Next Article in Journal
Physiological, Genetic, and Fermentative Traits of Oenococcus oeni Isolates from Spontaneous Malolactic Fermentation in Koshu Wine
Previous Article in Journal
Changes in Ruminal Dynamics and Microbial Populations Derived from Supplementation with a Protein Concentrate for Cattle with the Inclusion of Non-Conventional Feeding Sources
Previous Article in Special Issue
Bioprospecting the Endophytic Fungus, Lasiodiplodia theobromae MMPI, for the Integrated Production of Mycoprotein and Exocellular (1→6)-β-Glucan
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transcriptomic Analysis Reveals Opposing Roles of CEL1B in Sophorose- and Lactose-Induced Cellulase Expression in Trichoderma reesei Rut C30

1
School of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
2
Chongqing Key Laboratory of Digitalization in Pharmaceutical Processes, Chongqing University of Science and Technology, Chongqing 401331, China
*
Author to whom correspondence should be addressed.
Fermentation 2025, 11(8), 439; https://doi.org/10.3390/fermentation11080439 (registering DOI)
Submission received: 4 June 2025 / Revised: 26 July 2025 / Accepted: 27 July 2025 / Published: 31 July 2025

Abstract

The β-glucosidase CEL1B has been linked to regulating cellulase expression in Trichoderma reesei, yet its inducer-specific functions and broader regulatory roles remain poorly characterized. In this study, CRISPR-Cas9-mediated gene knockout was applied in the industrial high-producing T. reesei Rut C30 to investigate CEL1B function without the confounding effects of KU70 deletion. Unlike previous studies focused solely on cellulose or lactose induction, transcriptomic analysis of the CEL1B knockout strain revealed its regulatory roles under both lactose- and sophorose-rich conditions, with sophorose representing the most potent natural inducer of cellulase expression. Under lactose induction, CEL1B deletion resulted in a 52.4% increase in cellulase activity (p < 0.05), accompanied by transcriptome-wide upregulation of β-glucosidase genes (CEL3A: 729%, CEL3D: 666.8%, CEL3C: 110.9%), cellulose-sensing receptors (CRT1: 203.0%, CRT2: 105.8%), and key transcription factors (XYR1: 2.7-fold, ACE3: 2.8-fold, VIB1: 2.1-fold). Expression of ER proteostasis genes was significantly upregulated (BIP1: 3.3-fold, HSP70: 6.2-fold), contributing to enhanced enzyme secretion. Conversely, under sophorose induction, CEL1B deletion reduced cellulase activity by 25.7% (p < 0.05), which was associated with transcriptome profiling showing significant downregulation of β-glucosidase CEL3H (66.6%) and cellodextrin transporters (TrireC30_91594: 79.3%, TrireC30_127980: 76.3%), leading to reduced cellobiohydrolase expression (CEL7A: 57.8%, CEL6A: 67.8%). This first transcriptomic characterization of the CEL1B knockout strain reveals its dual opposing roles in modulating cellulase expression in response to lactose versus sophorose, providing new strategies for optimizing inducer-specific enzyme production in T. reesei.

1. Introduction

Amid the depletion of fossil fuels and escalating environmental challenges, the biorefinery industry—which utilizes the abundant lignocellulosic biomass of the Earth to produce renewable energy and bio-based chemicals—has attracted considerable attention. This emerging sector plays a crucial role in promoting global sustainable development and accelerating the transition to low-carbon energy systems [1,2,3]. Lignocellulose, primarily composed of cellulose, hemicellulose, and lignin, is inherently resistant to degradation due to its complex and heterogeneous structure [4,5]. Consequently, efficient cellulases are essential for the effective breakdown of lignocellulosic biomass, but the high cost of these enzymes remains a major bottleneck in industrial bioconversion processes [6]. Trichoderma reesei Rut C30, one of the most widely used industrial cellulase-producing strains, secretes a cellulase system dominated by cellobiohydrolase, endoglucanase, β-glucosidase, and various auxiliary proteins [7,8]. These enzymes act synergistically to convert lignocellulosic materials, such as corn stover, into fermentable glucose [9].
Cellobiohydrolase hydrolyzes cellulose chain ends to release cellobiose; endoglucanase cleaves glycosidic bonds within cellulose’s amorphous regions, thereby reducing its degree of polymerization, and β-glucosidase further converts cellobiose into glucose [9,10,11]. Auxiliary proteins—such as expansins and lytic polysaccharide monooxygenases—enhance enzymatic accessibility by disrupting cellulose crystallinity, loosening fibrillar networks, oxidizing or cleaving cellulose chains to create additional hydrolysis sites, and promoting substrate binding for core enzymes. These mechanisms collectively improve the catalytic efficiency of cellulolytic systems without directly hydrolyzing glycosidic bonds [12,13,14]. However, T. reesei produces these cellulases exclusively under inducible conditions [15,16,17].
Lactose is presently the most cost-effective soluble inducer for cellulase production in submerged liquid fermentation (SLF) [18,19,20]. However, lactose requires hydrolysis and transglycosylation by β-galactosidase or β-glucosidase to generate sophorose, the bioactive compound responsible for cellulase induction [21]. Consequently, lactose is not a direct inducer, which reduces the efficiency of cellulase biosynthesis in SLF. Sophorose (β-1,2-linked disaccharide), identified as the most potent inducer of cellulase synthesis in T. reesei, exhibits an induction capacity over 200-fold higher than lactose. Unfortunately, sophorose’s prohibitive cost precludes its industrial-scale application. Previous studies utilized a biocatalytically prepared glucose–sophorose mixture (MGD) as an inducer for T. reesei Rut C30, achieving cellulase titers of 90.3 FPU/mL [15]. Unlike lactose, direct sophorose application eliminates reliance on β-glucosidase-mediated transglycosylation. Furthermore, β-glucosidase silencing may inhibit sophorose degradation, thereby prolonging cellulase synthesis induction in T. reesei [22].
Previous studies have demonstrated that overexpression of β-glucosidase CEL1B disrupts sugar transport and endoplasmic reticulum homeostasis, thereby inhibiting cellulase synthesis during induction by cellulose or lactose. Conversely, deletion of CEL1B in T. reesei was found to enhance growth rates under lactose induction, suggesting that CEL1B may play a regulatory role in cellulase production [23]. However, its specific function under sophorose induction—the most potent inducer of cellulase synthesis—has not been reported. In our study, knockout of CEL1B resulted in a 25.7% decrease in cellulase activity under glucose–sophorose mixture (MGD) induction, but a 52.4% increase under lactose induction, highlighting its inducer-dependent dual effects on cellulase biosynthesis.
Although previous studies have explored the function of CEL1B in T. reesei KU70 or QM9414 [23,24], several limitations remain unresolved. Notably, functional analyses of CEL1B have been conducted using KU70-deficient strains, despite reports that deletion of KU70 may affect fungal growth [25], metabolism, and gene expression, potentially confounding the interpretation of targeted gene functions. Furthermore, other previous investigations were based on the T. reesei QM9414, which differs significantly from the industrial T. reesei Rut C30 in cellulase production capacity and regulatory background [23]; T. reesei Rut C30 harbors a mutated cre1 gene leading to partial release from carbon catabolite repression and substantially enhanced enzyme secretion [26]. In addition, prior studies have mainly focused on the effects of CEL1B under cellulose or lactose induction [24], while its role under sophorose-rich conditions—the most potent inducer of cellulase production—has not been systematically studied. Moreover, the impact of CEL1B deletion on global gene expression profiles has not yet been explored through transcriptomic analysis, limiting understanding of the underlying regulatory networks. To address these gaps, we employed a CRISPR-Cas9-based strategy to generate a CEL1B gene knockout strain directly in the Rut C30 background without KU70 disruption, and performed comprehensive transcriptomic profiling under different induction conditions. These efforts aim to elucidate the inducer-specific regulatory mechanisms of CEL1B and to provide new insights for optimizing cellulase production in T. reesei.

2. Materials and Methods

2.1. Strains

Trichoderma reesei Rut C30 (NRRL 11460) was obtained from the Agricultural Research Service (ARS) Culture Collection (NRRL; Peoria, IL, USA). The uracil-deficient strain T. reesei Δura5, utilized for CEL1B gene knockout in this study, was derived from T. reesei Rut C30, generated, and maintained in-house [9]. All T. reesei spores were stored in 50% glycerol at −80 °C.

2.2. Construction of CEL1B Gene Knockout T. reesei

The crRNA targeting the CEL1B gene of T. reesei (gene ID: TrireC30_77989) was designed using the CHOPCHOP online tool (http://chopchop.cbu.uib.no/ accessed on 3 June 2025), and the selected target sequence was 5′-CATTAACAAGCACTCCACCG-3′. The sgRNA targeting CEL1B was amplified by PCR using primers CEL1B-F (5′-CATTAACAAGCACTCCACCGGTTTTAGAGCTAGAAATAGC-3′) and CEL1B-R (5′-CACCGAGTCGGTGCTTTTTT-3′) and plasmid pTrgRNA (containing the tracrRNA sequence) as a template. The PCR products were then used as templates for a secondary amplification with primers T7-CEL1B-F (5′-TAATACGACTCACTATAGGGCATTAACAAGCACTCCACCG-3′) and T7-CEL1B-R (5′-CACCGAGTCGGTGCTTTTTT-3′), which included the T7 promoter sequence (5′-TAATACGACTCACTATAGGG-3′) [9]. The resulting DNA was transcribed into RNA using T7 RNA polymerase (Shanghai Biyuntain Biotechnology Co., Ltd., Shanghai, China), purified, and used to knock out CEL1B in T. reesei [27].
Protoplasts were prepared from T. reesei mycelia using the method described by Ogawa et al. [28], with minor modifications. A 500 μL spore suspension of T. reesei Δura5 was added to 50 mL of liquid potato dextrose medium containing 100 μL of uridine. The culture was incubated at 28 °C with agitation at 150 rpm for 12 h to facilitate germination of the spores. Subsequently, mix 3 mg/mL of Lywallzyme (Shanghai Macklin Biochemical Co., Ltd., Shanghai, China) with 2 mg/mL of Yatalase (Baori Doctor Biotechnology Co., Ltd., Beijing, China), and dissolve in 10 mL of OM solution (MgSO4·7H2O 73.9 g/L, Na2HPO4 4.5 g/L, NaH2PO4 8.17 g/L, pH = 5.8), and the germinated spores were further incubated for 12 h to digest cell walls. The resulting protoplasts were collected into 1.5 mL centrifuge tubes. Subsequently, 0.6 μg of the Poura5 expression cassette and 0.6 μg of sgRNA were introduced into the protoplast suspension, followed by incubation on ice for 30 min. After that, 600 μL of 60% (w/v) PEG4000 was added to the mixture, which was then left at room temperature for another 30 min. The transformation mixture was evenly plated onto minimal medium (MM) composed of 4 g/L Na2HPO4, 1.5 g/L KH2PO4, 1 g/L (NH4)2SO4, 0.2 g/L MgSO4·7H2O, 0.02 g/L CaCl2, 1 g/L NaNO3, 5 mg/L FeSO4·7H2O, 1.7 mg/L MnSO4·H2O, 1.4 mg/L ZnSO4·7H2O, 2 mg/L CoCl2, 20 g/L agar, and 10 g/L glucose. Plates were incubated at 28 °C for one week to allow colony formation [9]. Genomic DNA was extracted from transformants, and CEL1B knockout in T. reesei was confirmed by PCR and sequencing, following the method described by Ran et al. [21].

