Skip to main content

Tailoring the expression of Xyr1 leads to efficient production of lignocellulolytic enzymes in Trichoderma reesei for improved saccharification of corncob residues

Abstract

Background

The filamentous fungus Trichoderma reesei is extensively used for the industrial-scale cellulase production. It has been well known that the transcription factor Xyr1 plays an important role in the regulatory network controlling cellulase gene expression. However, the role of Xyr1 in the regulation of cellulase expression has not been comprehensively elucidated, which hinders further improvement of lignocellulolytic enzyme production.

Results

Here, the expression dosage of xyr1 was tailored in T. reesei by differentially overexpressing the xyr1 gene under the control of three strong promoters (Pegl2, Pcbh1, and Pcdna1), and the transcript abundance of xyr1 was elevated 5.8-, 12.6-, and 47.2-fold, respectively. We found expression of cellulase genes was significantly increased in the Pegl2-driven xyr1 overexpression strain QE2X, whereas relatively low in the Pcbh1- and Pcdna1-driven overexpression strains. We also found that the Pegl2-driven overexpression of xyr1 caused a more significant opening of chromatin in the core promoter region of the prominent cellulase genes. Furthermore, the cellulase activity showed a 3.2-fold increase in the strain QE2X, while insignificant improvement in the Pcbh1- and Pcdna1-driven strains. Finally, the saccharification efficiency toward acid-pretreated corncob residues containing high-content lignin by the crude enzyme from QE2X was increased by 57.2% compared to that from the parental strain. Moreover, LC–MS/MS and RT-qPCR analysis revealed that expression of accessory proteins (Cip1, Cip2, Swo1, and LPMOs) was greatly improved in QE2X, which partly explained the promoting effect of the Pegl2-driven overexpression on enzymatic hydrolysis of lignocellulose biomass.

Conclusions

Our results underpin that the precise tailoring expression of xyr1 is essential for highly efficient cellulase synthesis, which provide new insights into the role of Xyr1 in regulating cellulase expression in T. reesei. Moreover, these results also provides a prospective strategy for strain improvement to enhance the lignocellulolytic enzyme production for use in biorefinery applications.

Introduction

Lignocellulosic biomass is an abundantly available, inexpensive organic resource on the earth, which is basically composed of cellulose, hemicellulose, and lignin [1]. Biological degradation of lignocellulose biomass by lignocellulolytic enzymes into fermentable sugars for the production of biofuels and other biochemicals has received extensive attention [2, 3]. However, the high cost of cellulase production, which is the major barrier for practical applications, limits the development of an efficient and economically viable lignocellulosic feedstock based biorefinery [4].

The filamentous fungus Trichoderma reesei encodes a wide repertoire of cellulase, hemicellulase and accessory proteins to degrade cellulose and other biomass components, which makes it be extensively employed in the industrial cellulase production [5]. The cellulases secreted from T. reesei mainly include three enzyme components, namely, cellobiohydrolases (CBHs, EC 3.2.1.91), endoglucanases (EGs, EC3.2.1.4), and β-glucosidase (exactly BGL1, EC 3.2.1.21) [6]. In general, CBHs and EGs act cooperatively to degrade cellulose into cellobiose, and BGL1 ultimately hydrolyzes the oligosaccharide into glucose [6]. The three components work synergistically to accomplish a complete deconstruction of cellulose [7]. Optimizing the composition of cellulase complexes by genetic engineering to strengthen the synergism has been proved to be practical [8, 9], among which the overexpression of BGL or EGs is a powerful strategy for enhancing the synergistic effect [10,11,12]. Recently, Qian et al. demonstrated that the double overexpression of EG2 and BGL1 can significantly ameliorate the performance on the cellulase yield and saccharification efficiency in T. reesei [13]. Moreover, it is known that cellulase gene expression is stringently controlled by transcriptional regulatory networks in T. reesei [14]. Therefore, engineering the transcriptional factors can improve the entire cellulase system, which has been considered to be an effective way for enhancing cellulase production.

The energy-efficient production of cellulase in fungi is strictly regulated by a suit of transcription factors [15]. So far, several transcription factors involved in the modulation of cellulase gene expression have been identified, including at least five transcription activators (Xyr1, Ace2, Ace3, Vib1, and the Hap/2/3/5 complex) and four repressors (Cre1, Ace1, Rce1, and Ctf1) [16,17,18,19]. Other transcription factors, such as BglR, Pac1, and Crz1, have also been identified to regulate the biosynthesis of cellulase [20,21,22]. Especially, Xyr1 is the transcriptional activator modulating the expression of cellulase and hemicellulase genes in T. reesei [24]. Overexpression of xyr1 can markedly enhance cellulase production in T. reesei [26, 28]. In addition, Xyr1 also seems to be involved in the glucose repression of cellulase mediated by the carbon catabolite repressor Cre1, since its constitutive expression being able to reduce this repressive effect [4, 25, 26]. It has been reported that the expression levels of the main cellulase genes (cbh1 and cbh2) appeared to be strictly dependent on the expression level of xyr1 [25]. Zheng et al. overexpressed the xyr1 gene under the control of the copper repressible promoter Ptcu1 and the transcript level was improved by approximately twofold on Avicel [27]. However, it seemed that the efficiency of the tcu1 promoter in enhancing transcription of the major cellulase gene cbh1 remains lower compared to the strong pyruvate decarboxylase gene (pdc) promoter, with the transcript level of xyr1 was increased by 16-fold under the similar condition [27, 28]. As noted above, tuning the expression dosage of xyr1 may have profound effects on cellulase expression in T. reesei, which requires more systematical and in-depth investigation.

In the present study, the expression of xyr1 was tailored with three differential strong promoters (Pegl2, Pcbh1, and Pcdna1) in T. reesei. We found that the overexpression of xyr1 driven by Pegl2, not that driven by the stronger promoter Pcbh1 or Pcdna1, leads to more significantly enhanced cellulase production. Meanwhile, it also resulted in a significant opening of chromatin in the core promoter regions of cellulase genes, which was accompanied by the activation of the corresponding gene expression. Furthermore, the overexpression of xyr1 with Pegl2 also remarkably improved the levels of accessory proteins involved in lignocellulose degradation and thus promoted the saccharification efficiency toward acid-pretreated corncob residues.

Results

Overexpression of the xyr1 gene using the promoters with different strengths in T. reesei

To systematically investigate the effect of differential expression of xyr1 on cellulase production in T. reesei, the xyr1 overexpression cassettes driven by the cellulose-inducible promoters Pegl2, Pcbh1, and the strong constitutive promoter Pcdna1 were constructed and precisely integrated by homologous recombination into the hph locus of T. reesei QEB4, respectively (Fig. 1A). The strains harboring the xyr1 gene driven by the promoters Pegl2, Pcbh1, and Pcdna1 were designated QE2X, QCBX, and QCDX, respectively. The integration of the xyr1 gene into the genome of T. reesei QEB4 was verified by PCR (data not shown). Furthermore, the transcript levels of xyr1 were analyzed by RT-qPCR (Fig. 1B). It was found that xyr1 was significantly overexpressed in the strains, with the maximum transcript levels in the QCDX strain, followed by QCBX and QE2X, which were 47.2-, 12.6-, and 5.8-fold higher than the parental strain QEB4, respectively.

