch, J. The oligopeptide transport system of Bacillus subtilis plays a role in the initiation of sporulation. Mol. Microbiol. 1991, 5, 173–185. [Google Scholar] [CrossRef]
  32. Koide, A.; Hoch, J.A. Identification of a second oligopeptide transport system in Bacillus subtilis and determination of its role in sporulation. Mol. Microbiol. 1994, 13, 417–426. [Google Scholar] [CrossRef]
  33. Kolter, R.; Siegele, D.A.; Tormo, A. The stationary phase of the bacterial life cycle. Annu. Rev. Microbiol. 1993, 47, 855–874. [Google Scholar] [CrossRef] [PubMed]
  34. Shorenstein, R.G.; Losick, R. Purification and properties of the sigma subunit of ribonucleic acid polymerase from vegetative Bacillus subtilis. J. Biol. Chem. 1973, 248, 6163–6169. [Google Scholar] [CrossRef] [PubMed]
  35. Gross, C.; Chan, C.; Dombroski, A.; Gruber, T.; Sharp, M.; Tupy, J.; Young, B. The functional and regulatory roles of sigma factors in transcription. Cold Spring Harb. Symp. Quant. Biol. 1998, 63, 141–156. [Google Scholar] [CrossRef] [PubMed]
  36. Kawamura, F.; Yamashita, S.; Kobayashi, Y.; Takahashi, H.; Saito, H.; Doi, R.H. Mapping of sporulation-deficient suppressor mutations for a catabolite-resistant sporulation (crsA) mutant of the major sigma-43 factor of Bacillus subtilis. J. Gen. Appl. Microbiol. 1989, 35, 43–51. [Google Scholar] [CrossRef] [Green Version]
  37. Price, C.W.; Doi, R.H. Genetic mapping of rpoD implicates the major sigma factor of Bacillus subtilis RNA polymerase in sporulation initiation. Mol. Gen. Genet. 1985, 201, 88–95. [Google Scholar] [CrossRef] [PubMed]
  38. Leelakriangsak, M.; Zuber, P. Transcription from the P3 promoter of the Bacillus subtilis spx gene is induced in response to disulfide stress. J. Bacteriol. 2007, 189, 1727–1735. [Google Scholar] [CrossRef] [Green Version]
  39. Gaballa, A.; Antelmann, H.; Hamilton, C.J.; Helmann, J.D. Regulation of Bacillus subtilis bacillithiol biosynthesis operons by Spx. Microbiology 2013, 159, 2025–2035. [Google Scholar] [CrossRef] [Green Version]
  40. Loewen, P.C.; Hengge-Aronis, R. The role of the sigma factor σS (KatF) in bacterial global regulation. Annu. Rev. Microbiol. 1994, 48, 53–80. [Google Scholar] [CrossRef]
  41. Paget, M.S.; Helmann, J.D. The σ70 family of sigma factors. Genome Biol. 2003, 4, 203. [Google Scholar] [CrossRef] [Green Version]
  42. Helmann, J.D.; Márquez, L.M.; Chamberlin, M.J. Cloning, sequencing, and disruption of the Bacillus subtilis σ28 gene. J. Bacteriol. 1988, 170, 1568–1574. [Google Scholar] [CrossRef] [Green Version]
  43. Blackman, S.A.; Smith, T.J.; Foster, S.J. The role of autolysins during vegetative growth of Bacillus subtilis 168. Microbiology 1998, 144, 73–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Fredrick, K.L.; Helmann, J.D. Dual chemotaxis signaling pathways in Bacillus subtilis: A σD-dependent gene encodes a novel protein with both CheW and CheY homologous domains. J. Bacteriol. 1994, 176, 2727–2735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Mukherjee, S.; Kearns, D.B. The structure and regulation of flagella in Bacillus subtilis. Annu. Rev. Genet. 2014, 48, 319–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Helmann, J.D. Deciphering a complex genetic regulatory network: The Bacillus subtilis σW protein and intrinsic resistance to antimicrobial compounds. Sci. Prog. 2006, 89, 243–266. [Google Scholar] [CrossRef] [PubMed]