2.3. Cellulase Production

The T. reesei in which the CEL1B gene was successfully disrupted was designated as T. reesei ΔCEL1B. Spores from both the parental strain T. reesei Rut C30 and the ΔCEL1B mutant were cultivated on solid potato dextrose agar (PDA) plates at 28 °C for 7 d to promote sporulation. The harvested spores were then transferred to a seed culture medium containing 4 g/L glucose and 10 g/L corn steep liquor, and incubated at 28 °C with shaking at 150 rpm for 24 h. The resulting mycelial biomass was used to inoculate (8% v/v) a fermentation medium with either 10 g/L MGD or lactose as the sole carbon source.
The fermentation medium additionally included 2.8 g/L (NH4)2SO4, 4 g/L KH2PO4, 0.6 g/L MgSO4·7H2O, 0.8 g/L CaCl2, 10 mg/L FeSO4·7H2O, 1 g/L peptone, 0.3 g/L urea, 3.4 mg/L MnSO4·H2O, 2.8 mg/L ZnSO4·7H2O, 4 mg/L CoCl2, and 0.1 M Na2HPO4–citrate buffer (pH 5.0). Cultures were maintained at 28 °C under agitation (150 rpm), and samples were collected at specified time points. The supernatant and pellet were separated by centrifugation and subsequently used for cellulase activity measurement and biomass analysis, respectively [24].
The glucose–sophorose mixture (MGD) was synthesized via β-glucosidase-catalyzed transglycosylation. β-Glucosidase was added at 30 CBU per gram of glucose to a substrate containing 600 g/L glucose, and the reaction was conducted at 65 °C (pH 4.8) for 72 h. The mixture was heated to 100 °C for 5 min to inactivate β-glucosidase, yielding MGD. The final composition comprised 410.20 g/L glucose, 60.56 g/L laminaribiose, 9.34 g/L cellobiose, and 13.66 g/L sophorose [15].

2.4. RNA Isolation and High-Throughput RNA-Seq

Total RNA was extracted from T. reesei using a Plant Total RNA Purification Kit (Sangon Biotech, Shanghai, China), with three biological replicates prepared for RNA sequencing. RNA quantification and integrity analysis were performed using an Agilent 2100 Bioanalyzer (Agilent Technologies Co., Ltd., Beijing, China). The Agilent RNA 6000 Nano Kit (Agilent Technologies (Beijing, China) Co., Ltd.) was employed for quality control (QC), assessing RNA concentration, 28S/18S rRNA ratio, RNA Integrity Number (RIN), and fragment length distribution. RNA purity was evaluated using a NanoDropTM spectrophotometer (Thermo Fisher Scientific Co., Ltd., Shanghai, China).
RNA sequencing was carried out using the Illumina HiSeq 4000 platform (Shanghai Qingke Technology Co., Ltd., Shanghai, China). Following quality control, the high-quality reads were mapped to the T. reesei RUT C30 genome (https://mycocosm.jgi.doe.gov/cgi-bin/dispTranscript?db=TrireRUTC30_1&id=127115&useCoords=1 accessed on 3 June 2025) using the HISAT2 aligner (https://daehwankimlab.github.io/hisat2/ accessed on 3 June 2025). Subsequent alignment of clean reads to the reference genome was performed with Bowtie2 (http://ccb.jhu.edu/software/stringtie/ accessed on 3 June 2025), and gene expression levels were quantified using RSEM (http://ccb.jhu.edu/software/stringtie/gffcompare.shtml accessed on 3 June 2025). Differential gene expression analysis was conducted with DESeq2 (https://bio.tools/deseq accessed on 3 June 2025), applying a significance threshold of an absolute log2 fold change ≥1 and an adjusted p-value ≤ 0.05. Within DESeq2, the mean normalized gene count served as the filtering criterion, while statistical significance was assessed via the Wald test. Functional annotation and classification of differentially expressed genes were performed based on Gene Ontology (GO) and the Eukaryotic Orthologous Groups (KOG) system, using data from the T. reesei RUT C30 genome version 1.0.
All RNA-seq data from this study have been submitted to the National Center for Biotechnology Information (NCBI) under BioProject accession number PRJNA1260437.

2.5. Determination of Copy Numbers by qPCR Analysis

Genomic DNA was extracted from T. reesei transformants using a commercial fungal DNA isolation kit (Sangon Biotech, Shanghai, China) and used as the template for quantitative PCR (qPCR). The copy number of the Poura5 expression cassette was determined following the procedure outlined by Ran et al. [21], using the single-copy Poura5 gene as an internal reference.
To assess gene expression in T. reesei Rut C30 and ΔCEL1B, qPCR was performed using the protocol previously described. Primer sequences are provided in Table S1, and the sar1 gene was used as the housekeeping reference for normalization.

2.6. Analytical Methods

The concentration of reducing sugars was measured using the 3,5-dinitrosalicylic acid (DNS) method. In brief, 1.5 mL of the appropriately diluted sample was combined with 2 mL of DNS reagent and heated in boiling water for 5 min. After cooling to room temperature, absorbance was recorded at 540 nm, and sugar concentrations were determined using a glucose standard curve as reference [29].
To evaluate cellulase activity, Whatman No. 1 filter paper was used as the substrate. A 500 μL portion of the enzyme-containing supernatant was mixed with 1 mL of 0.2 mol/L sodium acetate–acetic acid buffer (pH 4.8) and a 1 × 6 cm strip of filter paper. The mixture was incubated at 50 °C for 1 h. The amount of reducing sugars released was quantified using the DNS method. One unit (U) of cellulase activity was defined as the amount of enzyme that releases 1 μmol of reducing sugar per minute per milliliter under the assay conditions [29].
Cellobiohydrolase activity was assessed using p-nitrophenyl-β-D-cellobioside (pNPC) as the substrate, following a previously reported protocol [30]. The assay mixture included 100 μL of diluted enzyme solution and 50 μL of 1 g/L pNPC, incubated at 50 °C for 30 min. The reaction was terminated by adding 150 μL of 10% (w/v) sodium carbonate. The release of p-nitrophenol (pNP) was quantified by measuring absorbance at 415 nm. One unit (U) of cellobiohydrolase activity was defined as the amount of enzyme required to produce 1 μg of pNP per minute under assay conditions.
Endoglucanase activity was determined using carboxymethyl cellulose (CMC) as the substrate. The reaction consisted of 500 μL of appropriately diluted enzyme and 1 mL of 2% (w/v) CMC solution, incubated at 50 °C for 30 min. The amount of reducing sugars released was measured using the DNS method. One unit (U) of endoglucanase activity was defined as the amount of enzyme required to release 1 μmol of reducing sugar per minute under assay conditions [29].
β-Glucosidase activity was measured using cellobiose as the substrate. A total of 200 μL of diluted enzyme was mixed with 200 μL of 15 mmol/L cellobiose solution and incubated at 50 °C for 30 min. The reaction was terminated by boiling, and glucose production was quantified using a commercial glucose biosensor (Sieman Technology Co., Ltd., Shenzhen, China). One unit (U) of β-glucosidase activity was defined as the amount of enzyme that hydrolyzes cellobiose to generate 1 μmol of glucose per minute per milliliter under the assay conditions [29].
Protein concentrations were determined using a bicinchoninic acid (BCA) assay kit (Cat. No.: BL521A, BioSharp, Beijing, China). In a 96-well microplate, 200 μL of working solution (prepared by mixing Reagents A and B in a 50:1 volume ratio) and 20 μL of the diluted enzyme solution were combined. The mixtures were incubated at 60 °C for 30 min, and absorbance was measured at 595 nm using a microplate reader (Beijing Kaiao Technology Co., Ltd., Beijing, China). Bovine serum albumin (BSA) was used to generate the standard calibration curve.
SDS-PAGE was conducted using 10% polyacrylamide gels and culture supernatants collected after 60 h of fermentation, which corresponded to the peak of cellulase production. Samples were prepared by mixing with 5× loading buffer at a 4:1 (v/v) ratio and boiling for 10 min. Electrophoresis was performed at a constant voltage until the tracking dye reached the gel bottom. Gels were stained with Coomassie Brilliant Blue R-250 and destained until protein bands were clearly visible.
All results are presented as means ± standard deviation (SD) from three independent biological replicates. Statistical analysis between groups was conducted using Student’s t-test, with significance considered at p < 0.05 (indicated by *), unless stated otherwise.

3. Results

3.1. Gene Knockout and Phenotypic Analysis of CEL1B in Trichoderma reesei RUT C30

Trichoderma reesei RUT C30 is widely used for industrial cellulase production due to its exceptional cellulase-producing capacity. The T. reesei genome contains 11 β-glucosidase genes, whose transglycosylation activity is crucial for cellulase synthesis induced by lactose or cellulose. Additionally, these genes regulate gene expression in a non-enzymatic manner. Among them, CEL1B encodes a key intracellular β-glucosidase in T. reesei [31]. Overexpression of the CEL1B gene under lactose induction reduces sugar transport capacity, triggers endoplasmic reticulum stress, and decreases cellulase activity by over 97.0% [24,31]. Consequently, this study aimed to knock out the CEL1B gene in RUT C30 to improve cellulase production. Meanwhile, a cost-effective glucose–sophorose soluble inducer was previously developed that efficiently induces cellulase synthesis in T. reesei, yet the role of CEL1B under sophorose induction remains unexplored. In previous study, we established a CRISPR-Cas9 genome editing system in T. reesei RUT C30 and derived an uracil auxotrophic strain, T. reesei ΔTrura5. Using the Poura5 expression cassette from Penicillium oxysporum as a selection marker [32], the cassette was integrated into the TrCEL1B gene, thereby inactivating the target gene while restoring the ura5 gene.
The knockout process of the CEL1B gene in T. reesei RUT C30 is illustrated (Figure 1). The gRNA was strategically positioned 656 bp downstream of the gene’s start codon, guiding the insertion of the Poura5 expression cassette at the target site for gene disruption as shown (Figure 1A). Six potential positive transformants were obtained and designated as T. reesei ΔCEL1B1, ΔCEL1B2, ΔCEL1B3, ΔCEL1B4, ΔCEL1B5, and ΔCEL1B6. PCR validation of these transformants showed that, except for ΔCEL1B3, T. reesei ΔCEL1B1, ΔCEL1B2, ΔCEL1B4, ΔCEL1B5, and ΔCEL1B6 all exhibited successful insertion of Poura5 into the CEL1B gene (Figure 1B). Two well-growing transformants, ΔCEL1B1 and ΔCEL1B4, were selected for further study. Figure 1C show the DNA sequencing results of T. reesei ΔCEL1B1 and ΔCEL1B4, indicating that the Poura5 gene expression cassette was inserted 822 bp downstream of the start codon in ΔCEL1B1, while in ΔCEL1B4, the exogenous DNA was inserted 320 bp downstream of the start codon. As shown in Figure 1D, the growth of T. reesei RUT C30, ΔURA5, ΔCEL1B1, and ΔCEL1B4 was examined on MM medium plates without uridine and PDA medium plates containing 5-fluoroorotic acid (5-FOA) and uridine. The results indicated that T. reesei RUT C30, ΔTrCEL1B1, and ΔTrCEL1B4 exhibited identical phenotypes, growing normally on MM medium without uridine but failing to grow on PDA medium containing 5-FOA. In contrast, T. reesei ΔURA5 was able to grow on PDA plates supplemented with 5-FOA and uridine, confirming the successful restoration of the ura5 gene in ΔCEL1B1 and ΔCEL1B4. Further analysis of the URA5 expression cassette copy number (Figure 1E) revealed that T. reesei RUT C30 had a copy number of 0, whereas both ΔTrCEL1B1 and ΔTrCEL1B4 had a copy number of 1, indicating the absence of off-target effects during the gene knockout process.