Fig. 1
figure 1

Construction and validation of the xyr1 overexpression strains. A Schematic diagrams of the expression cassettes used for differential overexpression of the xyr1 gene. The overexpression of xyr1 was controlled by three different promoters (Pegl2, Pcbh1, and Pcdna1), respectively. B Transcript levels of xyr1 which was determined by RT-qPCR in QEB4 and three independent transformants of each xyr1 overexpression strain (QE2X, QCBX, and QCDX). Strains were cultivated at 30 ℃ and 200 rpm in flasks using the CPM medium with 2% (w/v) Avicel as the sole carbon source for 72 h. The transcript level was normalized to that of actin (t-test, **p < 0.01). Results are means of three biological replicates and error bars indicate ± SD

Next, the three xyr1 overexpression strains QE2X, QCBX, and QCDX were cultured on minimal medium (MM) plates containing glucose, glycerol, and lactose as the carbon source, respectively, as well as on PDA plates to investigate the effect of differential xyr1 overexpression on mycelial phenotypes of T. reesei. As shown in Additional file 1: Fig. S1, the colonies of the recombinant strains extended normally and grew at a rate similar to that of the parental strain, indicating that different dosage levels of xyr1 had little effect on mycelial growth of T. reesei.

Thereafter, the recombinant strains were inoculated on microcrystalline cellulose (MCC), CMC, and CMC-esculin plates to assess the cellulase secretion capacity (Fig. 2). These strains displayed wider hydrolytic haloes or black zones around the colonies relative to the parental strain, suggesting that the cellulase secretion ability was enhanced to different extents. Especially, the cellulase secretion in QE2X strain with the moderately strong promoter Pegl2 was far greater than that in the other two strains with stronger promoters (Pcbh1 and Pcdna1) on the plates, implying that there may be a dose-dependent correlation between the expression level of xyr1 and the secretion of cellulase in T. reesei.

Fig. 2
figure 2

Detection of the cellulase secretion by T. reesei QEB4 and the xyr1 overexpression strains in different culture plates. The strains were assessed on the MCC plate (A), the CMC plate (C), and the CMC-esculin plate (E) to evaluate the secretion capacity of total cellulase, EGs and BGL, respectively. B The ratios between the diameter of the transparent zones and the colonies on MCC plate. D The hydrolytic halo diameters of the strains on CMC-Na plate. F The black zone diameters of the strains on CMC-esculin plate. *p < 0.05; **p < 0.01 (t-test). Results are means of three biological replicates and error bars indicate ± SD

Influences of differential overexpression of xyr1 on the transcriptional profile of cellulase genes

It is known that transcription of the cellulase-encoding genes are modulated by the transcriptional activator Xyr1 [23]. Here, the transcript levels of five main cellulase-encoding genes cbh1, cbh2, egl1, egl2, and bgl1 were measured by RT-qPCR in the xyr1 overexpression strains (Fig. 3A). It was found that the transcript quantities of the five cellulase genes in the QE2X strain with the lowest degree of xyr1 overexpression (5.8-fold) were 7.3-, 5.0-, 1.4-, 4.8-, and 5.8-fold higher than those of the parental strain, respectively. However, those in the QCBX strain with the stronger xyr1 overexpression (12.6-fold) were only slightly increased by 3.2-, 2.5-, 0.9-, 1.2-, and 2.6-fold, respectively. Unexpectedly, the transcript levels of these cellulase genes in the QCDX strain with the highest degree of xyr1 overexpression (47.2-fold) were not significantly improved. Taken together, these results demonstrate that cellulase gene transcription is precisely regulated by the xyr1 transcript abundance, i.e., the moderately strong overexpression of xyr1 with the Pegl2 promoter, but not the stronger overexpression with the Pcbh1 or Pcdna1 promoter, could lead to more pronounced improvement in cellulase expression.

Fig. 3
figure 3

Transcript analysis by RT-qPCR for genes encoding cellulases and regulators. Genes encoding cellulases (A) and transcription factors (B) were analyzed in the xyr1 overexpression strains with T. reesei QEB4 as the control. Strains were cultivated at 30℃ and 200 rpm in flasks using the CPM medium with 2% (w/v) Avicel as the sole carbon source for 72 h. The transcript levels of genes were normalized to that of actin (t-test, *p < 0.05, **p < 0.01, ns = not significant). Results are means of three biological replicates and error bars indicate ± SD

Furthermore, transcript analysis of several transcription factors involved in cellulase expression were performed (Fig. 3B). The positive transcriptional regulators-encoding genes ace2, ace3, and vib1 were virtually unchanged in all the recombinant strains. However, the negative transcriptional regulator-encoding genes cre1, ace1, and ctf1 were greatly decreased in both QE2X and QCBX. As for the QCDX strain, the expression of ace1 and ctf1 showed no significant changes, while the carbon catabolite repressor-encoding gene cre1 was downregulated. These findings suggest that overexpression of xyr1 probably resulted in downregulation of the negative regulators, thus facilitating improvement of the expression of cellulase genes.

Differential overexpression of xyr1 regulated the chromatin accessibility of cellulase genes and the native xyr1 gene

Eukaryotic gene expression is often accompanied by chromatin remodeling [29]. It was reported that the deletion of xyr1 in T. reesei resulted in the loss of expression of the cellulase genes cbh1 and cbh2 and the reduction in chromatin accessibility of these genes [30]. To gain further insight into the impact of xyr1 dosage, the chromatin packing of the core promoter region of cbh1 and cbh2 in the xyr1-overexpressing strains was analyzed by CHART-PCR (Fig. 4A, B). In all the recombinant strains, the chromatin accessibility index (CAI) in the promoter region of cbh1 and cbh2 was improved to varying degree which was corresponded with the high expression levels. This suggests that the xyr1 overexpression can lead to more open chromatin status of cellulase genes. As expected, remarkable chromatin opening of cbh1 and cbh2 was detected in QE2X, indicating that the egl2 promoter-driven xyr1 overexpression is more efficacious in promoting the chromatin accessibility of cellulase-encoding genes than the stronger cbh1/cdna1 promoter-driven overexpression.

Fig. 4
figure 4

Transcript and CHART analysis of T. reesei QEB4 and the xyr1 overexpression strains. The genes of cbh1 (A), cbh2 (B), and the native xyr1 (C) were determined by RT-qPCR and CHART-PCR, respectively. The strains were cultured in CPM with 2% (w/v) Avicel for 48 h. All data were normalized to the reference gene actin. The transcript levels are depicted on the x-axis and the chromatin accessibility indices (CAI) are plotted on the y-axis. Results are means of three biological replicates and error bars indicate ± SD

It was also known that the expression level of xyr1 may correlate with the degree of chromatin opening in its own promoter [31]. Hence, the chromatin packing of the native xyr1 core promoter region in the xyr1-overexpressing strains was analyzed (Fig. 4C). More open chromatin status was found in all the recombinant strains, with the most significant chromatin opening in QE2X. These results further corroborate the view that the dosage level of xyr1 differentially influenced the chromatin status of its own promoter.

Differential overexpression of xyr1 induced the unfolded protein response in the endoplasmic reticulum

As the major secreted proteins in T. reesei, the newly synthesized cellulase enzymes should enter the endoplasmic reticulum (ER) secretory pathway where they fold into the correct three-dimensional structure with the assistance of resident ER folding factors for secretion, whereas those that misfold would be efficiently degraded by the ER-associated degradation (ERAD) [32]. Meanwhile, the accumulation of unfolded proteins in the ER can cause ER stress, which elicits the unfolded protein response (UPR), a mechanism to upregulate UPR-regulated genes involved in protein folding, ERAD, and others for the restoration of ER homeostasis [33]. To investigate the effect of differential expression of xyr1 on the secretory pathway, the expression of genes encoding key components involved in protein folding (pdi1 and bip1) and the ERAD pathway (hrd1 and der1) was determined in the xyr1-overexpressing strains. As shown in Fig. 5, all tested genes (pdi1, bip1, hrd1, and der1) were significantly upregulated in QE2X, while only the folding factor genes (pdi1 and bip1) were upregulated in QCBX and QCDX. These above results demonstrate that the xyr1 overexpression could induce the UPR in the ER. In particular, the strong ER stress evoked in QE2X resulted in significant activation of the ERAD pathway, indicating a large amount of cellulase has been accumulated in the ER, i.e., saturation of the secretory pathway may be reached in this strain.