  47. Kingston, A.W.; Subramanian, C.; Rock, C.O.; Helmann, J.D. A σW-dependent stress response in Bacillus subtilis that reduces membrane fluidity. Mol. Microbiol. 2011, 81, 69–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Rodríguez Ayala, F.; Bartolini, M.; Grau, R. The stress-responsive alternative sigma factor SigB of Bacillus subtilis and its relatives: An old friend with new functions. Front. Microbiol. 2020, 11, 1761. [Google Scholar] [CrossRef]
  49. Sung, H.-M.; Yasbin, R.E. Adaptive, or stationary-phase, mutagenesis, a component of bacterial differentiation in Bacillus subtilis. J. Bacteriol. 2002, 184, 5641–5653. [Google Scholar] [CrossRef] [Green Version]
  50. Roy, E.M.; Griffith, K.L. Characterization of a novel iron acquisition activity that coordinates the iron response with population density under iron-replete conditions in Bacillus subtilis. J. Bacteriol. 2017, 199, e00487-16. [Google Scholar] [CrossRef] [Green Version]
  51. Petersohn, A.; Brigulla, M.; Haas, S.; Hoheisel, J.D.; Völker, U.; Hecker, M. Global analysis of the general stress response of Bacillus subtilis. J. Bacteriol. 2001, 183, 5617–5631. [Google Scholar] [CrossRef] [Green Version]
  52. Popp, P.F.; Gumerov, V.M.; Andrianova, E.P.; Bewersdorf, L.; Mascher, T.; Zhulin, I.B.; Wolf, D. Phyletic distribution and diversification of the phage shock protein stress response system in bacteria and archaea. mSystems 2022, 7, e01348-21. [Google Scholar] [CrossRef]
  53. Sonenshein, A.L. Control of sporulation initiation in Bacillus subtilis. Curr. Opin. Microbiol. 2000, 3, 561–566. [Google Scholar] [CrossRef] [PubMed]
  54. Ryter, A.; Schaeffer, P.; Ionesco, H. Cytologic classification, by their blockage stage, of sporulation mutants of Bacillus subtilis Marburg. Ann. de L’institut Pasteur 1966, 110, 305–315. [Google Scholar]
  55. Piggot, P.; Coote, J. Genetic aspects of bacterial endospore formation. Bacteriol. Rev. 1976, 40, 908–962. [Google Scholar] [CrossRef] [PubMed]
  56. Diaz, A.R.; Core, L.J.; Jiang, M.; Morelli, M.; Chiang, C.H.; Szurmant, H.; Perego, M. Bacillus subtilis RapA phosphatase domain interaction with its substrate, phosphorylated Spo0F, and its inhibitor, the PhrA peptide. J. Bacteriol. 2012, 194, 1378–1388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ohlsen, K.L.; Grimsley, J.K.; Hoch, J.A. Deactivation of the sporulation transcription factor Spo0A by the Spo0E protein phosphatase. Proc. Natl. Acad. Sci. USA 1994, 91, 1756–1760. [Google Scholar] [CrossRef] [PubMed]
  58. Wu, L.J.; Errington, J. RacA and the Soj-Spo0J system combine to effect polar chromosome segregation in sporulating Bacillus subtilis. Mol. Microbiol. 2003, 49, 1463–1475. [Google Scholar] [CrossRef] [Green Version]
  59. Winterhalter, C.; Stevens, D.; Fenyk, S.; Pelliciari, S.; Marchand, E.; Soultanas, P.; Ilangovan, A.; Murray, H. SirA inhibits the essential DnaA: DnaD interaction to block helicase recruitment during Bacillus subtilis sporulation. Nucleic Acids Res. 2023, 51, 4302–4321. [Google Scholar] [CrossRef]
  60. Rowland, S.L.; Burkholder, W.F.; Cunningham, K.A.; Maciejewski, M.W.; Grossman, A.D.; King, G.F. Structure and mechanism of action of Sda, an inhibitor of the histidine kinases that regulate initiation of sporulation in Bacillus subtilis. Mol. Cell 2004, 13, 689–701. [Google Scholar] [CrossRef]
  61. Carniol, K.; Ben-Yehuda, S.; King, N.; Losick, R. Genetic dissection of the sporulation protein SpoIIE and its role in asymmetric division in Bacillus subtilis. J. Bacteriol. 2005, 187, 3511–3520. [Google Scholar] [CrossRef] [Green Version]