3.2. Impact of CEL1B Gene Knockout on Cellulase Production

The synthesis of cellulase in T. reesei requires induction, with lactose being the most commonly used inducer [33]. Studies have shown that lactose must be hydrolyzed and transglycosylated by β-galactosidase or β-glucosidase to generate sophorose, which subsequently induces cellulase production [34]. Sophorose has been identified as the most efficient inducer of cellulase synthesis in T. reesei, exhibiting an induction capacity over 200 times higher than that of lactose. Previous studies demonstrated that a cost-effective glucose–sophorose mixture (MGD), prepared via β-glucosidase-catalyzed conversion of high-concentration glucose, can efficiently induce cellulase synthesis without relying on the transglycosylation activity of β-glucosidase [15]. However, the gene function of β-glucosidase CEL1B under lactose induction, particularly with MGD, remains uncharacterized.
T. reesei RUT C30, ΔTrCEL1B1, and ΔTrCEL1B4 were cultured with 10 g/L MGD or lactose as inducers for cellulase synthesis (Figure 2). Under MGD or lactose induction, cellulase production in all strains peaked at 60 h, with T. reesei ΔTrCEL1B1 and ΔTrCEL1B4 exhibiting identical production trends. When induced by MGD, cellulase and cellobiohydrolase activities in T. reesei ΔCEL1B were reduced by 25.7% and 22.7%, respectively, compared to T. reesei RUT C30 (p < 0.05), while β-glucosidase and endoglucanase activities showed no significant differences. This suggests that CEL1B gene knockout increased cellobiohydrolase production, thereby promoting cellulase synthesis. In contrast, under lactose induction, T. reesei ΔCEL1B exhibited increased activities of cellulase (52.4%), cellobiohydrolase (20.1%), β-glucosidase (34.1%), and endoglucanase (10.0%) compared to T. reesei RUT C30 (p < 0.05), indicating that the conversion of lactose to sophorose does not rely on the transglycosylation activity of CEL1B. In summary, sophorose and lactose exhibit distinct molecular mechanisms in inducing cellulase synthesis in T. reesei, and CEL1B negatively regulates cellulase synthesis in an enzyme activity-independent manner.
To further analyze extracellular protein concentrations, the results are shown in Figure 3. Under MGD induction, there was no significant difference in protein secretion between T. reesei RUT C30 and ΔTrCEL1B (p > 0.05). The extracellular protein concentration of T. reesei ΔTrCEL1B increased by 31.0% compared to T. reesei RUT C30 using lactose as inducer (p < 0.05), which was consistent with the cellulase production trend. Moreover, the results of sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) were in agreement with the extracellular protein concentration measurements.
Although a complemented strain was not constructed, two independent CEL1B knockout mutants were generated and analyzed. Both strains exhibited consistent changes in cellulase activity and gene expression, supporting that the observed effects are attributable to the loss of CEL1B.

3.3. Transcriptome Analysis

Given that T. reesei RUT C30 and ΔCEL1B exhibited opposite cellulase production patterns under lactose and MGD induction, respectively, transcriptomic analysis was performed to investigate the underlying molecular mechanisms. Sequencing was conducted on 12 samples, generating approximately 91.48 Gb of data per sample using the Illumina HiSeq platform. The raw sequencing reads contained low-quality bases, adapter contamination, and a high proportion of unknown bases (N), which were removed before downstream analysis. The quality metrics of the clean reads are presented in Table S2. Subsequently, clean reads were aligned to the reference genome using HISAT. The mapping results were consistent across samples (Table S3), confirming their comparability. Based on gene expression data, box plots (Figure S1A) and Principal Component Analysis (PCA) (Figure S1B) indicated that the normalized expression profiles of the samples were similar. All samples used for transcriptomic analysis were highly correlated (Figure S1C).
Based on gene expression analysis, differentially expressed genes (DEGs) were identified among samples or groups. Under MGD induction, 256 DEGs were identified between T. reesei RUT C30 and ΔCEL1B using DESeq2, with 57 genes upregulated and 199 genes downregulated. Similarly, under lactose induction, 949 DEGs were identified, of which 561 and 388 genes were upregulated and downregulated in RUT C30/ΔCEL1B (Figure 4A). The number of DEGs under lactose induction was 4.5 times higher than that under MGD induction, indicating that CEL1B gene knockout had a greater impact when lactose was used as an inducer. Gene Ontology (GO) enrichment analysis of the 199 and 899 CEL1B-specific DEGs revealed that, regardless of induction by MGD or lactose, the functions of DEGs were primarily related to lignocellulose metabolism (including cellulose, hemicellulose, and lignin) and cellulase activity (Figure 4B) and (Figure S2). However, under MGD induction, all lignocellulose metabolism-related genes were downregulated, explaining the decrease in cellulase production following CEL1B gene knockout. In contrast, under lactose induction, these genes were all upregulated, and CEL1B was also found to influence sugar transport processes.

3.4. GH Family

To comprehensively analyze the transcriptional differences in lignocellulose-degrading enzyme genes of T. reesei induced by MGD or lactose after knockout of the CEL1B gene, we examined the differential expression of all annotated glycoside hydrolase (GH) genes [35]. The average fragments per kilobase of transcript per million mapped reads (FPKM) were calculated for each GH family, and the sum of all FPKM averages within a GH family reflected the overall transcription level (Figure 5, Table S4). When MGD was used as the inducer, the total FPKM mean of all GH families in T. reesei ΔCEL1B decreased by 43.1% compared to T. reesei RUT C30. In contrast, lactose induction resulted in a 204% increase, consistent with cellulase production (Figure 2). Among all GH families, GH1 [36,37], GH5 [37,38,39], GH6 [40] and GH7 [41] accounted for the highest proportion, primarily encoding β-glucosidase, endoglucanase and cellobiohydrolase. Under MGD induction, the transcription levels of these four GH families in T. reesei ΔCEL1B constituted 58.0% of all GH genes, showing a 53.1% decrease compared to T. reesei RUT C30, with GH6 exhibiting the most significant reduction (65.4%), while GH1 decreased by 1.6%, GH5 by 60.9%, and GH7 by 55.9%. Conversely, under lactose induction, the transcription levels of GH1, GH5, GH6, and GH7 in T. reesei ΔCEL1B increased by 275.2% compared to T. reesei RUT C30, with GH1, GH5, GH6, and GH7 rising by 37.1%, 275.7%, 406.2%, and 366.6% respectively. These genes account for 50.0% of all GH genes.
Table 1 details the major lignocellulose-degrading genes differentially expressed in T. reesei Rut C30 and ΔCEL1B under different inducers, showing expression patterns consistent with cellulase fermentation results. When MGD was used as the inducer, the expression levels of the two cellobiohydrolases (CEL7A [33,42,43] and CEL6A [44,45,46]) in T. reesei ΔCEL1B decreased by 57.8% and 67.8%, respectively. Since cellobiohydrolases play a critical role in cellulose degradation, this may be the primary reason for the reduced cellulase production in T. reesei ΔCEL1B [47,48]. Among endoglucanases, CEL5A [49] and CEL12A [50,51] were downregulated by 69.8% and 61.2%, respectively, while CEL61A [52] and CEL45A [51] showed no significant difference. Similarly, the expression of the major extracellular β-glucosidase CEL3A [9,21,31,48] was not significantly altered, consistent with the β-glucosidase fermentation results. However, CEL3H expression decreased by 66.6%; previous studies reported that deletion of CEL3H impairs sophorose transport, which may explain the reduced cellulase production upon sophorose induction following CEL1B deletion [53]. In addition, auxiliary activity proteins (AA), including AA9 (lytic polysaccharide monooxygenase), swollenin, Cip1 [33,54], and Cip2 [55], were significantly downregulated. AA9 oxidatively disrupts the crystalline structure of cellulose, creating cleavage sites [56]; swollenin physically loosens the hydrogen-bond network of cellulose, increasing substrate accessibility [53]; Cip1 and Cip2, as non-catalytic auxiliaryproteins, enhance enzyme adsorption stability and promote inter-enzyme synergy, respectively, thereby optimizing enzyme–substrate interactions [33,54,55]. Together, these four components form a cooperative “pre-treatment–oxidation–synergism” system that significantly reduces the recalcitrance of cellulose and supports efficient lignocellulose bioconversion. Therefore, when MGD is used as the inducer, the cellulase cocktail produced by T. reesei ΔCEL1B is less effective in hydrolyzing lignocellulosic biomass such as corn stover. In contrast, under lactose induction, most lignocellulose-degrading genes were significantly upregulated in T. reesei ΔCEL1B, consistent with the fermentation results of filter paper activity, pNPCase, CMCase, and β-glucosidase. Notably, CEL3H expression was not significantly affected, suggesting that CEL1B does not directly regulate CEL3H expression and that its regulation is related to the type of inducer used. Whereas the expression of β-glucosidase CEL3A was significantly upregulated by 729.0% when lactose was used as an inducer, our previous study showed that deletion of CEL3A reduced cellulase production by 19.2%, suggesting that the transglycosylation activity of CEL3A is important for lactose induction. This is also an important reason for the increased cellulase production by T. reesei ΔCEL1B during lactose induction.