Fig. 5
figure 5

Transcript analysis of the genes encoding the ER-associated components by RT-qPCR. The UPR- (A) and ERAD- (B) related genes were analyzed in the xyr1 overexpression strains with T. reesei QEB4 as the control. Strains were cultivated at 30 ℃ and 200 rpm in flasks using CPM with 2% (w/v) Avicel as the sole carbon source for 72 h. The transcript levels of genes were normalized to that of actin (t-test, *p < 0.05, **p < 0.01, ns = not significant). Results are means of three biological replicates and error bars indicate ± SD

Pegl2-driven xyr1 overexpression greatly enhanced cellulase production in T. reesei

To investigate whether the cellulase production levels were improved in the xyr1 overexpression strains, the activities of cellulase in the fermentation broths were determined under Avicel-inducing conditions. The total cellulase activity of QE2X reached 8.4 IU/mL, 3.2-fold higher than that of the parental strain (2.0 IU/mL), using filter paper activity (FPA) as a characterization method, while the FPA in QCBX and QCDX was only elevated 0.9- and 0.8-fold compared to that of the parental strain, respectively (Fig. 6A). Consistently, QE2X also displayed a significant enhancement in the activities of the cellulase components, with 2.1-, 2.4-, and 1.8-fold increase in CBHs, EGs, and BGL activities, respectively. However, QCBX and QCDX only demonstrated 1.4- and 1.3-fold improvement in CBH activity, 1.9- and 1.7-fold improvement in EG activity as well as 1.2- and 1.1-fold improvement in BGL activity, respectively (Fig. 6B–D). In addition, the concentration of extracellular protein in QE2X reached 3.0 mg/mL, much higher than that in QCBX and QCDX (Fig. 6E). Taken together, these results indicate that the egl2 promoter-driven xyr1 overexpression can significantly enhance cellulase production in T. reesei.

Fig. 6
figure 6

Cellulase activities and total extracellular proteins of T. reesei QEB4 and the xyr1 overexpression strains. A FPase activity (FPA), B cellobiohydrolase (CBH) activity, C endoglucanase (EG) activity, D β-glucosidase (BGL) activity, and E the total extracellular protein of the fermentation supernatant from QEB4 and three recombinant strains cultured on 2% (w/v) Avicel for 7 days. *p < 0.05; **p < 0.01, ns = not significant (t-test). Results are means of three biological replicates and error bars indicate ± SD

Pegl2-driven overexpression of xyr1 optimized the lignocellulolytic enzyme system for saccharification of corncob residues

Among the three xyr1 overexpression strains, the Pegl2-driven overexpression strain QE2X showed the highest cellulase activity, and hence, it was selected for subsequent saccharification. Two differently pretreated corncob residues, acid-pretreated (ACR) and delignified (DCR) corncob residues, were utilized as substrates to be saccharified with the enzyme preparations produced by QE2X, with the parental strain QEB4 as control. As shown in Fig. 7, the glucose release from the QE2X enzyme preparation in the saccharification of ACR was 11.9 mg/mL, which was 57.2% higher than that from QEB4. However, no significant difference was observed in the saccharification of DCR between those from QE2X and QEB4. Since the difference in composition between the two corncob substrates is that ACR contains higher levels of lignin than DCR, these results demonstrate that the Pegl2-driven overexpression of xyr1 can optimize the lignocellulolytic enzyme system for saccharification of the high lignin-containing lignocellulosic substrates.

Fig. 7
figure 7

Saccharification of different pretreated corncob residues by T. reesei QEB4 and QE2X. Saccharification of acid-pretreated corncob residue (A) and delignified corncob residue (B) with the same FPA dosage. **p < 0.01, ns = not significant (t-test). Results are means of 3 biological replicates and error bars indicate ± SD

It is known that accessory proteins, such as glucuronoyl esterase (Cip1 and Cip2), swollenin (Swo1), and AA9 lytic polysaccharide monooxygenases (LPMOs), play important roles in enhancing lignocellulose degradation [34]. Especially, Cip1 can reduce the non-productive adsorption of lignin by cellulase and Cip2 acts in the cleavage of hemicellulose-lignin crosslinks [35, 36]. Ma et al. demonstrated by RNA sequencing in T. reesei that Xyr1 could stimulate the expression of those accessory protein-encoding genes [37]. Thus, the transcript levels of the above accessory protein-encoding genes were firstly examined. The transcript levels of cip1 and cip2 in QE2X were improved significantly, which were 21- and 18-fold higher than those in the parental strain QEB4, respectively (Fig. 8). Furthermore, the swollenin-encoding gene swo1 and two LPMO-encoding genes cel61a and cel61b were about 3.4-, 1.3-, and 0.7-fold higher in QE2X than those in QEB4, respectively (Fig. 8). Meanwhile, the LC–MS/MS analysis of the secretome of T. reesei QE2X and QEB4 was carried out. It was found that, besides the increased amounts of cellulases, the PSMs of all the above-mentioned accessory proteins in QE2X were also higher than those in QEB4 when equal protein was used for evaluation (Table 1), suggesting more accessory proteins were present in the enzyme preparations of QE2X. These results indicate that the Pegl2-driven overexpression of xyr1 can optimize the lignocellulolytic enzyme system of T. reesei by improving the expression of those accessory proteins, which is beneficial for efficient degradation of lignocellulosic biomass.

Fig. 8
figure 8

Transcript analysis for genes encoding accessory proteins of T. reesei QEB4 and QE2X by RT-qPCR. The transcript levels of genes were normalized to that of actin (t-test, *p < 0.05, **p < 0.01, ns = not significant). Results are means of three biological replicates and error bars indicate ± SD

Table 1 Identified accessory proteins in the 7d fermentation supernatants of T. reesei by LC–MS/MS

Discussion

In T. reesei, genetic engineering of transcriptional regulators has been used as a powerful tool for enhancing cellulase production. Xyr1 is the key transcriptional activator of lignocellulolytic genes, whose overexpression can improve cellulase yields under both inducing and non-inducing conditions [26]. However, the effect of xyr1 dosage regulation on cellulase expression has not yet been elucidated. Herein, the expression of xyr1 was tailored under promoters of different strengths and we found that the xyr1 overexpression with the Pegl2 promoter resulted in a substantial improvement in cellulase production and saccharification efficiency.

In previous studies, the promoter Ppdc was chosen to drive xyr1 expression for the construction of cellulase hyperproducing strain [4, 28, 38]. In addition, the promoter Ptcu1 was also used to the expression of xyr1, and the resultant strain could achieve full cellulase expression [27]. By roughly comparing the effects of the two promoters, we speculate that the overexpression of xyr1 driven by the stronger promoter may have a more pronounced influence on cellulase production. However, this does not imply that the maximization of cellulases can be achieved by extremely strong overexpression of xyr1. Bao et al. found that the moderate overexpression of Sec16 was more potent to increase protein secretion in Saccharomyces cerevisiae than the strong overexpression [39]. Thus, uncertain relationships between the degree of xyr1 overexpression and cellulase gene expression in T. reesei limit the potential for cellulase production to some extent. To further determine whether the effect of xyr1 overexpression depends on its dosage, we selected three strong promoters for tailoring the expression of xyr1: the main cellulase-encoding genes promoters Pegl2 and Pcbh1, as well as the strong constitutive promoter Pcdna1. Among the xyr1 overexpression strains, the strain QE2X with the egl2 promoter exhibited the highest cellulase production, which was consistent with the significantly enhanced cellulase secretion capacity and cellulase gene transcription (Figs. 2, 3A and 6). These findings further suggest that the robust upregulation of cellulase genes caused by the Pegl2-driven overexpression of xyr1 result in the outstanding cellulase production observed in the QE2X strain.