  62. Perez, A.R.; Abanes-De Mello, A.; Pogliano, K. SpoIIB localizes to active sites of septal biogenesis and spatially regulates septal thinning during engulfment in Bacillus subtilis. J. Bacteriol. 2000, 182, 1096–1108. [Google Scholar] [CrossRef] [Green Version]
  63. Peters, H., 3rd; Haldenwang, W.G. Synthesis and fractionation properties of SpoIIGA, a protein essential for pro-sigma E processing in Bacillus subtilis. J. Bacteriol. 1991, 173, 7821–7827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Gutierrez, J.; Smith, R.; Pogliano, K. SpoIID-mediated peptidoglycan degradation is required throughout engulfment during Bacillus subtilis sporulation. J. Bacteriol. 2010, 192, 3174–3186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Abanes-De Mello, A. The Roles of SpoIID, SpoIIM, and SpoIIP in Septal Thinning and Membrane Migration during Bacillus subtilis Engulfment; University of California: San Diego, CA, USA, 2003. [Google Scholar]
  66. Doan, T.; Morlot, C.; Meisner, J.; Serrano, M.; Henriques, A.O.; Moran, C.P., Jr.; Rudner, D.Z. Novel secretion apparatus maintains spore integrity and developmental gene expression in Bacillus subtilis. PLoS Genet. 2009, 5, e1000566. [Google Scholar] [CrossRef] [Green Version]
  67. McKenney, P.T.; Driks, A.; Eichenberger, P. The Bacillus subtilis endospore: Assembly and functions of the multilayered coat. Nat. Rev. Microbiol. 2013, 11, 33–44. [Google Scholar] [CrossRef] [PubMed]
  68. Debarbouille, M.; Martin-Verstraete, I.; Kunst, F.; Rapoport, G. The Bacillus subtilis sigL gene encodes an equivalent of sigma 54 from gram-negative bacteria. Proc. Natl. Acad. Sci. USA 1991, 88, 9092–9096. [Google Scholar] [CrossRef]
  69. Zhang, X.; Yang, T.-W.; Lin, Q.; Xu, M.-J.; Xia, H.-F.; Xu, Z.-H.; Li, H.-Z.; Rao, Z.-M. Isolation and identification of an acetoin high production bacterium that can reverse transform 2, 3-butanediol to acetoin at the decline phase of fermentation. World J. Microbiol. Biotechnol. 2011, 27, 2785–2790. [Google Scholar] [CrossRef]
  70. Wang, J.; Mei, H.; Zheng, C.; Qian, H.; Cui, C.; Fu, Y.; Su, J.; Liu, Z.; Yu, Z.; He, J. The metabolic regulation of sporulation and parasporal crystal formation in Bacillus thuringiensis revealed by transcriptomics and proteomics. Mol. Cell. Proteom. 2013, 12, 1363–1376. [Google Scholar] [CrossRef] [Green Version]
  71. Ould Ali, N.; Bignon, J.; Rapoport, G.; Debarbouille, M. Regulation of the acetoin catabolic pathway is controlled by sigma L in Bacillus subtilis. J. Bacteriol. 2001, 183, 2497–2504. [Google Scholar] [CrossRef] [Green Version]
  72. Engelberg-Kulka, H.; Amitai, S.; Kolodkin-Gal, I.; Hazan, R. Bacterial programmed cell death and multicellular behavior in bacteria. PLoS Genet. 2006, 2, e135. [Google Scholar] [CrossRef]
  73. González-Pastor, J.E.; Hobbs, E.C.; Losick, R. Cannibalism by sporulating bacteria. Science 2003, 301, 510–513. [Google Scholar] [CrossRef]
  74. Fujita, M.; González-Pastor, J.E.; Losick, R. High-and low-threshold genes in the Spo0A regulon of Bacillus subtilis. J. Bacteriol. 2005, 187, 1357–1368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Hobbs, E.C. Control of Cannibalism in Bacillus subtilis; Harvard University: Cambridge, MA, USA, 2006. [Google Scholar]
Figure 1. Optical density (OD600) and sporulation rate of B. subtilis DB104 during growth in LB.