3.5. Transporters

It is predicted that the T. reesei genome contains 113 sugar transporter proteins, compared to only 43 in Saccharomyces cerevisiae. This is expected, as T. reesei has a much broader range of sugar substrates than S. cerevisiae, which can grow only on a limited number of monosaccharides and disaccharides. The lifestyle of T. reesei requires the utilization of various pentoses, hexoses, and oligosaccharides, primarily derived from lignocellulose degradation [19,57]. Inducers of cellulase production in T. reesei include cellobiose, lactose, sorbose, and sophorose, while glucose inhibits this induction [9,15,33,58]. The mechanism by which T. reesei senses the presence of inducing sugars remains unclear; however, studies suggest that membrane proteins are involved in this process. As shown in Table S5 and Figure 6, 28 DEGs related to transporter proteins were identified under MGD induction, whereas 98 DEGs were identified under lactose induction.
Table 2 summarizes the expression profiles of transporter-encoding genes in T. reesei, including identified sugar transporters and cellulose sensors. Under MGD induction, four sugar transporter genes—TrireC30_91594, TrireC30_127980, TrireC30_33630 (Xlt1) [59], and TrireC30_138519 (Str1) [60,61]—were downregulated by 79.3%, 76.3%, 65.0%, and 52.0%, respectively, among 28 differentially expressed transporters. TrireC30_91594 and TrireC30_127980, predicted to encode cellodextrin transporters, exhibited reduced expression, potentially impairing sophorose transport under MGD and thereby attenuating cellulase gene induction. TrireC30_33630 (encoding the monosaccharide transporter Xlt1) and TrireC30_138519 (Str1) are established monosaccharide transporters. In T. reesei, monosaccharide transport (e.g., glucose and xylose) involves multiple genes, including the high-affinity transporter Hxt1 [62] and low-affinity transporter Stp1 [63]. Consequently, MGD induction minimally impacted monosaccharide transport in the T. reesei ΔCEL1B.
When lactose served as the inducer, 98 differentially expressed genes (DEGs) were linked to transporter-encoding genes, comprising 73 up- and 25 downregulated genes. Among these, 17 were sugar transporter genes (14 upregulated, 3 downregulated), representing 17.3% of transporter-related DEGs. The cellulose sensor Crt1 (TrireC30_109243), a predicted β-disaccharide transporter critical for cellulase induction, exhibited a 203% upregulation in T. reesei ΔCEL1B [64,65,66,67]. Additionally, Crt2 (TrireC30_137001) expression increased by 105.8% [64]. Although Crt2 is not directly involved in β-disaccharide transport, it regulates cellulase gene expression by activating the transcription factor Xyr1 and subsequently Crt1. These elevated expression levels of Crt1 and Crt2 likely contribute significantly to enhanced cellulase production. With the exception of Hxt1 [62], all other mono- and disaccharide transporters were significantly upregulated, indicating that CEL1B deletion enhances cellulase biosynthesis under lactose induction. The divergent expression profiles of sugar transporters between MGD and lactose conditions suggest inducer-specific regulation. This supports a model where CEL1B deletion promotes lactose-to-sophorose conversion, amplifying cellulase induction.

3.6. Transcription Factors

The T. reesei genome contains 758 predicted transcription factor (TF)-encoding genes. Under MGD induction, six TFs exhibited differential expression (three upregulated, three downregulated), while lactose induction altered 46 TFs (26 upregulated, 20 downregulated) (Table S6). The expression profiles of TF-encoding genes regulating lignocellulase expression are summarized in Table 3. Under MGD induction, no significant changes (p > 0.05) occurred in known cellulase gene regulators. In contrast, lactose induction in T. reesei ΔCEL1B upregulated cellulase-activating TFs Xyr1 (2.7-fold), Ace3 (2.8-fold), and Vib1 (2.1-fold) compared to T. reesei Rut C30, consistent with cellulase production trends. Xyr1 and Ace3 are indispensable for cellulase gene expression, as their deletion severely impairs production and transcription [55,68,69,70,71,72,73], Ace3 [74,75,76,77]. Vib1 further enhances cellulase expression; its overexpression in T. reesei Rut C30 increased cellulase yields and secretion by 200.0% and 219.0%, respectively [76,78,79]. The elevated cellulase production in T. reesei ΔCEL1B under lactose likely stems from TF upregulation. Since lactose requires β-glucosidase-mediated transglycosylation to generate sophorose—the true cellulase inducer—CEL1B is not central to this conversion. Direct use of sophorose-containing MGD reduced only cellobiohydrolase in T. reesei ΔCEL1B, minimally affecting overall cellulase production. Conversely, lactose induction significantly boosted cellulase yields in T. reesei ΔCEL1B, likely due to enhanced sophorose formation and induction efficiency.
As shown in Figure 7, the two most differentially expressed transcription factors (TFs) in T. reesei ΔCEL1B—TrireC30_106452 and TrireC30_133861—showed divergent expression patterns depending on the inducer. Under MGD induction, their mRNA levels were suppressed by 70.7% and 52.0%, respectively, whereas lactose induction markedly upregulated them by 185.6% and 257.1%. Both TFs belong to the Gcn5-related N-acetyltransferase (GNAT) family [80,81], which regulates cellulase gene transcription via histone acetylation. GNAT acetyltransferases, such as Gcn5 [82,83], catalyze lysine acetylation on histone tails (e.g., H3K9/K14) at promoter regions, loosening chromatin structure to activate transcription. The pronounced upregulation of these GNAT members under lactose correlates with enhanced cellulase production, while their repression under MGD aligns with reduced enzyme yields, suggesting GNAT-mediated chromatin remodeling directly modulates cellulase expression. Notably, no other differentially expressed TFs exhibited this regulatory association, highlighting the specificity of GNAT-driven epigenetic control. These findings establish GNAT acetyltransferases as critical regulators of cellulase biosynthesis, with their inducer-dependent expression shifts reflecting metabolic adaptations caused by CEL1B gene deletion, favoring lactose-to-sophorose conversion over direct transcriptional regulation.

3.7. Endoplasmic Reticulum (ER) Protein Processing Pathway

In filamentous fungi, nascent polypeptides translocated into the endoplasmic reticulum undergo coordinated processing events, including signal peptide cleavage, glycosylation, and chaperone-mediated structural maturation [84]. Notably, a comparative analysis demonstrated that T. reesei ΔCEL1B exhibited significantly enhanced secretory capacity under lactose induction compared to the parental T. reesei Rut C30 strain (p < 0.05), whereas both strains showed comparable extracellular protein yields when cultured with MGD as the inducer. These findings underscore the critical challenge of maintaining the fidelity of the secretory pathway, wherein aberrant polypeptides are systematically recognized and targeted for cytoplasmic degradation via the ubiquitin–proteasome system (UPS) [53].
As illustrated in Table 4, transcriptional profiling revealed differential expression patterns of genes involved in protein folding and post-translational processing. Under lactose induction, eight DEGs were identified, all of which were highly expressed. The 6.2-fold upregulation of the Hsp70 nucleotide exchange factor (TrireC30_124246) suggests an enhanced protein quality control capacity, augmenting Hsp70 chaperone activity to manage cellular stress and protein misfolding [85,86]. The observed upregulation of molecular chaperones—including Hsp90 (TrireC30_102206, 2.0-fold) [86], the DnaJ-domain co-chaperone (TrireC30_122594, 1.1-fold), and BiP1 (TrireC30_25648, 3.3-fold) [84,85,87]—suggests a coordinated enhancement of protein quality control mechanisms, likely improving folding efficiency (via Hsp70–Hsp40 collaboration), stabilizing stress-responsive clients (through Hsp90 activity), and reinforcing endoplasmic reticulum homeostasis (via the BiP1-driven unfolded protein response).
The marked upregulation of ERManI (TrireC30_83445: 2.8-fold; TrireC30_101105: 2.1-fold) [87], which encodes 1,2-α-mannosidase critical for N-glycan trimming and glycoprotein quality control in the ER, suggests enhanced processing of misfolded glycoproteins, potentially accelerating ER-associated degradation (ERAD) or maturation pathways. Concurrently, the slight increase in NEF (TrireC30_137683: 1.0-fold), an Armadillo/β-catenin-like repeat-containing protein, may synergize with molecular chaperones to regulate protein folding or substrate release, collectively fine-tuning ER proteostasis under stress conditions [24,33]. The significant downregulation of BiP1 (53.8% reduction) and SPCS1 (TrireC30_116265, 54.7% reduction) [24] in T. reesei ΔCEL1B under MGD induction indicates disruptions in endoplasmic reticulum (ER) proteostasis and secretory pathway efficiency. In summary, the use of lactose as an inducer avoids excessive ER stress, which is advantageous for both T. reesei survival and cellulase production. Notably, the contrasting responses of the ER protein processing pathway under lactose versus MGD induction in the T. reesei ΔCEL1B strain suggest that CEL1B deletion primarily facilitates lactose-to-sophorose conversion rather than directly modulating ER proteostasis.

3.8. RT-qPCR

The transcriptomic data were validated by RT-qPCR analysis of mRNA expression levels for 22 genes in T. reesei Rut C30 and ΔCEL1B (Figure 8). The Log2 fold change (Log2FC) values obtained from both transcriptomic analysis and RT-qPCR (averaged from three replicates) showed a strong Pearson correlation (R2 = 0.9101), indicating the reliability of the transcriptome data.

4. Conclusions

The knockout of CEL1B in Trichoderma reesei exerts inducer-specific regulatory effects on cellulase biosynthesis. Under lactose induction, CEL1B gene deletion elevated cellulase production by 52.4% (p < 0.05), driven by the upregulation of β-glucosidase genes (Cel3A: 729.0%, Cel3D, and Cel3C), which enhanced lactose-to-sophorose conversion. This activated cellulose sensors Crt1 (203% upregulation) and Crt2 (105.8% upregulation), stimulating transcription factors Xyr1 (2.7-fold), Ace3 (2.8-fold), and Vib1 (2.1-fold) to amplify cellulase gene expression. Concurrently, the upregulation of ER proteostasis genes (BiP1: 3.3-fold, Hsp70: 6.2-fold, Hsp90: 2.0-fold) improved enzyme folding and secretion efficiency. In contrast, under MGD induction, CEL1B knockout reduced cellulase activity by 25.7% (p < 0.05) through the downregulation of β-glucosidase Cel3H (66.6%) and cellodextrin transporters (TrireC30_91594: 79.3%, TrireC30_127980: 76.3%), which impaired sophorose transport and suppressed cellobiohydrolases (CEL7A: 57.8%, CEL6A: 67.8%). These results demonstrate that CEL1B acts as a metabolic switch, repressing cellulase synthesis under sophorose-rich conditions while enhancing lactose-driven induction via transcriptional and secretory pathway optimization. This dual regulatory mechanism provides a foundation for engineering T. reesei with tailored inducer responsiveness for industrial lignocellulose bioconversion.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/fermentation11080439/s1, Figure S1: Correlation between samples and gene expression distribution of RNA-seq data. (A) Gene expression box plot; (B) Principal Component Analysis (PCA) of samples; (C) Heat map of Pearson correlation between samples; Figure S2: Hierarchical analysis diagram of GO enrichment; Table S1: PCR primers mainly used for the knockout of CEL1B and primers for qPCR analysis; Table S2: Clean reads quality metrics; Table S3: Summary of Genome Mapping; Table S4: All carbohydrate-active enzyme (CAZy) genes when ΔCEL1B is cultured in MGD or lactose; Table S5: Upregulated downregulated transporter genes in the presence of MGD and lactose after the knockout of CEL1B; Table S6: Upregulated downregulated transcription factor genes in the presence of MGD and lactose after the knockout of CEL1B.