It has been reported that the level of xyr1 transcription tightly regulate the main cellulase genes expression [25]. However, our results showed that the Pegl2-driven xyr1 overexpression (5.8-fold) markedly improved both the transcription and chromatin accessibility of cellulase genes, while the effects of the xyr1 overexpression driven by the cbh1 and cdna1 promoters (12.6- and 47.2-fold) were not particularly remarkable (Figs. 3A, 4A, B). It has been reported that upon cellulase induction, Xyr1 is synthesized and rapidly imports into the nucleus in order to regulate cellulase gene expression [40]. In order to investigate the effect of differential xyr1 overexpression on the Xyr1 protein level, the nuclear protein extracts of the T. reesei strains were also prepared and determined by LC–MS/MS in this study. More peptides of Xyr1 could be detected in QE2X than the other two strains, QCBX and QCDX (Additional file 3: Table S2). That is, the Pegl2-driven xyr1 overexpression, instead of the Pcbh1/Pcdna1-driven overexpression, significantly improved the amount of the Xyr1 protein in the nucleus. It is known that a phenomenon of “quelling” in filamentous fungi displays the characteristic feature of post-transcriptional gene silencing (PTGS), which functions by mediating mRNA degradation or translational suppression [41, 42]. There may be a “quelling” mechanism in T. reesei in which the excessive mRNA of xyr1 causes translation repression, ultimately leading to a reduction in the amount of Xyr1 in the nucleus. Thus, it can be speculated that the transcript levels of cbh1 and cbh2 do not correspond to the transcript levels of xyr1 to some extent, but strictly to its protein levels.

Meanwhile, Xyr1 has been implicated in chromatin remodeling with histone modification, which has a regulatory role on cellulase gene expression in T. reesei [43]. Mello-de-Sousa et al. have demonstrated that the deletion of xyr1 could trigger chromatin compaction in the upstream regions of the cellulase genes (cbh1 and cbh2) under inducing conditions, which suggests that Xyr1 is necessary for the chromatin opening of these regions [30]. Our results further confirmed this conclusion. The cbh1 and cbh2 promoters of the xyr1-overexpressing strains were more accessible compared to the parental strain (Fig. 4A, B). However, the chromatin packing pattern of cbh1 and cbh2 genes did not exactly follow the transcript level of the xyr1 gene. Among the three strong promoters tested, the moderately strong one, i.e., the Pegl2-driven overexpression of xyr1 was more beneficial for the chromatin accessibility of cellulase-encoding genes, which resulted in the higher gene expression. Furthermore, we also confirmed that Xyr1 could influence the chromatin remodeling of xyr1 itself. The opening of the native xyr1 promoter is more pronounced in the xyr1-overexpressing strains than in the parental strain (Fig. 4C). Till et al. have reported the presence of Xyr1 regulatory element in the xyr1 promoter, which is involved in the Xyr1 autoregulatory mechanism [44]. Thus, the increase of chromatin accessibility of the xyr1 gene may be related to the above mechanism, which warrants further investigation.

BGL is the rate-limiting enzyme in the cellulose hydrolysis process, whose overexpression is beneficial for optimization of cellulase system [12]. However, the overexpression of xyr1 is not adequate to mediate the high-level expression of BGL, which may be attributed to the presence of less Xyr1 binding sites (XBS) on the bgl promoter [4, 45]. In the parental strain QEB4, the introduction of bgl gene under control of the strong cbh1 promoter containing more XBS makes the native Xyr1 a limiting step to further improve the expression of bgl [13]. In this study, the overexpression of xyr1 in the strain QEB4 resulted in an increase in the production of BGL by a maximum of 1.8-fold (Fig. 6D). This result indicates that simultaneous engineering of transcription factors and cellulase genes could be a feasible strategy for improvement of cellulase production.

It is known that the efficiency of saccharification of lignocellulosic substrates is closely related to the performance of lignocellulases [11]. However, the composition of the enzyme complexes secreted by T. reesei may not be optimal for industrial applications due to the presence of certain enzymes in limiting amounts [9, 46]. Optimization of enzyme complex with different proportions of cellulase components and accessory proteins has been employed for highly efficient bioconversion of lignocellulosic biomass [12, 47]. In addition, lignocellulose is embedded in an amorphous matrix of cross-linked lignin and hemicellulose, which limits the cellulase accessibility to substrates [48, 49]. Especially, lignin is considered as the rate-limiting factor in the hydrolysis of lignocellulosic biomass as it impedes the access of cellulase to cellulose leading to non-productive binding [50, 51]. Hence, delignification of lignocellulosic biomass can improve the saccharification efficiency and saccharide yields [52, 53]. BSA and surfactants have also been demonstrated to prevent cellulase–lignin interactions to some extent [54, 55]. Furthermore, exogenous addition of Cip1 promoted the degradation of lignocellulosic substrates with high lignin [36]. Analysis of the extracellular proteins of QE2X by LC–MS/MS demonstrated the higher amounts of accessory proteins (Cip1, Cip2, Swo1, and LPMO) in QE2X than the parental strain QEB4 (Table 1), which altered the diversity of enzyme mixtures. Thus, the hydrolysis efficiency of ACR through the crude enzyme produced by QE2X had been greatly improved, showing the Pegl2-driven xyr1 overexpression could optimize the lignocellulolytic enzyme system by improving expression of accessory proteins.

Conclusion

In the present study, we showed that the Pegl2-driven xyr1 overexpression significantly improved production of lignocellulolytic enzymes in T. reesei (Fig. 9). The total cellulase activity of QE2X was up to 8.4 IU/mL under inducing condition. The glucose release from ACR by the enzyme mixture of QE2X was improved by 57.2%. To the best of our knowledge, this is the first report regarding the roles of xyr1 dosage level on cellulase expression in T. reesei. The significantly enhanced production of cellulase in QE2X is attributed to the moderate upregulation of transcription factor, with the underlying mechanisms require further exploration. In conclusion, this study demonstrates that selecting the promoter with proper dosage to drive the expression of key regulators is a feasible strategy to enhance lignocellulolytic enzyme production for the conversion of lignocellulose biomass.

Fig. 9
figure 9

Schematic summary showing the proposed model for the dosage regulation of xyr1 in T. reesei. A Compared to the strong xyr1 overexpression with the cbh1/cdna1 promoter, the moderate xyr1 overexpression with the egl2 promoter can result in more open chromatin and stronger expression of cellulase gene, thus significantly enhancing cellulase production. B The lignocellulolytic enzyme production and saccharification efficiency were significantly improved in T. reesei QE2X, because of the high expression of cellulase and accessory proteins

Materials and methods

Strains, medium, and culture conditions

T. reesei QEB4, an EG2-BGL1 double overexpression strain [13], was used as the parental strain in this study. The strain QEB4 was constructed from the uracil auxotrophic strain QP4 whose pyr4 gene was replaced with the resistance gene hph. The fungal strain and its derivatives were cultivated on potato dextrose agar (PDA) plates at 30 ℃ for 5–7 days to produce conidia, which were harvested, and then inoculated with approximately 106 spores/mL into 50 mL of liquid minimal medium (MM) [56] before incubation for 36 h, 30 ℃ and 200 rpm. Then, equal amounts (0.4 g cell wet weight) of mycelia were transferred into 100 mL of the cellulase production medium (CPM) in 500 mL Erlenmeyer flasks. The CPM solution was composed as follows: 2% (w/v) Avicel, 0.1% CaCl2·2H2O, 0.5% (NH4)2SO4, 0.5% KH2PO4, 0.06% MgSO4·7H2O, and 2% corn steep liquor.

Construction of the xyr1 overexpression strains

The xyr1 overexpression cassettes were constructed with the double-joint PCR method [57]. The Phanta® Super-Fidelity DNA Polymerase (Vazyme Biotech Co., Ltd., Nanjing, China) was used for PCR amplification, and DNA fragments were purified using Gel Extraction Kit (Omega, USA). The primers were designed using the primer premier 5.0 software. Primer synthesis and DNA sequencing were performed at BGI Genomics (Qingdao, China). The primers used in this study are presented in Additional file 2: Table S1. The plasmid T-ptrA was used as the template to clone the ptrA expression cassette by PtrA-F/PtrA-R. The 5′- and 3′-flank fragments of hph, the three promoters (egl2, cbh1, and cdna1), and the encoding region and terminator of xyr1 were amplified from the genomic DNA of T. reesei QEB4 using the primer pairs Hph-UF/Hph-UR, Hph-DF/Hph-DR, Egl2-F/Egl2-R, Cbh1-F/Cbh1-R, Cdna1-F/Cdna1-R, and Xyr1-F/Xyr1-R, respectively. Subsequently, the purified DNA fragments were fused together in 1:2:4:2:1 molar ratio of 5′-flanking region:ptrA: promoter:xyr1:3′-flanking region and were used as the template to amplify the final expression cassettes by Hph-cUF/Hph-cDR. The cassettes were transformed into the protoplasts of T. reesei QEB4 using the method described previously, respectively [56]. The strains were screened on MM agar plates containing 300 μg/mL pyrithiamine, and the purified candidate transformants were identified and named as QE2X, QCBX, and QCDX, respectively.

RNA extraction and RT-qPCR analysis

The fungal mycelia were collected and flash-frozen in liquid nitrogen. Total RNA was extracted using the Trizol reagent (TaKaRa, Japan), and the RNA concentration was quantified by using a NanoDrop LiTE Spectrophotometer (THERMO SCIENTIFIC, United States). 1 μg RNA was reverse transcribed to cDNA using PrimeScript RT reagent Kit (TaKaRa, Japan) following the manufacturer’s protocol. RT-qPCR analysis was performed with SYBR Premix Ex Taq™ (TaKaRa, Japan) and the LigntCycler 480 system (Roche, Mannheim, Germany) using the primers listed in Additional file 2: Table S1. Three biological replicates were carried out for each sample. Transcript levels of target genes were analyzed according to the ΔΔCt method using the actin gene for normalization.

Growth and secretion ability analysis

To analyze the growth morphology on plate, the parental and recombinant strains were pre-grown on MM agar plates and then equal amounts of mycelia were transferred in biological duplicates onto PDA plates or MM plates containing 2% glucose, 2% glycerol, or 2% lactose, respectively, and incubated at 30 ℃ for 2 days. Mycelia diameters were measured every 12 h for 2 days and recorded as representation of growth rates.

To determine the secretion ability of total cellulase, the equal amount of mycelia was inoculated on MCC plate containing 0.05% ball-milled Avicel, 0.02% peptone on the upper layer, and water agar on the bottom layer. After 5 days incubation, the double-layer MCC plate with hydrolysis zones was measured and recorded by photographs. In addition, the secretion ability of EGs and BGL1 in recombinant strains was assayed by the CMC-esculin plates (1% CMC-Na, 0.3% esculin, 0.05% ferric citrate and 2% agar) and the CMC plates (1% CMC-Na, 0.1% yeast extract and 2% agar), respectively.

Enzyme activity and protein assay

The activities of FPase (FPA) and EGs were determined by measuring the released reducing sugar with filter paper and CMC-Na as substrates, respectively. Determination of FPA was performed in a 2 mL reaction system containing 500 μL of the appropriately diluted culture supernatant, 1.5 mL of 50 mM citrate buffer (pH 4.8), and 0.05 g of Whatman No.1 filter paper. The EGs activity assay was carried out at 50 ℃ in a 2 mL reaction system including 500 μL of the suitably diluted supernatant and 1.5 mL of 0.5% CMC sodium in 50 mM citrate buffer (PH 4.8). The DNS method was used to measure the amount of released reducing sugars [58]. The CBHs and BGL activities were determined by measuring the amount of released p-nitrophenol using p-nitrophenyl-D-cellobioside (pNPC, Sigma, USA) and p-nitrophenyl-β-D-glucopyranoside (pNPG, Sigma, USA) as the substrates, respectively. The assays were performed in 200 μL reaction mixtures containing 50 μL of the diluted supernatant, 50 μL of the respective substrate, and 100 μL of 50 mM citrate buffer (pH 4.8) and then incubated at 50 ℃ for 30 min. One unit (U) of enzyme activity was defined as the amount of enzymes liberating 1 µmoL reducing sugars (FPA, EGs) or p-nitrophenol (CBHs, BGL1) per minute under the test conditions. Total extracellular proteins were assayed as previously described by Ellilä et al. [4]. Briefly, proteins were precipitated with ice-cold acetone and the concentration was quantified using the Bradford Kit (Sangon Biotech, Shanghai, China) with bovine serum albumin (BSA) as a standard. To facilitate analyzing the amount of the Xyr1 protein, total nuclear protein extracts were prepared from the T. reesei strains cultured on 2% (w/v) Avicel for 3 days. And the extraction experiment was performed using the Filamentous fungi Nuclear Protein Extraction Kit (BestBio, Shanghai, China) according to the manufacturer’s instructions.

Saccharification of the pretreated corncob residues

For the saccharification assay, ACR and DCR were applied as substrates in the saccharification process. ACR consists of 62.6% cellulose, 2.4% hemicellulose, 17.7% lignin, and 6.8% ash, while DCR contains 65.7% cellulose, 1.8% hemicellulose, 3.2% lignin, and 5.9% ash [59]. The crude enzyme preparations were placed in 30 mL reaction volume in 100 mL flasks containing 50 mM citrate buffer (pH 4.8), 1.5 g substrate, and 30 μL Proclin 300 preservative, and the reaction mixture was incubated at 200 rpm, 50 ℃. Enzyme loading was adjusted to the same FPA (10 FPU/g substrate). Glucose released was detected by the SBA-40C biological sensor analyzer (BISAS, Shandong, China) after incubation for 48 h.

Southern blot analysis

The specific probe was amplified from the genome of T. reesei QEB4 using the primer pair Hph-pF/Hph-pR. Chromosomal DNA of the parental and recombinant strains was digested thoroughly by Mph1103I and then hybridized by the probe. Finally, the probe-hybridized DNA fragments were detected and visualized using a DIG-High Prime DNA Labeling and Detection Starter Kit I (Roche, Germany) according to the manufacturer’s instructions.

Chromatin accessibility real‑time PCR (CHART‑PCR) assays

CHART-PCR assay were performed according to a previously described protocol [60]. Briefly, mycelia were collected after 48 h of induction, filtered, and ground in liquid nitrogen. Subsequently, 0.1 g powder was incubated with 10 μL RNase-free DNase I (TaKaRa Biotechnology) for 5 min at 37 ℃ in 1 mL of nuclease digestion buffer (250 mM sucrose, 0.05 mM CaCl2, 3 mM MgCl2, 15 mM NaCl, 60 mM KCl, 5 mM DTT, and 15 mM pH 7.5 Tris–HCl). The reaction was terminated by adding 100 μL of termination buffer (20 mM EDTA and 2% SDS). Then, two rounds of phenol–chloroform extraction and one round of chloroform extraction were followed for protein extraction. The supernatant was treated with RNase A (10 μg/mL) at 37 ℃ for 15 min and precipitated with 0.3 M NaAc and two volumes of ethanol. The DNA obtained were dissolved in 30 μL of double-distilled water. A negative control reaction without DNase I was included. Quantitative PCR analysis on the DNase I-treatment samples was performed to measure the relative abundance of target regions. Each sample was prepared in triplicate. The chromatin accessibility index (CAI) was defined as CAI = Dc/Ds, where Dc is the amount of intact DNA detected for the promoter regions of actin gene and Ds is the amount of intact DNA detected for each target region. The amount of intact input DNA for each sample was calculated by comparing the thresholds of the PCR amplification plots with the standard curve produced for each primer set using serial dilutions of undigested genomic DNA. All primer sequences are provided in Additional file 2: Table S1.

LC–MS/MS

LC–MS/MS analyses were performed according to the method described previously with appropriate modifications [61]. The fermentation supernatants of three biological replicates were mixed in equal proportions and then subjected to the measurement. The sample (100 μg of protein, measured by the Bradford method) was denatured in an equal volume of 8 M urea and reduced in 5 mM DTT at 56 ℃ for 30 min. The samples were then alkylated in 14 mM iodoacetamide (IAA) for 1 h in darkness. The unreacted IAA was quenched by the addition of 5 mM DTT for 15 min. The samples were digested by adding trypsin (Sigma, USA) with 1 mM CaCl2 as a co-factor at 37 ℃ for 12 h, and reaction was terminated by adjusting to pH 2 with trifluoroacetic acid. The digested oligopeptides were desalted on C18 Sep-Pak cartridges (Waters Associates, USA) and were further dissolved in 0.1% formic acid and then were subjected to nanoelectrospray ionization, followed by analysis in LTQ Orbitrap Velos Pro (Thermo Scientific, USA) coupled with HPLC system. The Orbitrap resolution was set at 70,000, and data from LC–MS/MS analysis were searched by proteome discovered software 1.4 (Thermo Fisher Scientific). Sequences were mapped based on the reference genome of T. reesei, acquired from the JGI Genome Portal (https://mycocosm.jgi.doe.gov/Trire2/Trire2.home.html). Furthermore, the relative protein abundance was characterized by peptide spectrum matches (PSMs), which could correlate linearly with the protein abundance [62].

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

CBHs:

Cellobiohydrolases

EGs:

Endoglucanases

BGL:

β-Glucosidase

MM:

Minimal medium

CPM:

Cellulase production medium

CMC-Na:

Sodium carboxymethy cellulose

pNPG:

4-Nitrophenyl-β-D-glucopyranoside

pNPC:

P-nitrophenyl-β-D-cellobioside

FPA:

Filter paper activity

BSA:

Bovine serum albumin

ACR:

Acid-pretreated corncob residues

DCR:

Delignified corncob residues

CAI:

Chromatin accessibility index

PSMs:

Peptide spectrum matches

XBS:

Xyr1 binding sites

PTGS:

Post-transcriptional gene silencing

References

  1. Dashtban M, Schraft H, Qin W. Fungal bioconversion of lignocellulosic residues; opportunities & perspectives. Int J Biol Sci. 2009;5(6):578–95.

    Article  CAS  Google Scholar 

  2. Guo H, Chang Y, Lee DJ. Enzymatic saccharification of lignocellulosic biorefinery: research focuses. Bioresour Technol. 2018;252:198–215.

    Article  CAS  Google Scholar 

  3. Liu G, Qu Y. Engineering of filamentous fungi for efficient conversion of lignocellulose: tools, recent advances and prospects. Biotechnol Adv. 2019;37(4):519–29.

    Article  CAS  Google Scholar 

  4. Ellilä S, Fonseca L, Uchima C, Cota J, Goldman GH, Saloheimo M, Sacon V, Siika-Aho M. Development of a low-cost cellulase production process using Trichoderma reesei for Brazilian biorefineries. Biotechnol Biofuels. 2017;10:30.

    Article  Google Scholar 

  5. Liu G, Qin Y, Li Z, Qu Y. Development of highly efficient, low-cost lignocellulolytic enzyme systems in the post-genomic era. Biotechnol Adv. 2013;31(6):962–75.

    Article  CAS  Google Scholar 

  6. Kubicek CP, Mikus M, Schuster A, Schmoll M, Seiboth B. Metabolic engineering strategies for the improvement of cellulase production by Hypocrea jecorina. Biotechnol Biofuels. 2009;2:19.

    Article  Google Scholar 

  7. Bischof RH, Ramoni J, Seiboth B. Cellulases and beyond: the first 70 years of the enzyme producer Trichoderma reesei. Microb Cell Fact. 2016;15(1):106.

    Article  Google Scholar 

  8. Druzhinina IS, Kubicek CP. Genetic engineering of Trichoderma reesei cellulases and their production. Microb Biotechnol. 2017;10(6):1485–99.

    Article  CAS  Google Scholar 

  9. Fang H, Li C, Zhao J, Zhao C. Biotechnological advances and trends in engineering Trichoderma reesei towards cellulase hyperproducer. Biotechnol Bioprocess Eng. 2021;26(4):517–28.

    Article  CAS  Google Scholar 

  10. Miettinen-Oinonen A, Suominen P. Enhanced production of Trichoderma reesei endoglucanases and use of the new cellulase preparations in producing the stonewashed effect on denim fabric. Appl Environ Microbiol. 2002;68(8):3956–64.

    Article  CAS  Google Scholar 

  11. Li Y, Zhang X, Xiong L, Mehmood MA, Zhao X, Bai F. On-site cellulase production and efficient saccharification of corn stover employing cbh2 overexpressing Trichoderma reesei with novel induction system. Biores Technol. 2017;238:643–9.

    Article  CAS  Google Scholar 

  12. Gao J, Qian Y, Wang Y, Qu Y, Zhong Y. Production of the versatile cellulase for cellulose bioconversion and cellulase inducer synthesis by genetic improvement of Trichoderma reesei. Biotechnol Biofuels. 2017;10:272.

    Article  Google Scholar 

  13. Qian Y, Zhong L, Gao J, Sun N, Wang Y, Sun G, Qu Y, Zhong Y. Production of highly efficient cellulase mixtures by genetically exploiting the potentials of Trichoderma reesei endogenous cellulases for hydrolysis of corncob residues. Microb Cell Fact. 2017;16(1):207.

    Article  Google Scholar 

  14. Tani S, Kawaguchi T, Kobayashi T. Complex regulation of hydrolytic enzyme genes for cellulosic biomass degradation in filamentous fungi. Appl Microbiol Biotechnol. 2014;98(11):4829–37.

    Article  CAS  Google Scholar 

  15. Amore A, Giacobbe S, Faraco V. Regulation of cellulase and hemicellulase gene expression in fungi. Curr Genomics. 2013;14(4):230–49.

    Article  CAS  Google Scholar 

  16. Portnoy T, Margeot A, Seidl-Seiboth V, Le Crom S, Ben Chaabane F, Linke R, Seiboth B, Kubicek CP. Differential regulation of the cellulase transcription factors XYR1, ACE2, and ACE1 in Trichoderma reesei strains producing high and low levels of cellulase. Eukaryot Cell. 2011;10(2):262–71.

    Article  CAS  Google Scholar 

  17. Zhang F, Zhao X, Bai F. Improvement of cellulase production in Trichoderma reesei Rut-C30 by overexpression of a novel regulatory gene Trvib-1. Biores Technol. 2018;247:676–83.

    Article  CAS  Google Scholar 

  18. Cao Y, Zheng F, Wang L, Zhao G, Chen G, Zhang W, Liu W. Rce1, a novel transcriptional repressor, regulates cellulase gene expression by antagonizing the transactivator Xyr1 in Trichoderma reesei. Mol Microbiol. 2017;105(1):65–83.

    Article  CAS  Google Scholar 

  19. Meng QS, Zhang F, Liu CG, Zhao XQ, Bai FW. Identification of a novel repressor encoded by the putative gene ctf1 for cellulase biosynthesis in Trichoderma reesei through artificial zinc finger engineering. Biotechnol Bioeng. 2020;117(6):1747–60.

    Article  CAS  Google Scholar 

  20. Nitta M, Furukawa T, Shida Y, Mori K, Kuhara S, Morikawa Y, Ogasawara W. A new Zn(II)(2)Cys(6)-type transcription factor BglR regulates β-glucosidase expression in Trichoderma reesei. Fungal Genet Biol. 2012;49(5):388–97.

    Article  CAS  Google Scholar 

  21. He R, Ma L, Li C, Jia W, Li D, Zhang D, Chen S. Trpac1, a pH response transcription regulator, is involved in cellulase gene expression in Trichoderma reesei. Enzym Microb Technol. 2014;67:17–26.

    Article  CAS  Google Scholar 

  22. Chen L, Zou G, Wang J, Wang J, Liu R, Jiang Y, Zhao G, Zhou Z. Characterization of the Ca(2+) -responsive signaling pathway in regulating the expression and secretion of cellulases in Trichoderma reesei Rut-C30. Mol Microbiol. 2016;100(3):560–75.

    Article  CAS  Google Scholar 

  23. Mach-Aigner AR, Pucher ME, Steiger MG, Bauer GE, Preis SJ, Mach RL. Transcriptional regulation of xyr1, encoding the main regulator of the xylanolytic and cellulolytic enzyme system in Hypocrea jecorina. Appl Environ Microbiol. 2008;74(21):6554–62.

    Article  CAS  Google Scholar 

  24. Stricker AR, Grosstessner-Hain K, Würleitner E, Mach RL. Xyr1 (xylanase regulator 1) regulates both the hydrolytic enzyme system and D-xylose metabolism in Hypocrea jecorina. Eukaryot Cell. 2006;5(12):2128–37.

    Article  CAS  Google Scholar 

  25. Derntl C, Gudynaite-Savitch L, Calixte S, White T, Mach RL, Mach-Aigner AR. Mutation of the Xylanase regulator 1 causes a glucose blind hydrolase expressing phenotype in industrially used Trichoderma strains. Biotechnol Biofuels. 2013;6(1):62.

    Article  CAS  Google Scholar 

  26. Lv X, Zheng F, Li C, Zhang W, Chen G, Liu W. Characterization of a copper responsive promoter and its mediated overexpression of the xylanase regulator 1 results in an induction-independent production of cellulases in Trichoderma reesei. Biotechnol Biofuels. 2015;8:67.

    Article  Google Scholar 

  27. Zheng F, Yang R, Cao Y, Zhang W, Lv X, Meng X, Zhong Y, Chen G, Zhou Q, Liu W. Engineering Trichoderma reesei for hyperproduction of cellulases on glucose to efficiently saccharify pretreated corncobs. J Agric Food Chem. 2020;68(45):12671–82.

    Article  CAS  Google Scholar 

  28. Wang S, Liu G, Wang J, Yu J, Huang B, Xing M. Enhancing cellulase production in Trichoderma reesei RUT C30 through combined manipulation of activating and repressing genes. J Ind Microbiol Biotechnol. 2013;40(6):633–41.

    Article  CAS  Google Scholar 

  29. Wu C. Chromatin remodeling and the control of gene expression. J Biol Chem. 1997;272(45):28171–4.

    Article  CAS  Google Scholar 

  30. Mello-de-Sousa TM, Rassinger A, Pucher ME, dos Santos CL, Persinoti GF, Silva-Rocha R, Poças-Fonseca MJ, Mach RL, Nascimento Silva R, Mach-Aigner AR. The impact of chromatin remodelling on cellulase expression in Trichoderma reesei. BMC Genomics. 2015;16(1):588.

    Article  Google Scholar 

  31. Mello-de-Sousa TM, Rassinger A, Derntl C, Poças-Fonseca MJ, Mach RL, Mach-Aigner AR. The relation between promoter chromatin status, Xyr1 and cellulase Ex-pression in Trichoderma reesei. Curr Genomics. 2016;17(2):145–52.

    Article  CAS  Google Scholar 

  32. Sun Z, Brodsky JL. Protein quality control in the secretory pathway. J Cell Biol. 2019;218(10):3171–87.

    Article  CAS  Google Scholar 

  33. Cao SS, Kaufman RJ. Unfolded protein response. Curr Biol. 2012;22(16):R622–6.

    Article  CAS  Google Scholar 

  34. Lehmann L, Rønnest NP, Jørgensen CI, Olsson L, Stocks SM, Jørgensen HS, Hobley T. Linking hydrolysis performance to Trichoderma reesei cellulolytic enzyme profle. Biotechnol Bioeng. 2016;113:1001–10.

    Article  CAS  Google Scholar 

  35. 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 Fact. 2021;20(1):136.

    Article  CAS  Google Scholar 

  36. Li XL, Spániková S, de Vries RP, Biely P. Identification of genes encoding microbial glucuronoyl esterases. FEBS Lett. 2007;581(21):4029–35.

    Article  CAS  Google Scholar 

  37. Ma L, Chen L, Zhang L, Zou G, Liu R, Jiang Y, Zhou Z. RNA sequencing reveals Xyr1 as a transcription factor regulating gene expression beyond carbohydrate metabolism. Biomed Res Int. 2016;2016:4841756.

    Article  Google Scholar 

  38. Fonseca LM, Parreiras LS, Murakami MT. Rational engineering of the Trichoderma reesei RUT-C30 strain into an industrially relevant platform for cellulase production. Biotechnol Biofuels. 2020;13:93.

    Article  CAS  Google Scholar 

  39. Bao J, Huang M, Petranovic D, Nielsen J. Moderate expression of SEC16 increases protein secretion by Saccharomyces cerevisiae. Appl Environ Microbiol. 2017;83(14):e03400-e3416.

    Article  CAS  Google Scholar 

  40. Lichius A, Seidl-Seiboth V, Seiboth B, Kubicek CP. Nucleo-cytoplasmic shuttling dynamics of the transcriptional regulators XYR1 and CRE1 under conditions of cellulase and xylanase gene expression in Trichoderma reesei. Mol Microbiol. 2014;94(5):1162–78.

    Article  CAS  Google Scholar 

  41. Li L, Chang SS, Liu Y. RNA interference pathways in filamentous fungi. Cell Mol Life Sci. 2010;67(22):3849–63.

    Article  CAS  Google Scholar 

  42. Romano N, Macino G. Quelling: transient inactivation of gene expression in Neurospora crassa by transformation with homologous sequences. Mol Microbiol. 1992;6(22):3343–53.

    Article  CAS  Google Scholar 

  43. Gupta VK, Steindorff AS, de Paula RG, Silva-Rocha R, Mach-Aigner AR, Mach RL, Silva RN. The post-genomic era of Trichoderma reesei: What’s next? Trends Biotechnol. 2016;34(12):970–82.

    Article  CAS  Google Scholar 

  44. Till P, Derntl C, Kiesenhofer DP, Mach RL, Yaver D, Mach-Aigner AR. Regulation of gene expression by the action of a fungal lncRNA on a transactivator. RNA Biol. 2020;17(1):47–61.

    Article  CAS  Google Scholar 

  45. Xia Y, Yang L, Xia L. Combined strategy of transcription factor manipulation and β-glucosidase gene overexpression in Trichoderma reesei and its application in lignocellulose bioconversion. J Ind Microbiol Biotechnol. 2018;45(9):803–11.

    Article  CAS  Google Scholar 

  46. Zhou J, Wang YH, Chu J, Luo LZ, Zhuang YP, Zhang SL. Optimization of cellulase mixture for efficient hydrolysis of steam-exploded corn stover by statistically designed experiments. Bioresour Technol. 2009;100(2):819–25.

    Article  CAS  Google Scholar 

  47. Sun FF, Hong J, Hu J, Saddler JN, Fang X, Zhang Z, Shen S. Accessory enzymes influence cellulase hydrolysis of the model substrate and the realistic lignocellulosic biomass. Enzyme Microb Technol. 2015;79–80:42–8.

    Article  Google Scholar 

  48. Jørgensen H, Kristensen JB, Felby C. Enzymatic conversion of lignocellulose into fermentable sugars: challenges and opportunities. Biofuels Bioprod Biorefin. 2007;1:119–34.

    Article  Google Scholar 

  49. Jeoh T, Ishizawa CI, Davis MF, Himmel ME, Adney WS, Johnson DK. Cellulase digestibility of pretreated biomass is limited by cellulose accessibility. Biotechnol Bioeng. 2007;98:112–22.

    Article  CAS  Google Scholar 

  50. Orth AB, Denny M, Tien M. Overproduction of lignin-degrading enzymes by an isolate of Phanerochaete chrysosporium. Appl Environ Microbiol. 1991;57(9):2591–6.

    Article  CAS  Google Scholar 

  51. Shrestha S, Fonoll X, Khanal SK, Raskin L. Biological strategies for enhanced hydrolysis of lignocellulosic biomass during anaerobic digestion: current status and future perspectives. Bioresour Technol. 2017;245(Pt A):1245–57.

    Article  CAS  Google Scholar 

  52. Gupta R, Mehta G, Khasa YP, Kuhad RC. Fungal delignification of lignocellulosic biomass improves the saccharification of cellulosics. Biodegradation. 2011;22(4):797–804.

    Article  CAS  Google Scholar 

  53. Várnai A, Viikari L, Marjamaa K, Siika-aho M. Adsorption of monocomponent enzymes in enzyme mixture analyzed quantitatively during hydrolysis of lignocellulose substrates. Bioresour Technol. 2011;102:1220–7.

    Article  Google Scholar 

  54. Yang B, Wyman CE. BSA treatment to enhance enzymatic hydrolysis of cellulose in lignin containing substrates. Biotechnol Bioeng. 2006;94:611–7.

    Article  CAS  Google Scholar 

  55. Kristensen JB, Borjesson J, Bruun MH, Tjerneld F, Jorgensen H. Use of surface active additives in enzymatic hydrolysis of wheat straw lignocellulose. Enzyme Microb Technol. 2007;40:888–95.

    Article  CAS  Google Scholar 

  56. Penttilä M, Nevalainen H, Rättö M, Salminen E, Knowles J. A versatile transformation system for the cellulolytic filamentous fungus Trichoderma reesei. Gene. 1987;61(2):155–64.

    Article  Google Scholar 

  57. Yu JH, Hamari Z, Han KH, Seo JA, Reyes-Domínguez Y, Scazzocchio C. Double-joint PCR: a PCR-based molecular tool for gene manipulations in filamentous fungi. Fungal Genet Biol. 2004;41(11):973-81.73.

    Article  CAS  Google Scholar 

  58. Ghose TK. Measurement of cellulase activities. Pure Appl Chem. 1987;59:257–68.

    Article  CAS  Google Scholar 

  59. Liu K, Lin X, Yue J, Li X, Fang X, Zhu M, Lin J, Qu Y, Xiao L. High concentration ethanol production from corncob residues by fed-batch strategy. Bioresour Technol. 2010;101(13):4952–8.

    Article  CAS  Google Scholar 

  60. Mello-de-Sousa TM, Gorsche R, Rassinger A, Poças-Fonseca MJ, Mach RL, Mach-Aigner AR. A truncated form of the Carbon catabolite repressor 1 increases cellulase production in Trichoderma reesei. Biotechnol Biofuels. 2014;7:129.

    Article  Google Scholar 

  61. Ribeiro DA, Cota J, Alvarez TM, Brüchli F, Bragato J, Pereira BM, Pauletti BA, Jackson G, Pimenta MT, Murakami MT, Camassola M, Ruller R, Dillon AJ, Pradella JG, Paes Leme AF, Squina FM. The Penicillium echinulatum secretome on sugar cane bagasse. PLoS ONE. 2012;7(12):e50571.

    Article  CAS  Google Scholar 

  62. Zhou JY, Schepmoes AA, Zhang X, Moore RJ, Monroe ME, Lee JH, Camp DG, Smith RD, Qian WJ. Improved LC-MS/MS spectral counting statistics by recovering low-scoring spectra matched to confidently identified peptide sequences. J Proteome Res. 2010;9(11):5698–6604.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the grants from the National Key R&D Program of China (2021YFC2101300; No. 2018YFA0900503), the National Natural Science Foundation of China (No. 31970070), and the Youth Interdisciplinary Science and Innovative Research Groups of Shandong University (2020QNQT006). The authors would like to thank Jingyao Qu and Zhifeng Li of the Core Facilities for Life and Environmental Science, State Key laboratory of Microbial Technology of Shandong University for help and guidance in LC-MS/MS and RT-qPCR.

Author information

Authors and Affiliations

Authors

Contributions

LS conceived the work and drafted the manuscript. AY and YW performed the experiments and analyzed the data. YW participated in the experiment and collected the data. HL and YZ designed the work and revised the manuscript. All the authors read and approved the final manuscript.

Corresponding author

Correspondence to Yaohua Zhong.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All the authors have agreed to submit the work to the journal.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

Figure S1. Growth assay of T. reesei QEB4 and the xyr1 overexpression strains. (A) Colonial phenotypes of the parental strain and three recombinant strains on MM with 2% glucose, 2% glycerol, and 2% lactose or on PDA. Plates were incubated at 30℃ and photos were taken at 48 h. Growth rates of hyphae were determined after cultivation on the MM plates with the following carbon sources: glucose (B), glycerol (C), and lactose (D), and on the PDA plates (E) at 30°C for 48 h. Results are means of three biological replicates and error bars indicate ± SD

Additional file 2: Table S1.

Primers used in this study

Additional file 3: Table S2.

The Xyr1 protein identified from the nucleus of T. reesei by LC-MS/MS

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shen, L., Yan, A., Wang, Y. et al. Tailoring the expression of Xyr1 leads to efficient production of lignocellulolytic enzymes in Trichoderma reesei for improved saccharification of corncob residues. Biotechnol Biofuels 15, 142 (2022). https://doi.org/10.1186/s13068-022-02240-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13068-022-02240-9

Keywords