Figure 1. Optical density (OD600) and sporulation rate of B. subtilis DB104 during growth in LB.
Microorganisms 11 01928 g001
Figure 2. Analysis of the transcriptomic data. (a) Multidimensional scaling plot and (b) Pearson correlation coefficient of all samples. (c) Venn diagram analysis and (d) the numbers of DEGs identified by pairwise times with a |fold change| ≥ 2.
Figure 2. Analysis of the transcriptomic data. (a) Multidimensional scaling plot and (b) Pearson correlation coefficient of all samples. (c) Venn diagram analysis and (d) the numbers of DEGs identified by pairwise times with a |fold change| ≥ 2.
Microorganisms 11 01928 g002
Figure 3. K-mean clustering analysis of differentially expressed genes (DEGs). (a) Heatmap of each cluster over time (columns) for all DEGs. (b) The expression patterns of each cluster.
Figure 3. K-mean clustering analysis of differentially expressed genes (DEGs). (a) Heatmap of each cluster over time (columns) for all DEGs. (b) The expression patterns of each cluster.
Microorganisms 11 01928 g003
Figure 4. Expression patterns of DEGs. Z-score value of (a) basal body, (b) filament, and (c) hook genes of B. subtilis flagella. (d) Z-score value of the skf operon. (e) Z-score value of the sdpABC and sdpRI operons.
Figure 4. Expression patterns of DEGs. Z-score value of (a) basal body, (b) filament, and (c) hook genes of B. subtilis flagella. (d) Z-score value of the skf operon. (e) Z-score value of the sdpABC and sdpRI operons.
Microorganisms 11 01928 g004
Figure 5. Sporulation stage of B. subtilis and expression patterns of genes involved in sporulation. Gene expression is visualized as the Z-score. The red and green colors represent the up-regulated and down-regulated genes, respectively.
Figure 5. Sporulation stage of B. subtilis and expression patterns of genes involved in sporulation. Gene expression is visualized as the Z-score. The red and green colors represent the up-regulated and down-regulated genes, respectively.
Microorganisms 11 01928 g005
Figure 6. Expression patterns of the genes involved in spore coat expression. Gene expression is visualized as the Z-score. The red and blue colors represent the up-regulated and down-regulated gene, respectively. E represents the σE-dependent genes. K represents the σK-dependent genes.
Figure 6. Expression patterns of the genes involved in spore coat expression. Gene expression is visualized as the Z-score. The red and blue colors represent the up-regulated and down-regulated gene, respectively. E represents the σE-dependent genes. K represents the σK-dependent genes.
Microorganisms 11 01928 g006
Figure 7. Real-time quantitative PCR validation of RNA-seq data for most up- and down-regulated DEGs. The right and left y-axis represent the TPM value of RT-qPCR and fold change of RNA-seq, respectively. The fold-change values were generated for RT-qPCR samples by comparing the expression of genes at each timepoint vs. at 8 h. RT-qPCR analysis for RNA expression of target genes was normalized against rpsJ. RT-qPCR data are presented as the mean ± standard deviation (n = 3).
Figure 7. Real-time quantitative PCR validation of RNA-seq data for most up- and down-regulated DEGs. The right and left y-axis represent the TPM value of RT-qPCR and fold change of RNA-seq, respectively. The fold-change values were generated for RT-qPCR samples by comparing the expression of genes at each timepoint vs. at 8 h. RT-qPCR analysis for RNA expression of target genes was normalized against rpsJ. RT-qPCR data are presented as the mean ± standard deviation (n = 3).