Author Contributions

Conceptualization, Y.L.; methodology, Y.L.; software, J.C.; visualization, J.C., L.W. and X.H.; formal analysis, J.C.; investigation, J.F.; data curation, L.W.; writing—original draft, Y.L. and L.W.; writing—review and editing, X.Z. and T.T.; supervision, X.Z.; funding acquisition, Y.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financially funded by the National Natural Science Foundation of China (Grant numbers 22378033), the Natural Science Foundation Project of Chongqing, the Chongqing Science and Technology Commission (CN) (Grant No. CSTB2022NSCQ-MSX0544), the Major Project of Science and Technological Research of Chongqing Municipal Education Commission (Grant No. KJZD-M202401502), the Youth Project of Science and Technology Research Program of Chongqing Municipal Education Commission (Grant No. KJQN202301546), and also supported by the National Undergraduate Training Programs for Innovation and Entrepreneurship of China (No. 202511551014).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data generated in this study has been deposited in the National Center for Biotechnology Information (NCBI). It can be accessed via the following link: https://www.ncbi.nlm.nih.gov/bioproject/PRJNA1260437 (accessed on 3 June 2025).

Acknowledgments

We thank all of the members of the laboratory for their support and constructive comments, and all authors included in this section have consented to the acknowledgement.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
MGDGlucose–sophorose mixture
GH familiesGlycoside Hydrolase families
T. reeseiTrichoderma reesei
SLFSubmerged liquid fermentation
Sophoroseβ-1,2-linked disaccharide
CRISPR-Cas9Clustered Regularly Interspaced Short Palindromic Repeats associated with Cas9
sgRNAsingle guide RNA
DEGsDifferentially expressed genes
GOGene Ontology
KOGEukaryotic Orthologous Groups
SRASequence Read Archive
qPCRQuantitative Polymerase Chain Reaction
DNS3,5-dinitrosalicylic acid
pNPCp-nitrophenyl-β-D-cellobioside
pNPp-nitrophenol
CMCCarboxymethyl cellulose
BCABicinchoninic acid
SDS-PAGESulfate polyacrylamide gel electrophoresis
5-FOA5-fluoroorotic acid
PCAPrincipal Component Analysis
FPKMFragments per kilobase of transcript per million mapped reads
TFsTranscription factors
GNATGcn5-related N-acetyltransferase
EREndoplasmic reticulum
ARSAgricultural Research Service

References

  1. Li, G.; Wang, R.; Pang, J.; Wang, A.; Li, N.; Zhang, T. Production of renewable hydrocarbon biofuels with lignocellulose and its derivatives over heterogeneous catalysts. Chem. Rev. 2024, 124, 2889–2954. [Google Scholar] [CrossRef] [PubMed]
  2. Machineni, L.; Deepanraj, B.; Chew, K.W.; Rao, A.G. Biohydrogen production from lignocellulosic feedstock: Abiotic and biotic methods. Renew. Sustain. Energy Rev. 2023, 182, 113344. [Google Scholar] [CrossRef]
  3. Shivhare, A.; Kumar, A.; Srivastava, R. Metal phosphate catalysts to upgrade lignocellulose biomass into value-added chemicals and biofuels. Green Chem. 2021, 23, 3818–3841. [Google Scholar] [CrossRef]
  4. Woiciechowski, A.L.; Neto, C.J.D.; de Souza Vandenberghe, L.P.; de Carvalho Neto, D.P.; Sydney, A.C.N.; Letti, L.A.J.; Karp, S.G.; Torres, L.A.Z.; Soccol, C.R. Lignocellulosic biomass: Acid and alkaline pretreatments and their effects on biomass recalcitrance—Conventional processing and recent advances. Bioresour. Technol. 2020, 304, 122848. [Google Scholar] [CrossRef]
  5. Das, N.; Jena, P.K.; Padhi, D.; Mohanty, M.K.; Sahoo, G. A comprehensive review of characterization, pretreatment and its applications on different lignocellulosic biomass for bioethanol production. Biomass Convers. Biorefinery 2021, 13, 1503–1527. [Google Scholar] [CrossRef]
  6. Zhang, Z.; Zhang, J.; Li, H.; Zhao, Q.; Li, X.; Liu, G.; Lu, X.; Zhao, J.; Qu, Y. The combination of continuous-pulse feeding hydrolysates with soybean hulls induction during fed-batch fermentation of Trichoderma reesei b5 significantly elevated the cellulase production and its degradation ability on lignocellulosic biomass. Int. J. Biol. Macromol. 2025, 307, 142244. [Google Scholar] [CrossRef]
  7. Fonseca, L.M.; Parreiras, L.S.; Murakami, M.T. Rational engineering of the Trichoderma reesei RUT-C30 strain into an industrially relevant platform for cellulase production. Biotechnol. Biofuels 2020, 13, 93. [Google Scholar] [CrossRef]
  8. Al-Qassab, A.A.; Zakaria, M.R.; Yunus, R.; Salleh, M.A.M.; Mokhtar, M.N. Investigating process parameters to enhance (hemi)cellulolytic enzymes activity produced by Trichoderma reesei RUT-C30 using deoiled oil palm mesocarp fiber in solid-state fermentation. Int. J. Biol. Macromol. 2024, 276, 134030. [Google Scholar] [CrossRef]
  9. Chen, Y.; Gao, Y.; Wang, Z.; Peng, N.; Ran, X.; Chen, T.; Liu, L.; Li, Y. The Influence of Trctf1 Gene Knockout by CRISPR–Cas9 on Cellulase Synthesis by Trichoderma reesei with Various Soluble Inducers. Fermentation 2023, 9, 746. [Google Scholar] [CrossRef]
  10. Aich, S.; Datta, S. Engineering of a highly thermostable endoglucanase from the GH7 family of bipolaris sorokiniana for higher catalytic efficiency. Appl. Microbiol. Biotechnol. 2020, 104, 3935–3945. [Google Scholar] [CrossRef] [PubMed]
  11. Contreras, F.; Pramanik, S.; Rozhkova, A.M.; Zorov, I.N.; Korotkova, O.; Sinitsyn, A.P.; Schwaneberg, U.; Davari, M.D. Engineering robust cellulases for tailored lignocellulosic degradation cocktails. Int. J. Mol. Sci. 2020, 21, 1589. [Google Scholar] [CrossRef] [PubMed]
  12. Marjamaa, K.; Rahikainen, J.; Karjalainen, M.; Maiorova, N.; Holopainen-Mantila, U.; Molinier, M.; Aro, N.; Nygren, H.; Mikkelson, A.; Koivula, A.; et al. Oxidative modification of cellulosic fibres by lytic polysaccharide monooxygenase AA9A from Trichoderma reesei. Cellulose 2022, 29, 6021–6038. [Google Scholar] [CrossRef]
  13. Adnan, M.; Ma, X.; Xie, Y.; Waheed, A.; Liu, G. Heterologously expressed cellobiose dehydrogenase acts as efficient electron-donor of lytic polysaccharide monooxygenase for cellulose degradation in Trichoderma reesei. Int. J. Mol. Sci. 2023, 24, 17202. [Google Scholar] [CrossRef]
  14. Jia, H.; Feng, X.; Huang, J.; Guo, Y.; Zhang, D.; Li, X.; Zhao, J. Recombinant family 1 carbohydrate-binding modules derived from fungal cellulase enhance enzymatic degradation of lignocellulose as novel effective accessory protein. Front. Microbiol. 2022, 13, 876466. [Google Scholar] [CrossRef]
  15. Li, Y.; Liu, C.; Bai, F.; Zhao, X. Overproduction of cellulase by Trichoderma reesei RUT C30 through batch-feeding of synthesized low-cost sugar mixture. Bioresour. Technol. 2016, 216, 503–510. [Google Scholar] [CrossRef] [PubMed]
  16. Su, Y.; Xu, Y.; Yu, X. From induction to secretion: A complicated route for cellulase production in Trichoderma reesei. Bioresour. Bioprocess. 2021, 8, 107. [Google Scholar] [CrossRef]
  17. Zhang, P.; Li, Q.; Chen, Y.; Peng, N.; Liu, W.; Wang, X.; Li, Y. Induction of cellulase production in Trichoderma reesei by a glucose–sophorose mixture as an inducer prepared using stevioside. RSC Adv. 2022, 12, 17392–17400. [Google Scholar] [CrossRef] [PubMed]
  18. Noguchi, T.; Saito, H.; Nishiyama, R.; Yoshida, N.; Matsubayashi, T.; Teshima, Y.; Yamada, C.; Hiramatsu, S.; Yamada, K.; Kagawa, Y. Isolation of a cellulase hyperproducing mutant strain of Trichoderma reesei. Bioresour. Technol. Rep. 2021, 15, 100733. [Google Scholar] [CrossRef]
  19. Wang, H.; Pang, A.; Li, B.; Huo, L.; Wu, F.; Lin, F. Intracellular sugar transporters facilitate cellulase synthesis in Trichoderma reesei using lactose. Biomolecules 2023, 13, 295. [Google Scholar] [CrossRef]
  20. Li, C.; Lin, F.; Zhou, L.; Qin, L.; Li, B.; Zhou, Z.; Jin, M.; Chen, Z. Cellulase hyper-production by Trichoderma reesei mutant SEU-7 on lactose. Biotechnol. Biofuels 2017, 10, 228. [Google Scholar] [CrossRef]
  21. Ran, X.; Gao, Y.; He, X.; Wang, Z.; Mo, Y.; Li, Y. Enhanced glucose-1-phosphate production from corn stover using cellulases with reduced β-glucosidase activity via Trbgl1 gene knockout in Trichoderma reesei Rut C30. Enzyme Microb. Technol. 2024, 180, 110503. [Google Scholar] [CrossRef] [PubMed]
  22. Long, T.; Zhang, P.; Yu, J.; Gao, Y.; Ran, X.; Li, Y. Regulation of β-Disaccharide Accumulation by β-Glucosidase Inhibitors to Enhance Cellulase Production in Trichoderma reesei. Fermentation 2022, 8, 232. [Google Scholar] [CrossRef]
  23. Xu, J.; Zhao, G.; Kou, Y.; Zhang, W.; Zhou, Q.; Chen, G.; Liu, W. Intracellular β-Glucosidases CEL1a and CEL1B Are Essential for Cellulase Induction on Lactose in Trichoderma reesei. Eukaryot. Cell 2014, 13, 1001–1013. [Google Scholar] [CrossRef]
  24. Pang, A.; Luo, Y.; Hu, X.; Zhang, F.; Wang, H.; Gao, Y.; Durrani, S.; Li, C.; Shi, X.; Wu, F.; et al. Transmembrane transport process and endoplasmic reticulum function facilitate the role of gene CEL1B in cellulase production of Trichoderma reesei. Microb. Cell Factories 2022, 21, 90. [Google Scholar] [CrossRef]
  25. Jung, K.W.; Jung, J.H.; Park, H.Y. Functional roles of homologous recombination and non-homologous end joining in DNA damage response and microevolution in cryptococcus neoformans. J. Fungi 2021, 7, 566. [Google Scholar] [CrossRef] [PubMed]
  26. Rassinger, A.; Gacek-Matthews, A.; Strauss, J.; Mach, R.L.; Mach-Aigner, A.R. Truncation of the transcriptional repressor protein Cre1 in Trichoderma reesei rut-C30 turns it into an activator. Fungal Biol. Biotechnol. 2018, 5, 15. [Google Scholar] [CrossRef] [PubMed]
  27. He, J.; Yu, B.; Zhang, K.; Ding, X.; Chen, D. Expression of a Trichoderma reesei beta-xylanase gene in Escherichia coli and activity of the enzyme on fiber-bound substrates. Protein Expr. Purif. 2009, 67, 1–6. [Google Scholar] [CrossRef]
  28. Ogawa, K.; Ohara, H.; Koide, T.; Toyama, N. Intraspecific hybridization of Trichoderma reesei by protoplast fusion. J. Ferment. Bioeng. 1989, 67, 3. [Google Scholar] [CrossRef]
  29. Ghose, T.K. Measurement of cellulase activities. Pure Appl. Chem. 1987, 59, 257–268. [Google Scholar] [CrossRef]
  30. Jalak, J.; Väljamäe, P. Mechanism of initial rapid rate retardation in cellobiohydrolase catalyzed cellulose hydrolysis. Biotechnol. Bioeng. 2010, 106, 871–883. [Google Scholar] [CrossRef]
  31. Pang, A.; Wang, H.; Luo, Y.; Yang, Z.; Liu, Z.; Wang, Z.; Li, B.; Yang, S.; Zhou, Z.; Lu, X.; et al. Dissecting cellular function and distribution of β-glucosidases in Trichoderma reesei. mBio 2021, 12, e03671-20. [Google Scholar] [CrossRef]
  32. Chai, S.; Zhu, Z.; Tian, E.; Xiao, M.; Wang, Y.; Zou, G.; Zhou, Z. Building a versatile protein production platform using engineered Trichoderma reesei. ACS Synth. Biol. 2021, 11, 486–496. [Google Scholar] [CrossRef]
  33. Li, Y.; Yu, J.; Zhang, P.; Long, T.; Mo, Y.; Li, J.; Li, Q. Comparative transcriptome analysis of Trichoderma reesei reveals different gene regulatory networks induced by synthetic mixtures of glucose and β-disaccharide. Bioresour. Bioprocess. 2021, 8, 57. [Google Scholar] [CrossRef] [PubMed]
  34. Delgado-Fernandez, P.; Plaza-Vinuesa, L.; Lizasoain-Sánchez, S.; de Las, B.; Muñoz, R.; Jimeno, M.L.; García-Doyagüez, E.; Moreno, F.J.; Corzo, N. Hydrolysis of lactose and transglycosylation of selected sugar alcohols by LacA β-galactosidase from lactobacillus plantarum WCFS1. J. Agric. Food Chem. 2020, 68, 7040–7050. [Google Scholar] [CrossRef]
  35. Häkkinen, M.; Arvas, M.; Oja, M.; Aro, N.; Penttilä, M.; Saloheimo, M.; Pakula, T.M. Re-annotation of the CAZy genes of Trichoderma reesei and transcription in the presence of lignocellulosic substrates. Microb. Cell Factories 2012, 11, 134. [Google Scholar] [CrossRef]
  36. Florindo, R.N.; Souza, V.P.; Mutti, H.S.; Camilo, C.; Manzine, L.R.; Marana, S.R.; Polikarpov, I.; Nascimento, A.S. Structural insights into β-glucosidase transglycosylation based on biochemical, structural and computational analysis of two GH1 enzymes from trichoderma harzianum. New Biotechnol. 2018, 40, 218–227. [Google Scholar] [CrossRef] [PubMed]
  37. Nawaz, K.; Anwar, W.; Subhani, M.N.; Akhter, A.; Iftikhar, S.; Khan, H.A.A.; Shahid, A.A. Glucanase gene of trichoderma; new strategy for the management of root rot disease in chili. J. Soil Sci. Plant Nutr. 2023, 24, 354–370. [Google Scholar] [CrossRef]
  38. von Freiesleben, P.; Spodsberg, N.; Stenbæk, A.; Stålbrand, H.; Krogh, K.B.R.M.; Meyer, A.S. Boosting of enzymatic softwood saccharification by fungal GH5 and GH26 endomannanases. Biotechnol. Biofuels 2018, 11, 194. [Google Scholar] [CrossRef] [PubMed]
  39. Li, B.; Walton, J.D. Functional diversity for biomass deconstruction in family 5 subfamily 5 (GH5_5) of fungal endo-β1,4-glucanases. Appl. Microbiol. Biotechnol. 2017, 101, 4093–4101. [Google Scholar] [CrossRef]
  40. Christensen, S.J.; Badino, S.F.; Cavaleiro, A.M.; Borch, K.; Westh, P. Functional analysis of chimeric TrCel6A enzymes with different carbohydrate binding modules. Protein Eng. Des. Sel. 2019, 32, 401–409. [Google Scholar] [CrossRef]
  41. Haataja, T.; Gado, J.E.; Nutt, A.; Anderson, N.T.; Nilsson, M.; Momeni, M.H.; Isaksson, R.; Väljamäe, P.; Johansson, G.; Payne, C.M.; et al. Enzyme kinetics by GH7 cellobiohydrolases on chromogenic substrates is dictated by non-productive binding: Insights from crystal structures and MD simulation. FEBS J. 2022, 290, 379–399. [Google Scholar] [CrossRef]
  42. Haviland, Z.K.; Nong, D.; Zexer, N.; Tien, M.; Anderson, C.T.; Hancock, W.O. Lignin impairs Cel7A degradation of in vitro lignified cellulose by impeding enzyme movement and not by acting as a sink. Biotechnol. Biofuels 2024, 17, 7. [Google Scholar] [CrossRef]
  43. Zhang, J.; Hong, Y.; Li, K.; Sun, Y.; Yao, C.; Ling, J.; Zhong, Y. Enhancing the production of a heterologous trametes laccase (LacA) by replacement of the major cellulase CBH1 in Trichoderma reesei. J. Ind. Microbiol. Biotechnol. 2023, 50, kuad002. [Google Scholar] [CrossRef]
  44. Zhang, J.; Li, K.; Sun, Y.; Yao, C.; Liu, W.; Liu, H.; Zhong, Y. An efficient CRISPR/Cas9 genome editing system based on a multiple sgRNA processing platform in Trichoderma reesei for strain improvement and enzyme production. Biotechnol. Biofuels Bioprod. 2024, 17, 22. [Google Scholar] [CrossRef] [PubMed]
  45. Arai, T.; Wada, M.; Nishiguchi, H.; Takimura, Y.; Ishii, J. Inducer-free recombinant protein production in Trichoderma reesei: Secretory production of endogenous enzymes and heterologous nanobodies using glucose as the sole carbon source. Microb. Cell Factories 2023, 22, 103. [Google Scholar] [CrossRef] [PubMed]
  46. Ren, M.; Wang, Y.; Liu, G.; Zuo, B.; Zhang, Y.; Wang, Y.; Liu, W.; Liu, X.; Zhong, Y. The effects of deletion of cellobiohydrolase genes on carbon source-dependent growth and enzymatic lignocellulose hydrolysis in Trichoderma reesei. J. Microbiol. 2020, 58, 687–695. [Google Scholar] [CrossRef]
  47. Zhao, C.; Zhao, J.; Han, J.; Mei, Y.; Fang, H. Improved consolidated bioprocessing for itaconic acid production by simultaneous optimization of cellulase and metabolic pathway of neurospora crassa. Biotechnol. Biofuels 2024, 17, 57. [Google Scholar] [CrossRef]
  48. Lee, D.S.; Song, Y.; Lee, Y.G.; Bae, H.J. Comparative evaluation of adsorption of major enzymes in a cellulase cocktail obtained from Trichoderma reesei onto different types of lignin. Polymers 2022, 14, 167. [Google Scholar] [CrossRef]
  49. Wang, Y.; Liu, R.; Liu, H.; Li, X.; Shen, L.; Zhang, W.; Song, X.; Liu, W.; Liu, X.; Zhong, Y. Development of a powerful synthetic hybrid promoter to improve the cellulase system of Trichoderma reesei for efficient saccharification of corncob residues. Microb. Cell Factories 2022, 21, 5. [Google Scholar] [CrossRef] [PubMed]
  50. Fitz, E.; Gamauf, C.; Seiboth, B.; Wanka, F. Deletion of the small GTPase rac1 in Trichoderma reesei provokes hyperbranching and impacts growth and cellulase production. Fungal Biol. Biotechnol. 2019, 6, 16. [Google Scholar] [CrossRef]
  51. Karlsson, J.; Siika-aho, M.; Tenkanen, M.; Tjerneld, F. Enzymatic properties of the low molecular mass endoglucanases Cel12A (EG III) and Cel45A (EG V) of Trichoderma reesei. J. Biotechnol. 2002, 99, 63–78. [Google Scholar] [CrossRef] [PubMed]
  52. Pierce, B.C.; Agger, J.W.; Wichmann, J.; Meyer, A.S. Oxidative cleavage and hydrolytic boosting of cellulose in soybean spent flakes by Trichoderma reesei Cel61A lytic polysaccharide monooxygenase. Enzym. Microb. Technol. 2017, 98, 58–66. [Google Scholar] [CrossRef] [PubMed]
  53. Pang, A.; Wang, H.; Luo, Y.; Zhang, F.; Wu, F.; Zhou, Z.; Lu, Z.; Lin, F. Investigating the cellular functions of β-glucosidases for synthesis of lignocellulose-degrading enzymes in Trichoderma reesei. Eng. Microbiol. 2023, 3, 100105. [Google Scholar] [CrossRef]
  54. Jia, H.; Sun, W.; Li, X.; Zhao, J. Cellulose induced protein 1 (Cip1) from Trichoderma reesei enhances the enzymatic hydrolysis of pretreated lignocellulose. Microb. Cell Factories 2021, 20, 136. [Google Scholar] [CrossRef]
  55. Shen, L.; Yan, A.; Wang, Y.; Wang, Y.; Liu, H.; Zhong, Y. Tailoring the expression of Xyr1 leads to efficient production of lignocellulolytic enzymes in Trichoderma reesei for improved saccharification of corncob residues. Biotechnol. Biofuels 2022, 15, 142. [Google Scholar] [CrossRef]
  56. Keller, M.B.; Badino, S.F.; Blossom, B.M.; McBrayer, B.; Borch, K.; Westh, P. Promoting and impeding effects of lytic polysaccharide monooxygenases on glycoside hydrolase activity. ACS Sustain. Chem. Eng. 2020, 8, 14117–14126. [Google Scholar] [CrossRef]
  57. Wang, H.; Shi, X.; Huo, L.; Tu, J.; Li, C.; Wu, F.; Lin, F. The discovery of novel ER-localized cellobiose transporters involved in cellulase biosynthesis in Trichoderma reesei. J. Basic Microbiol. 2024, 65, e2400573. [Google Scholar] [CrossRef]
  58. Antoniêto, A.C.C.; de Paula, R.G.; Castro, L.d.S.; Silva-Rocha, R.; Persinoti, G.F.; Silva, R.N. Trichoderma reesei CRE1-mediated Carbon Catabolite Repression in Response to Sophorose Through RNA Sequencing Analysis. Curr. Genomics 2016, 17, 119–131. [Google Scholar] [CrossRef]
  59. Havukainen, S.; Pujol-Giménez, J.; Valkonen, M.; Hediger, M.A.; Landowski, C.P. Functional characterization of a highly specific l-arabinose transporter from Trichoderma reesei. Microb. Cell Factories 2021, 20, 177. [Google Scholar] [CrossRef]
  60. Sloothaak, J.; Tamayo-Ramos, J.A.; Odoni, D.I.; Laothanachareon, T.; Derntl, C.; Mach-Aigner, A.R.; Santos, V.A.P.M.D.; Schaap, P.J. Identification and functional characterization of novel xylose transporters from the cell factories Aspergillus niger and Trichoderma reesei. Biotechnol. Biofuels 2016, 9, 148. [Google Scholar] [CrossRef] [PubMed]
  61. Huang, Z.; Chen, X.; Qin, L.; Wu, H.; Su, X.; Dong, Z. A novel major facilitator transporter TrSTR1 is essential for pentose utilization and involved in xylanase induction in Trichoderma reesei. Biochem. Biophys. Res. Commun. 2015, 460, 663–669. [Google Scholar] [CrossRef]
  62. Saloheimo, A.; Rauta, J.; Stasyk, O.V.; Sibirny, A.A.; Penttilä, M.; Ruohonen, L. Xylose transport studies with xylose-utilizing saccharomyces cerevisiae strains expressing heterologous and homologous permeases. Appl. Microbiol. Biotechnol. 2007, 74, 1041–1052. [Google Scholar] [CrossRef]
  63. Zhang, W.; Cao, Y.; Chen, G.; Liu, W. Identification of the structural determinants for efficient glucose transport via segment swapping between two fungal glucose transporters. RSC Adv. 2017, 7, 25109–25117. [Google Scholar] [CrossRef]
  64. Yan, S.; Xu, Y.; Yu, X. Role of cellulose response transporter-like protein CRT2 in cellulase induction in Trichoderma reesei. Biotechnol. Biofuels 2023, 16, 118. [Google Scholar] [CrossRef]
  65. Wang, H.; Pang, A.; Wang, W.; Li, B.; Li, C.; Wu, F.; Lin, F. Discovery of ER-localized sugar transporters for cellulase production with lac1 being essential. Biotechnol. Biofuels 2022, 15, 132. [Google Scholar] [CrossRef]
  66. Wang, Z.; Yang, R.; Lv, W.; Zhang, W.; Meng, X.; Liu, W. Functional characterization of sugar transporter CRT1 reveals differential roles of its C-terminal region in sugar transport and cellulase induction in Trichoderma reesei. Microbiol. Spectr. 2022, 10, e0087222. [Google Scholar] [CrossRef]
  67. Havukainen, S.; Valkonen, M.; Koivuranta, K.; Landowski, C.P. Studies on sugar transporter CRT1 reveal new characteristics that are critical for cellulase induction in Trichoderma reesei. Biotechnol. Biofuels 2020, 13, 158. [Google Scholar] [CrossRef] [PubMed]
  68. Fang, Y.; Meng, X.; Liu, L.; Li, Z.; Jia, K.; Liu, W. Simultaneous in vivo assembly and targeted genome integration of gene clusters in Trichoderma reesei. ACS Synth. Biol. 2025, 14, 575–584. [Google Scholar] [CrossRef] [PubMed]
  69. Sveholm, E.; Mattila, H.; Aro, N.; Valkonen, M.; Paasela, T.; Pakula, T.M. Transcriptomic and metabolic changes in Trichoderma reesei caused by mutation in xylanase regulator 1 (xyr1). Biotechnol. Biofuels 2024, 17, 106. [Google Scholar] [CrossRef] [PubMed]
  70. Liu, L.; Wang, Z.; Fang, Y.; Yang, R.; Pu, Y.; Meng, X.; Liu, W. Small GTPase Rab7 is involved in stress adaptation to carbon starvation to ensure the induced cellulase biosynthesis in Trichoderma reesei. Biotechnol. Biofuels 2024, 17, 55. [Google Scholar] [CrossRef]
  71. Maués, D.B.; Maraschin, J.C.; Duarte, D.Â.; Antoniêto, A.C.C.; Silva, R.N. Overexpression of the Transcription Factor Azf1 Reveals Novel Regulatory Functions and Impacts β-Glucosidase Production in Trichoderma reesei. J. Fungi 2023, 9, 1173. [Google Scholar] [CrossRef] [PubMed]
  72. Lv, D.; Zhang, W.; Meng, X.; Liu, W. Single mutation in transcriptional activator Xyr1 enhances cellulase and xylanase production in Trichoderma reesei on glucose. J. Agric. Food Chem. 2023, 71, 11993–12003. [Google Scholar] [CrossRef] [PubMed]
  73. Lv, D.; Zhang, W.; Meng, X.; Liu, W. A novel fusion transcription factor drives high cellulase and xylanase production on glucose in Trichoderma reesei. Bioresour. Technol. 2023, 370, 128520. [Google Scholar] [CrossRef]
  74. Li, N.; Qiu, Z.; Cai, W.; Shen, Y.; Wei, D.; Chen, Y.; Wang, W. The ras small GTPase RSR1 regulates cellulase production in Trichoderma reesei. Biotechnol. Biofuels 2023, 16, 87. [Google Scholar] [CrossRef]
  75. Li, N.; Li, J.; Chen, Y.; Shen, Y.; Wei, D.; Wang, W. Mechanism of Zn2+ regulation of cellulase production in Trichoderma reesei Rut-C30. Biotechnol. Biofuels Bioprod. 2023, 16, 73. [Google Scholar] [CrossRef]
  76. Arai, T.; Ichinose, S.; Shibata, N.; Kakeshita, H.; Kodama, H.; Igarashi, K.; Takimura, Y. Inducer-free cellulase production system based on the constitutive expression of mutated XYR1 and ACE3 in the industrial fungus Trichoderma reesei. Sci. Rep. 2022, 12, 19445. [Google Scholar] [CrossRef] [PubMed]
  77. Chen, Y.; Wang, W.; Liu, P.; Lin, A.; Fan, X.; Wu, C.; Li, N.; Wei, L.; Wei, D. The novel repressor Rce2 competes with Ace3 to regulate cellulase gene expression in the filamentous fungus Trichoderma reesei. Mol. Microbiol. 2021, 116, 1298–1314. [Google Scholar] [CrossRef]
  78. Sun, Y.; Qian, Y.; Zhang, J.; Yao, C.; Wang, Y.; Liu, H.; Zhong, Y. Development of a novel expression platform for heterologous protein production via deleting the p53-like regulator Vib1 in Trichoderma reesei. Enzym. Microb. Technol. 2022, 155, 109993. [Google Scholar] [CrossRef]
  79. Chen, X.; Song, B.; Liu, M.; Qin, L.; Dong, Z. Understanding the role of Trichoderma reesei Vib1 in gene expression during cellulose degradation. J. Fungi 2021, 7, 613. [Google Scholar] [CrossRef]
  80. Ullah, S.F.; Oreb, M.; Boles, E.; Srivastava, V.; Seidl-Seiboth, V.; Seiboth, B.; Kappel, L. N-acetylglucosamine sensing in the filamentous soil fungus Trichoderma reesei. FEBS J. 2025, 292, 3072–3090. [Google Scholar] [CrossRef]
  81. Fekete, E.; Karaffa, L.; Aghcheh, R.K.; Németh, Z.; Fekete, É.; Orosz, A.; Paholcsek, M.; Stágel, A.; Kubicek, C.P. The transcriptome of lae1 mutants of Trichoderma reesei cultivated at constant growth rates reveals new targets of LAE1 function. BMC Genom. 2014, 15, 447. [Google Scholar] [CrossRef] [PubMed]
  82. Xin, Q.; Gong, Y.; Lv, X.; Chen, G.; Liu, W. Trichoderma reesei histone acetyltransferase Gcn5 regulates fungal growth, conidiation, and cellulase gene expression. Curr. Microbiol. 2013, 67, 580–589. [Google Scholar] [CrossRef] [PubMed]
  83. Zheng, F.; Cao, Y.; Lv, X.; Wang, L.; Li, C.; Zhang, W.; Chen, G.; Liu, W. A copper-responsive promoter replacement system to investigate gene functions in Trichoderma reesei: A case study in characterizing SAGA genes. Appl. Microbiol. Biotechnol. 2016, 101, 2067–2078. [Google Scholar] [CrossRef]
  84. Shen, L.; Gao, J.; Wang, Y.; Li, X.; Liu, H.; Zhong, Y. Engineering the endoplasmic reticulum secretory pathway in Trichoderma reesei for improved cellulase production. Enzym. Microb. Technol. 2021, 152, 109923. [Google Scholar] [CrossRef]
  85. Heydenreich, R.M.J. The Impact of the Unfolded Protein Response on Enzyme Secretion in Trichoderma reesei. Ph.D. Thesis, Technische Universität Wien, Vienna, Austria, 2020. Available online: https://repositum.tuwien.at/handle/20.500.12708/15474 (accessed on 19 April 2025).
  86. Neves-da-Rocha, J.; Santos-Saboya, M.J.; Lopes, M.E.; Rossi, A.; Martinez-Rossi, N. Insights and perspectives on the role of proteostasis and heat shock proteins in fungal infections. Microorganisms 2023, 11, 1878. [Google Scholar] [CrossRef]
  87. Zhang, F.; Li, J.; Champreda, V.; Liu, C.; Bai, F.; Zhao, X. Global reprogramming of gene transcription in Trichoderma reesei by overexpressing an artificial transcription factor for improved cellulase production and identification of Ypr1 as an associated regulator. Front. Bioeng. Biotechnol. 2020, 8, 649. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The Trcel1b gene was knocked out in T. reesei Δura5 using the CRISPR-Cas9 system. (A) Schematic of the cel1b gene knockout strategy and the transformants T. reesei ΔCEL1B1 and ΔCEL1B4. (B) PCR confirmation of the successful insertion of the Poura5 expression cassette into the cel1b gene. (C) DNA sequencing of the Poura5 expression cassette in T. reesei ΔCEL1B1 and ΔCEL1B4. (D) Growth of T. reesei RUT C30, Δura5, ΔCEL1B1, and ΔCEL1B4 on MM plates without uracil and PDA plates containing 5-fluoroorotic acid. (E) Copy number determination of the Poura5 expression cassette in the genomes of T. reesei RUT C30, ΔCEL1B1, and ΔCEL1B4 by qPCR.
Figure 1. The Trcel1b gene was knocked out in T. reesei Δura5 using the CRISPR-Cas9 system. (A) Schematic of the cel1b gene knockout strategy and the transformants T. reesei ΔCEL1B1 and ΔCEL1B4. (B) PCR confirmation of the successful insertion of the Poura5 expression cassette into the cel1b gene. (C) DNA sequencing of the Poura5 expression cassette in T. reesei ΔCEL1B1 and ΔCEL1B4. (D) Growth of T. reesei RUT C30, Δura5, ΔCEL1B1, and ΔCEL1B4 on MM plates without uracil and PDA plates containing 5-fluoroorotic acid. (E) Copy number determination of the Poura5 expression cassette in the genomes of T. reesei RUT C30, ΔCEL1B1, and ΔCEL1B4 by qPCR.
Fermentation 11 00439 g001
Figure 2. Cellulase production by T. reesei RUT C30, ΔCEL1B1, and ΔCEL1B4 using 10 g/L MGD or lactose as the carbon source. Each value represents the mean of three biological replicates. Error bars indicate standard deviation. Statistical significance was assessed using the t-test; p < 0.05 was considered statistically significant (*), and p > 0.05 was denoted as not significant (ns).
Figure 2. Cellulase production by T. reesei RUT C30, ΔCEL1B1, and ΔCEL1B4 using 10 g/L MGD or lactose as the carbon source. Each value represents the mean of three biological replicates. Error bars indicate standard deviation. Statistical significance was assessed using the t-test; p < 0.05 was considered statistically significant (*), and p > 0.05 was denoted as not significant (ns).
Fermentation 11 00439 g002
Figure 3. Extracellular protein concentrations and SDS-PAGE analysis of T. reesei RUT C30, ΔCEL1B1, and ΔCEL1B4 induced by 10 g/L MGD or lactose. Statistical significance was assessed using the t-test; p < 0.05 was considered statistically significant (*), and p > 0.05 was denoted as not significant (ns).
Figure 3. Extracellular protein concentrations and SDS-PAGE analysis of T. reesei RUT C30, ΔCEL1B1, and ΔCEL1B4 induced by 10 g/L MGD or lactose. Statistical significance was assessed using the t-test; p < 0.05 was considered statistically significant (*), and p > 0.05 was denoted as not significant (ns).
Fermentation 11 00439 g003
Figure 4. Transcriptomic analysis of differentially expressed genes (DEGs) in T. reesei RUT C30 and ΔCEL1B under MGD or lactose induction. (A) Volcano plots showing changes in gene expression levels; red dots indicate significantly upregulated genes, green/blue dots indicate significantly downregulated genes, and black dots indicate genes without significant changes. (B) Gene Ontology (GO) enrichment analysis of DEGs.
Figure 4. Transcriptomic analysis of differentially expressed genes (DEGs) in T. reesei RUT C30 and ΔCEL1B under MGD or lactose induction. (A) Volcano plots showing changes in gene expression levels; red dots indicate significantly upregulated genes, green/blue dots indicate significantly downregulated genes, and black dots indicate genes without significant changes. (B) Gene Ontology (GO) enrichment analysis of DEGs.
Fermentation 11 00439 g004
Figure 5. Expression profiles of glycoside hydrolase (GH) family genes (CAZy) based on transcriptomic data. Average FPKM (fragments per kilobase of transcript per million mapped reads) values of GH genes expressed by T. reesei RUT C30 and ΔCEL1B when cultured in MGD or lactose.
Figure 5. Expression profiles of glycoside hydrolase (GH) family genes (CAZy) based on transcriptomic data. Average FPKM (fragments per kilobase of transcript per million mapped reads) values of GH genes expressed by T. reesei RUT C30 and ΔCEL1B when cultured in MGD or lactose.
Fermentation 11 00439 g005
Figure 6. Top 10 differentially expressed transporter genes in T. reesei RUT C30 and ΔCEL1B under MGD or lactose induction. Data are shown as heatmaps, with color coding of relative expression values as follows: red = 2; white = 0; purple = −2 (values represent log2 fold changes). Previously reported transporters are annotated.
Figure 6. Top 10 differentially expressed transporter genes in T. reesei RUT C30 and ΔCEL1B under MGD or lactose induction. Data are shown as heatmaps, with color coding of relative expression values as follows: red = 2; white = 0; purple = −2 (values represent log2 fold changes). Previously reported transporters are annotated.
Fermentation 11 00439 g006
Figure 7. Top 10 differentially expressed transcription factor genes in T. reesei RUT C30 and ΔCEL1B under MGD or lactose induction. Data are shown as heatmaps, with color coding of relative expression values as follows: red = 3; white = 0; purple = −2 (values represent log2 fold changes).
Figure 7. Top 10 differentially expressed transcription factor genes in T. reesei RUT C30 and ΔCEL1B under MGD or lactose induction. Data are shown as heatmaps, with color coding of relative expression values as follows: red = 3; white = 0; purple = −2 (values represent log2 fold changes).
Fermentation 11 00439 g007
Figure 8. Correlation between RNA-seq and RT-qPCR data. Comparison of log2 fold changes for 21 genes measured by RNA-seq and RT-qPCR. cDNA was amplified from each RNA-seq sample for RT-qPCR analysis. A strong and statistically significant Pearson correlation was observed between gene expression levels determined by RT-qPCR and RNA-seq.
Figure 8. Correlation between RNA-seq and RT-qPCR data. Comparison of log2 fold changes for 21 genes measured by RNA-seq and RT-qPCR. cDNA was amplified from each RNA-seq sample for RT-qPCR analysis. A strong and statistically significant Pearson correlation was observed between gene expression levels determined by RT-qPCR and RNA-seq.
Fermentation 11 00439 g008
Table 1. Log2-fold change (FC) of the main secreted cellulases expressed in MGD/lactose.
Table 1. Log2-fold change (FC) of the main secreted cellulases expressed in MGD/lactose.
CategoryGene IDDescription/NameMGD Log2FCLac Log2FC
Cellulase125125Exoglucanase I/CEL7A−1.32.4
122470Exoglucanase II/CEL6A−1.72.6
5304Endoglucanase I/CEL7B−1.63.4
72489Endoglucanase III/CEL5A−1.72.5
124438Endoglucanase II/CEL12A−1.43.3
74496Endoglucanase V/CEL45ANSNS
136547β-glucosidase/CEL3A−0.83.0
82126β-glucosidase/CEL3H−1.6NS
Auxiliary protein104220Swollenin−1.52.3
139633Endoglucanase IV (AA9)NS2.0
122518Endoglucanase VII (AA9)−1.21.8
121449Cellulose binding domain/CIP1−1.52.2
125575Cellulose binding domain/CIP2−1.62.3
NS, nonsignificant at p > 0.05; Lac: Lactose.
Table 2. Log2-fold change (FC) of characterized transporters in MGD/lactose.
Table 2. Log2-fold change (FC) of characterized transporters in MGD/lactose.
Gene IDNameMGD Up/DwonLac Up/DwonDescription
97259Hgt1NSUpGlucose transporter
109243Crt1NSUpCellulose sensor
136988Stp1NSUpCellobiose transporter
138519Str1DownUpXylose transporter
33630Xlt1DownUpXylose transporter
38765 NSUpXylose transporter
91594 DownUpCellobiose/Xylose transporter
127980 DownUpCellodextrin transporter
55710 NSUpPermease of the major facilitator superfamily
137001Crt2NSUpCellulose sensor
137795 NSUpCellobiose transporter
79984 NSUpCellobiose transporter
124396Hxt1NSDownGlucose transporter
NS, nonsignificant at p > 0.05; Lac: Lactose.
Table 3. Log2-fold change (FC) of characterized transcription factors involved in the regulation of cellulase genes.
Table 3. Log2-fold change (FC) of characterized transcription factors involved in the regulation of cellulase genes.
Gene IDNamePositive/Negative-ActingMGD Up/DownMGD Log2FCLac Up/DownLac Log2FC
98788Xyr1PositiveNS−0.1 Up1.7
98455Ace3PositiveNS−0.7 Up1.8
125610Vib1PositiveNS−0.2 Up1.3
122363Ace1NegativeNS0.2 NS−0.6
32395Ace2PositiveNS−0.1 NS0.5
93466Hap2PositiveNS0.1 NS−0.1
24298Hap3PositiveNS−0.1 NS0.2
95791PaccNegativeNS0.6 NS−0.2
140814AreaPositiveNS0.3 NS0.1
91236BglrPositiveNS0.3 NS0.1
68701Clr-1PositiveNS0.3 NS0.3
76250Clr-2PositiveNS−0.2 NS0.7
93861Ypr1PositiveNS−0.5 NS0.8
112571Tmac1PositiveNS0.2 NS−0.3
23706Cre1NegativeNS−0.6 NS0.7
6520Rce1NegativeNS−0.2 NS0.7
NS, nonsignificant at p > 0.05; Lac: lactose.
Table 4. Differential transcription of genes involved in protein processing in the endoplasmic reticulum (ER).
Table 4. Differential transcription of genes involved in protein processing in the endoplasmic reticulum (ER).
Gene IDNameMGD Up/DwonLac Up/DwonDescription
124246Fes1NSUpHsp70 nucleotide exchange factor
121926 NSUpHeat shock protein 70 kDa
102206 NSUpHeat shock protein 90
122594 NSUpDnaJ domain-containing protein
25648Bip1DownUpHeat shock protein 70 kDa
83445ERManINSUp1, 2-α-mannosidase
137683NEFNSUpArmadillo/beta-catenin-like repeat-containing protein
101105ERManINSUp1, 2-α-mannosidase
116265SPCS1DownNSMicrosomal signal peptidase 12 kDa subunit
NS, nonsignificant at p > 0.05; Lac: Lactose.
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

Wang, L.; Fan, J.; He, X.; Cheng, J.; Zhang, X.; Tian, T.; Li, Y. Transcriptomic Analysis Reveals Opposing Roles of CEL1B in Sophorose- and Lactose-Induced Cellulase Expression in Trichoderma reesei Rut C30. Fermentation 2025, 11, 439. https://doi.org/10.3390/fermentation11080439

AMA Style

Wang L, Fan J, He X, Cheng J, Zhang X, Tian T, Li Y. Transcriptomic Analysis Reveals Opposing Roles of CEL1B in Sophorose- and Lactose-Induced Cellulase Expression in Trichoderma reesei Rut C30. Fermentation. 2025; 11(8):439. https://doi.org/10.3390/fermentation11080439

Chicago/Turabian Style

Wang, Lu, Junping Fan, Xiao He, Jian Cheng, Xinyan Zhang, Tian Tian, and Yonghao Li. 2025. "Transcriptomic Analysis Reveals Opposing Roles of CEL1B in Sophorose- and Lactose-Induced Cellulase Expression in Trichoderma reesei Rut C30" Fermentation 11, no. 8: 439. https://doi.org/10.3390/fermentation11080439

APA Style

Wang, L., Fan, J., He, X., Cheng, J., Zhang, X., Tian, T., & Li, Y. (2025). Transcriptomic Analysis Reveals Opposing Roles of CEL1B in Sophorose- and Lactose-Induced Cellulase Expression in Trichoderma reesei Rut C30. Fermentation, 11(8), 439. https://doi.org/10.3390/fermentation11080439

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