Microorganisms 11 01928 g007
Table 1. Transcripts per million (TPM) of the highest differentially expressed genes (DEGs).
Table 1. Transcripts per million (TPM) of the highest differentially expressed genes (DEGs).
GeneDescription8 h10 h12 h15 h18 h24 hTotal
cotXspore coat protein (insoluble fraction)290627221,51675,55818,170115,668
sspEsmall acid-soluble spore protein (gamma-type SASP)14038217,43968,24210,15517,838114,196
yczNputative spore and germination protein59067385168511,54597,628111,972
gerEtranscriptional regulator of late spore coat genes38214912317,07150,09532,767100,587
cotYouter spore coat protein (crust layer, insoluble fraction)87823621,14765,66710,29397,429
cotZspore coat protein (insoluble fraction, crust layer)913536419,37355,345836183,588
sdpCprecursor of killing factor SdpC15,17532,30313,4132336980666670,873
skfAsporulation killing factor A365316,99037,15765312708305470,093
cotVspore coat protein (insoluble fraction)2544435940438,99217,22665,956
cgeBmaturation of the outermost layer of the spore34912278467522,33235,83063,386
cgeAspore outermost layer component319151108329917,99541,38263,254
cotDspore coat protein (inner)112395411,74438,21710,03060,197
yczMputative type I toxin314482240486714249,15757,820
sspBsmall acid-soluble spore protein (beta-type SASP)384808235,5912720428650,766
safAmorphogenetic protein associated with SpoVID6551618,99817,4023633470550,260
Table 2. The top 10 most up- and down-regulated DEGs according to fold change.
Table 2. The top 10 most up- and down-regulated DEGs according to fold change.
GeneDescriptionPairwise TimeFold Changep-ValueAdj. p-Value a
cotDspore coat protein (inner)15 h/12 h308.742.28 × 10−434.45 × 10−40
spoIIIDtranscriptional regulator (stage III sporulation)10 h/8 h257.782.07 × 10−388.08 × 10−35
yqfXconserved protein of unknown function expressed in germinating spores12 h/10 h236.641.16 × 10−251.13 × 10−22
spmHglucose-1-phosphate cytidylyltransferase (sporulation)15 h/12 h223.841.08 × 10−323.84 × 10−30
yhdBconserved hypothetical protein12 h/10 h217.381.10 × 10−183.92 × 10−16
ypzDputative germination protein15 h/12 h210.051.35 × 10−314.41 × 10−29
cotNEinner spore coat protein12 h/10 h203.644.51 × 10−112.75 × 10−9
yurSconserved protein of unknown function15 h/12 h175.042.15 × 10−244.41 × 10−22
cotGspore morphogenetic protein15 h/12 h172.245.10 × 10−451.99 × 10−41
sppOspore protein cse1510 h/8 h166.919.57 × 10−201.97 × 10−17
gsiBgeneral stress protein glucose starvation induced15 h/12 h−16.301.02 × 10−93.60 × 10−8
frlOfructose amino acid-binding lipoprotein12 h/10 h−16.782.48 × 10−101.31 × 10−8
cheYregulator of chemotaxis and motility10 h/8 h−17.472.11 × 10−104.17 × 10−9
gbsBcholine dehydrogenase12 h/10 h−20.412.69 × 10−52.07 × 10−4
frlNfructose-amino acid permease12 h/10 h−20.812.75 × 10−111.76 × 10−9
frlMfructose-amino acid permease12 h/10 h−25.961.06 × 10−129.63 × 10−11
gbsAglycine betaine aldehyde dehydrogenase, NAD+-dependent12 h/10 h−36.863.30 × 10−75.59 × 10−6
gabT4-aminobutyrate aminotransferase10 h/8 h−39.083.16 × 10−183.75 × 10−16
xptxanthine phosphoribosyltransferase10 h/8 h−48.692.65 × 10−183.30 × 10−16
pbuXxanthine permease10 h/8 h−88.521.81 × 10−193.07 × 10−17
a p-value after FDR correction.
Table 3. The top 10 KEGG pathways of up- or down-regulated DEGs.
Table 3. The top 10 KEGG pathways of up- or down-regulated DEGs.
Pathway IDKEGG Pathway10 h/8 h12 h/10 h15 h/12 h18 h/15 h24 h/18 h
Up-regulated
bsu01100Metabolic pathways11112087390
bsu01110Biosynthesis of secondary metabolites495341210
bsu01120Microbial metabolism in diverse environments31362900
bsu00500Starch and sucrose metabolism11151170
bsu01250Biosynthesis of nucleotide sugars810780
bsu01230Biosynthesis of amino acids810780
bsu00541O-Antigen nucleotide sugar biosynthesis0020011
bsu02010ABC transporters0001110
bsu00520Amino sugar and nucleotide sugar metabolism012900
bsu00650Butanoate metabolism810000
Down-regulated
bsu01100Metabolic pathways00736125
bsu01120Microbial metabolism in diverse environments383402310
bsu02040Flagellar assembly3122000
bsu02010ABC transporters2626000
bsu02030Bacterial chemotaxis3111000
bsu01230Biosynthesis of amino acids029000
bsu02020Two-component system280000
bsu00230Purine metabolism1701000
bsu00650Butanoate metabolism76600
bsu00020Citrate cycle (TCA cycle)810000
Table 4. Transcripts per million (TPM) of sigma-factor-encoding genes.
Table 4. Transcripts per million (TPM) of sigma-factor-encoding genes.
GeneDescription8 h10 h12 h15 h18 h24 h
sigARNA polymerase major sigma-43 factor (sigma-A)9848631747757459593
sigBRNA polymerase sigma-37 factor (sigma-B)383618317105120120
sigDRNA polymerase sigma-28 factor (sigma-D)3523373210
sigERNA polymerase sporulation-specific sigma-29 factor (sigma-E)95565275492162
sigFRNA polymerase sporulation-specific sigma factor (sigma-F)2015853169681215741873
sigGRNA polymerase sporulation-specific sigma factor (sigma-G)152107311591444140287
sigIRNA polymerase sigma factor (heat stress responsive)810106417
sigKcRNA polymerase sporulation-specific sigma-K factor precursor (C-terminal fragment)151124267166
sigKnRNA polymerase sporulation-specific sigma-K factor precursor (N-terminal half)93723153887102
sigLRNA polymerase sigma-54 factor (sigma-L)1982381569739124
sigMRNA polymerase ECF (extracytoplasmic function)-type sigma factor (sigma-M)1917178420
sigXRNA polymerase ECF (extracytoplasmic function)-type sigma factor (sigma-X)354222221540
sigWRNA polymerase ECF (extracytoplasmic function)-type sigma factor (sigma-W)130412111100254150437
sigYRNA polymerase ECF (extracytoplasmic function)-type sigma factor (sigma Y)814169818
ylaAconserved protein of unknown function001001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jun, J.-S.; Jeong, H.-E.; Moon, S.-Y.; Shin, S.-H.; Hong, K.-W. Time-Course Transcriptome Analysis of Bacillus subtilis DB104 during Growth. Microorganisms 2023, 11, 1928. https://doi.org/10.3390/microorganisms11081928

AMA Style

Jun J-S, Jeong H-E, Moon S-Y, Shin S-H, Hong K-W. Time-Course Transcriptome Analysis of Bacillus subtilis DB104 during Growth. Microorganisms. 2023; 11(8):1928. https://doi.org/10.3390/microorganisms11081928

Chicago/Turabian Style

Jun, Ji-Su, Hyang-Eun Jeong, Su-Yeong Moon, Se-Hee Shin, and Kwang-Won Hong. 2023. "Time-Course Transcriptome Analysis of Bacillus subtilis DB104 during Growth" Microorganisms 11, no. 8: 1928. https://doi.org/10.3390/microorganisms11081928

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop