40
1 Modulating ER-Golgi cargo receptors for improving secretion of carrier-fused 1 heterologous protein in the filamentous fungus Aspergillus oryzae 2 3 Running title 4 ER-Golgi cargo receptor and heterologous proteins 5 6 Authors 7 Huy-Dung Hoang, Jun-ichi Maruyama and Katsuhiko Kitamoto* 8 9 Department of Biotechnology, The University of Tokyo, Tokyo, Japan. 10 11 *Corresponding author 12 Mailing address: Department of Biotechnology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo- 13 ku, Tokyo, Japan. 14 Tel: +81-3-5841-5161 15 Fax: +81-3-5841-8033 16 Email: [email protected] 17 AEM Accepts, published online ahead of print on 31 October 2014 Appl. Environ. Microbiol. doi:10.1128/AEM.02133-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved. on March 31, 2019 by guest http://aem.asm.org/ Downloaded from

Downloaded from //aem.asm.org/content/aem/early/2014/10/27/AEM...58 bottlenecks in transcription and post -transcriptional processes (8). Despite the demonstrated 59 efficacy of these

Embed Size (px)

Citation preview

1

Modulating ER-Golgi cargo receptors for improving secretion of carrier-fused 1

heterologous protein in the filamentous fungus Aspergillus oryzae 2

3

Running title 4

ER-Golgi cargo receptor and heterologous proteins 5

6

Authors 7

Huy-Dung Hoang, Jun-ichi Maruyama and Katsuhiko Kitamoto* 8

9

Department of Biotechnology, The University of Tokyo, Tokyo, Japan. 10

11

*Corresponding author 12

Mailing address: Department of Biotechnology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-13

ku, Tokyo, Japan. 14

Tel: +81-3-5841-5161 15

Fax: +81-3-5841-8033 16

Email: [email protected] 17

AEM Accepts, published online ahead of print on 31 October 2014Appl. Environ. Microbiol. doi:10.1128/AEM.02133-14Copyright © 2014, American Society for Microbiology. All Rights Reserved.

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

2

Abstract 18

19

Filamentous fungi are excellent hosts for industrial protein production due to their superior 20

secretory capacity; however, the yield of heterologous eukaryotic proteins is generally lower 21

compared to that of fungal or endogenous proteins. Although activating protein folding 22

machinery in the ER improves the yield, the importance of intracellular transport machinery for 23

heterologous protein secretion is poorly understood. Here, using Aspergillus oryzae as a model 24

filamentous fungus, we studied the involvement of two putative lectin-like cargo receptors, 25

AoVip36 and AoEmp47, in secretion of heterologous protein expressed in fusion with the 26

endogenous enzyme α-amylase as carrier. Fluorescence microscopy revealed that mDsRed-27

tagged AoVip36 localized in the Golgi, whereas AoEmp47 showed localization in both the ER 28

and Golgi. Deletion of AoVip36 and AoEmp47 improved heterologous protein secretion, but 29

only AoVip36 deletion had a negative effect on secretion of α-amylase. Analysis of ER-enriched 30

cell fractions revealed that AoVip36 and AoEmp47 were involved in the retention of 31

heterologous proteins in the ER. However, the overexpression of each cargo receptor had 32

different effects on heterologous protein secretion: AoVip36 enhanced the secretion, whereas 33

AoEmp47 promoted the intracellular retention. Taken together, our data suggest that AoVip36 34

and AoEmp47 hinder the secretion of heterologous proteins by promoting their retention in the 35

ER, but that AoVip36 also promotes the secretion of heterologous proteins. Moreover, we found 36

that genetic deletion of these putative ER-Golgi cargo receptors significantly improves 37

heterologous protein production. The present study is the first to propose that ER-Golgi transport 38

is a bottleneck for heterologous protein production in filamentous fungi. 39

40

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

3

Introduction 41

42

Filamentous fungi possess a prominent secretory capacity and thus are excellent hosts for 43

recombinant protein production. Numerous approaches and attempts have been made to produce 44

industrially valuable proteins in filamentous fungi, such as Aspergillus and Trichoderma (1). 45

However, higher eukaryotic proteins are generally inefficiently produced and secreted in these 46

fungi compared to fungal or endogenous proteins. Several bottlenecks in the heterologous 47

protein production process have been identified to date and a few limiting factors have been 48

overcome by genetically modifying the expression host. In particular, reducing protease activity 49

is necessary to limit the degradation of heterologous proteins, as was demonstrated by the three-50

fold increase in the level of heterologous proteins in the culture supernatant of an Aspergillus 51

oryzae strain with ten successively deleted protease genes (2). Heterologous protein production 52

by A. oryzae was also effectively improved by the repression of vacuolar protein sorting and 53

autophagy (3, 4). The genetic fusion of a target protein with an endogenous protein carrier is a 54

commonly used strategy to increase heterologous protein yields in filamentous fungi. Effective 55

carrier proteins are abundantly secreted enzymes, such as glucoamylase, α-amylase, and 56

cellobiohydrolase (5–7), and are thought to allow the heterologous fusion protein to overcome 57

bottlenecks in transcription and post-transcriptional processes (8). Despite the demonstrated 58

efficacy of these approaches, heterologous protein secretion in recombinant strains remains far 59

below that of endogenous proteins, indicating that other bottlenecks for heterologous protein 60

production and secretion must be overcome before filamentous fungi can reach maximal 61

production capacity. 62

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

4

One approach for increasing the expression of heterologous proteins is the use of a strong 63

promoter; however, increased target protein synthesis may lead to overloading of the folding 64

capacity of the ER. In Aspergilli, expression of human protein tissue plasminogen activator or a 65

fused form of α-amylase-prochymosin triggered the unfolded protein response (UPR) due to ER 66

stress (9, 10). Overexpression of molecular chaperones such as BiP and protein disulfide 67

isomerase to assist protein folding improves the yield of heterologous proteins (11, 12), a 68

response that may be attributable to alleviation of ER stress resulting from increased folding 69

activity. When the UPR is triggered, the expression of a set of UPR-associated proteins, 70

including molecular chaperones, vesicular traffic components, and ER-associated degradation 71

(ERAD) proteins, is up-regulated in an attempt to resolve the ER stress by increasing the folding, 72

transport, and degradation of proteins (13). Constitutive up-regulation of UPR by expression of 73

the activated form of the transcription factor HacA was shown to improve the production of 74

bovine chymosin in Aspergillus niger (14) and the plant taste-modifying protein neoculin in A. 75

oryzae (7). Although these observations suggest that ER stress is a major hindrance for 76

heterologous protein production in filamentous fungi, the underlying cause of the ER stress 77

associated with heterologous protein expression remains unclear. 78

One possible cause of ER stress is the inefficient transport of heterologous proteins between 79

the ER and Golgi. Cargo proteins are transported between the ER and Golgi by vesicular 80

trafficking. Coat protein complex II (COPII)-coated vesicles transport secretory proteins in the 81

anterograde direction, whereas COPI-coated vesicles carry proteins in the retrograde direction, 82

from the Golgi back to the ER (15). During the budding of COPII vesicles, a class of membrane 83

proteins called ER-Golgi cargo receptors is responsible for recruiting cargo proteins into the 84

vesicles, thereby promoting their transport (16). The binding of cargo receptors to target proteins 85

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

5

is highly specific, as demonstrated by the defective secretion of a specific set of proteins upon 86

deletion of a single cargo receptor. For example, the mammalian lectin-type cargo receptor 87

ERGIC-53 selectively exports a subset of glycosylated proteins (17). Previous studies have also 88

found that another lectin-type cargo receptor, VIP36, promotes secretion of the glycoproteins rat 89

parotid gland α-amylase and clusterin (18). VIP36 has also been shown to recycle human α1-90

antitrypsin from the Golgi back to the ER, and upon silencing of VIP36, α1-antitrypsin secretion 91

is increased (19), demonstrating that this cargo receptor has a protein retention function. As 92

VIP36 forms a stable complex with the molecular chaperone BiP (20), it appears to be involved 93

in the quality control of secretory proteins. Because glycoproteins such as α-amylase are 94

commonly used as carrier proteins, lectin-type cargo receptors might affect the secretion of 95

carrier-fused heterologous proteins and are therefore potentially good models for studying the 96

interaction between cargo receptors and heterologous proteins in A. oryzae. This study aimed to 97

investigate the involvement of lectin-type cargo receptors in the intracellular trafficking of 98

carrier-fused heterologous proteins in the filamentous fungus A. oryzae. 99

100

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

6

MATERIALS AND METHODS 101

102

Strains, media and growth conditions. A. oryzae strain RIB40 (21) was used as a DNA 103

donor. Escherichia coli DH5α was used as a host for DNA manipulation. A. oryzae strains 104

NSlD1 (niaD- sC

- ΔligD) and NSPlD1 (niaD

- sC

- ΔpyrG ΔligD) (22) were used as hosts for gene 105

expression and deletion, respectively. A. oryzae strain NS-CaG1 (niaD- sC

- adeA

- AoclxA-106

egfp::adeA) expressing the ER marker calnexin (23) fused to EGFP was used as a host for 107

transformation in the microscopic observation experiments. All strains were maintained on PD 108

agar medium (Nissui Pharmaceutical, Tokyo, Japan). M medium (2% glucose, 0.2% NH4Cl, 109

0.1% (NH4)2SO4, 0.05% KCl, 0.05% NaCl, 0.1% KH2PO4, 0.05% MgSO4·7H2O, and 0.002% 110

FeSO4·7H2O [pH 5.5]) supplemented with 0.15% methionine (M+Met) was used as selective 111

medium for pyrG+ niaD

- sC

- strains used for gene deletion. CD medium (2% glucose, 0.3% 112

NaNO3, 0.2% KCl, 0.1% KH2PO4, 0.05% MgSO4·7H2O, and 0.002% FeSO4·7H2O [pH 5.5]) 113

supplemented with 0.0015% methionine (CD+Met) was used as selective medium for niaD+ sC

- 114

strains and as a growth medium for microscopic observation experiments. 5×DPY medium (10% 115

dextrin, 5% polypeptone, 2.5% yeast extract, 0.5% KH2PO4, and 0.05% MgSO4·7H2O [pH 5.5]) 116

was used as general growth medium unless otherwise stated. 117

118

Generation of a putative cargo receptor-deletion mutant. For generation of a plasmid for 119

the deletion of Aovip36, the 1.5-kb upstream and downstream regions of Aovip36 120

(AO090026000428) were amplified using the primer pairs X-uVIP36-F/X-uVIP36-R and X-121

dVIP36-F/X-dVIP36-R, respectively. The amplified fragments were cloned into the entry vector 122

pDONR-P4-P1R and PDONR-p2R-p3 using the BP reaction (MultiSite Gateway Cloning Kit; 123

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

7

Life Technologies, Carlsbad, CA) to generate the plasmids p5’uVp and p3’dVp, respectively. 124

Using a similar method, the plasmids p5’uEp and p3’dEp, containing the 1.5-kb upstream and 125

downstream regions of Aoemp47 (AO090102000145), respectively, were constructed using the 126

primer sets X-uEp-F/X-uEp-R and X-dEp-F/X-dEp-R, respectively. The two fragments were 127

then connected with upstream and downstream ends of the selective marker pyrG from the 128

plasmid pgEpG using the LR reaction of MultiSite Gateway cloning kit. Linear deletion 129

fragments were then amplified by PCR with the primer sets X-uVIP36-F/X-dVIP36-R for 130

Aovip36 and X-uEp-F/X-dEp-R for Aoemp47. The amplified products were then purified and 131

introduced into A. oryzae strain NSPlD1 using a previously described transformation method 132

(24), and deletion strains were selected on M+Met agar medium. Deletion of cargo receptor 133

genes by the pyrG marker was confirmed by Southern analysis using the upstream region of each 134

gene as probes. The sequences of all primers used in this study are listed in Table S1 in the 135

supplemental material. 136

137

Construction of expression plasmids. For the generation of an expression plasmid for N-138

terminally mDsRed-tagged AoVip36, the signal sequence of AoVip36, mDsRed without a stop 139

codon, and the remaining coding sequence of Aovip36 were amplified using the primer sets 140

attB1-Vp-F/ssVp-mDsRed-R, ssVp-mDsRed-F/mDsRed-Vp-R, and mDsRed-Vp-F/AttB2-Vp-R, 141

respectively. Fusion PCR with the primer pair attB1-Vp-F and attB2-Vp-R was used to insert 142

mDsRed between the signal sequence and the remaining Aovip36 coding sequence. Similarly, the 143

signal sequence of AoEmp47, mDsRed without a stop codon, and the remaining coding sequence 144

of Aoemp47 were amplified using the primer sets attB1-Ep-F/ssEp-mDsRed-R, ssEp-mDsRed-145

F/mDsRed-Ep-R, and mDsRed-Ep-F/AttB2-Ep-R, respectively. Fusion PCR with the primer pair 146

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

8

attB1-Ep-F and attB2-Ep-R was used to insert the mDsRed sequence between the Aoemp47 147

signal sequence and the remaining coding sequence. The fusion products for AoVip36 and 148

AoEmp47 were then cloned into center entry vector pDONR-P221 using the BP clonase reaction 149

to generate pCRVp and pCREp, respectively. The LR clonase reaction with the 5’ entry clone 150

pg5’PaB, containing the amyB promoter, and the 3’ entry clone pg3’TaN, containing the amyB 151

terminator and niaD marker, was then performed to generate a plasmid expressing mDsRed 152

fused versions of AoVip36 (pEpARVpN) and AoEmp47 (pEpAREpN) under control the of 153

amyB promoter. 154

To express heterologous proteins, plasmids for expressing AmyB-fused prochymosin 155

(proCHY) (pgAKCN) (25) or EGFP (pNamyBEGFP) (23) were used for transformation. 156

Integration of a single copy of the plasmids into the niaD locus was confirmed by Southern 157

analysis. Two to three transformants were selected and expression of heterologous protein was 158

confirmed by enzyme activity (proCHY) or immunoblot (AmyB-EGFP). There were no 159

differences in the secretion levels, one transformants was selected for further analysis. 160

For overexpressing mDsRed-AoVip36 and mDsRed-AoEmp47, LR clonase reactions of 161

pCRVp or pCREp with pg5’PaB and pg3’TaSO, containing the amyB terminator and A. oryzae 162

sC marker, were performed to generate an overexpression plasmid. The plasmids were 163

transformed into the strain SlD-aG2 expressing AmyB-EGFP to ensure similar levels of AmyB-164

EGFP expression in the resulted transformants. Expression of mDsRed-tagged cargo receptors in 165

the transformants were confirmed by fluorescence microscopy. There were no differences in the 166

fluorescence pattern of both mDsRed-tagged cargo receptor and AmyB-EGFP between two 167

independent transformants for each construct, and therefore one transformant was selected for 168

further analysis. 169

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

9

170

Construction of a strain endogenously expressing AoGrh1-EGFP. For inserting EGFP at 171

the C-terminus of AoGrh1 (AO090001000523), a 1.5-kb region containing the 0.5-kb 5’ 172

upstream region and entire open reading frame (ORF) of Aogrh1 without a stop codon was PCR 173

amplified with the primers aB4-uGr1-F and aB1r-uGr1-R, and was then cloned into a 5’ entry 174

vector to generate the plasmid p5’uGr1. A 1.5-kb downstream region of Aogrh1 was amplified 175

by PCR with primers aB2r-dGr1-F and aB3-dGr1-R and then cloned into a 3’ entry vector to 176

generate the plasmid p3’dGr1. The two entry clones were subjected to the LR clonase reaction 177

with the center entry clone pgEGFPadeA, which contained the egfp gene and amyB terminator, 178

and destination vector pDEST R4-R3, generating the plasmid pgGr1GA. The insertion cassette 179

for expressing the AoGrh1-EGFP fusion protein from the Aogrh1 gene locus was amplified by 180

PCR using primers aB4-uGr1-F and aB3-dGr1-R from the plasmid pgGr1GA, and was then 181

introduced into strain NSR-ΔlD-2. Insertion of the egfp gene at the 3’ end of the Aogrh1 ORF 182

was confirmed by colony PCR with the primer pair aB4-uGr1-F and aB3-dGr1-R (data not 183

shown). 184

185

Milk-clotting assay for quantification of secreted chymosin. Milk-clotting assay for 186

chymosin activity was performed according to a previously reported method (3). Briefly, 100 µl 187

of sample was mixed with 900 μl of a skim milk solution (12% skim milk in 10 mM CaCl2) in a 188

2-ml eppendorf tube, which was then incubated at 30°C with constant shaking at 50 rpm. The 189

clotting point was defined at the time the solution stopped visibly moving due to formation of a 190

milk clot. The clotting times were recorded and compared with a standard solution prepared with 191

rennin (Sigma-Aldrich, St. Louis, MO) to determine the chymosin concentration in the test 192

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

10

samples. The total amount of proteins in the culture supernatant was measured with a Protein 193

Assay kit (Bio-Rad, Hercules, CA) according to the manufacturer’s instructions. 194

195

Fluorescence microscopy observation. For fluorescence microscopy, 105 conidia were 196

inoculated into 100 μl M+Met liquid medium in 35-mm glass-based dishes (Asahi Techno Glass, 197

Chiba, Japan) and incubated for 18 h at 30°C. Hyphae were then observed with an IX71 inverted 198

microscope (Olympus, Tokyo, Japan) equipped with a 100×neofluor objective lens (1.4 199

numerical aperture). GFP and mDsRed excitations were performed with 488 nm (Furukawa 200

Electric, Tokyo, Japan) and 561 nm (Melles Griot, Carlsbad, CA) semiconductor lasers, 201

respectively, and emitted light was filtered with GFP, DsRed, and DualView filters (Nippon 202

Roper, Chiba, Japan). The CSU22 confocal scanning system (Yokogawa Electronic, Tokyo, 203

Japan) equipped with an Andor iXon cooled digital charge-coupled device camera (Andor 204

Technology PLC, Belfast, UK) and Andor iQ Software (Andor Technology) was used for 205

acquiring and analyzing images. 206

For quantifying colocalization, mean fraction of mDsRed-tagged cargo receptors overlapping 207

the ER or Golgi in the three independent images was calculated using Mander’s coefficient (MC) 208

(26) and the ImageJ (National Institute of Health, Bethesda, MD) plug-in JaCoP. The MC value 209

of 1 means that the two fluorescence signals completely overlap with each other while a MC 210

value of 0 means that there is no overlap between the two fluorescence signals. 211

Localization analysis of AmyB-EGFP in the strains overexpressing mDsRed-tagged AoVip36 212

and AoEmp47 was performed by observing at least 20 independent hyphae. All the hyphae 213

showed similar localization patterns of AmyB-EGFP and mDsRed-tagged cargo receptors, and 214

the representative images were captured. 215

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

11

216

SDS-PAGE and immunoblotting. For SDS-PAGE, protein samples were mixed with 1/5 217

volume 5×Laemli Sample buffer and were then boiled in a water bath for 5 min and immediately 218

placed on ice for 5 min. Samples and molecular markers were separated on a 10% 219

polyacrylamide gel. For immunoblotting, proteins in the gel were transferred onto Immobilon-P 220

PVDF membranes (Millipore, Bedford, MA) using a semi-dry blotting system (Nihon Eido, 221

Tokyo, Japan). For detecting chymosin, a polyclonal rabbit anti-chymosin antibody (Nordic 222

Immunological Laboratories, Tilburg, Netherlands) was used. For detecting EGFP, Living 223

Colors® A.V. Monoclonal Antibody (Clontech, Palo Alto, CA) was used. Chemiluminescence 224

was detected using an ECL Advance Western blotting Detection Kit (GE Healthcare, 225

Buckinghamshire, UK) and images were obtained with an LAS-4000 Mini System (GE 226

Healthcare). Quantification of immunoblotting analysis was performed using ImageJ software 227

(National Institute of Health, Bethesda, MD). The band intensities in the same gel were 228

compared between the control and deletion/overexpression strains, and the relative values were 229

calculated by normalizing that of the control strain as 100%. 230

231

Cell disruption, ER enrichment, and immunoblotting. Conidia were inoculated into 20 ml 232

5×DPY liquid medium and cultivated at 30°C with shaking at 150 rpm. Mycelia were collected 233

and disrupted using metal beads in a Multi-Bead Shocker (Yasui Kikai, Osaka, Japan). Disrupted 234

mycelia were homogenized in extraction buffer (250 mM sucrose, 50 mM Tris, and 1 mM 235

PMSF) supplemented with 1% Protease Inhibitor Cocktail (Sigma) and briefly centrifuged at 236

500×g for 5 min to remove unbroken cells and cell debris. For enrichment of ER contents, the 237

obtained supernatant was further centrifuged at 20,000×g for 30 min, and both the resulting 238

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

12

pellet and supernatant were collected for subsequent analysis. Protein concentration of samples 239

was determined using a Protein Assay kit (Bio-Rad). Cell lysate samples containing 2 µg protein 240

were subjected to SDS-PAGE and immunoblotting, as described above. 241

242

α-Amylase activity assay. α-Amylase in the culture supernatant was quantified using an α-243

Amylase Quantification Kit (Kikkoman, Tokyo, Japan), as instructed by the manufacturer. All 244

culture supernatant samples were diluted 1/50 before quantification. 245

246

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

13

RESULTS 247

248

Subcellular localization of putative lectin-type cargo receptors in A. oryzae. A search for 249

homologs of VIP36 in two A. oryzae genome databases, the Comprehensive A. oryzae Genome 250

Database (CAoGD; http://nribf21.nrib.go.jp/CFGD/) and Database of the Genome Analyzed at 251

NITE (DOGAN; http://www.bio.nite.go.jp/dogan/project/view/AO), resulted in the identification 252

of two entries with high sequence similarity to VIP36: AO090026000428 and AO090102000145. 253

The predicted product of the AO090026000428 gene shared high similarity to human VIP36 and 254

was therefore named AoVip36. AoVip36 had a domain organization similar to that of human 255

VIP36, consisting of a signal sequence, lectin-like domain, and a predicted transmembrane 256

region near the C-terminus (Fig. 1A). AoVip36 showed 27% overall identity with human VIP36, 257

with the lectin-like domain showing 34% identity with the corresponding domain of human 258

VIP36. 259

The predicted AO090102000145 gene product had higher amino acid sequence similarity to 260

the Saccharomyces cerevisiae cargo receptor Emp47p than human VIP36, and was therefore 261

named AoEmp47. The domain organization of AoEmp47 was also similar to that of S. cerevisiae 262

Emp47p and consisted of a signal sequence, a lectin-like domain followed by a coiled-coil 263

domain, and a predicted transmembrane region located near the C-terminus (Fig. 1B). The 264

coiled-coil domain of S. cerevisiae Emp47p was reported to promote self-oligomerization, which 265

is essential for the exit from the ER (27). AoEmp47 showed 16% overall identity with S. 266

cerevisiae Emp47p, and the lectin-like domain shared 18% identity to that of S. cerevisiae 267

Emp47p. Moreover, a tyrosine containing motif (YxxΦ) and a dilysine motif were also found at 268

the C-terminus of AoEmp47 (Fig. 1C). Previous studies in S. cerevisiae Emp47p found that the 269

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

14

tyrosine-containing motif is important for Golgi localization and interaction with COPI and 270

COPII components, while the dilysine motif is required for ER recycling (28, 29). 271

To determine the subcellular localization of AoVip36 and AoEmp47, mDsRed-tagged 272

versions of the two putative cargo receptors were expressed in A. oryzae under control of the 273

amyB promoter. In the fusion construct, mDsRed was inserted after the N-terminal signal 274

sequence of the receptor (Fig. S1A in the supplemental material). The ER membrane protein 275

AoClxA fused to EGFP was used as an ER marker (23), and an A. oryzae homolog 276

(AO090001000523) of Aspergillus nidulans GrhA, called AoGrh1, was used as a Golgi marker. 277

GrhA is a homolog of the human Golgi-associated protein GRASP65 and localizes to cis-Golgi 278

in A. nidulans (30). EGFP was inserted at the C-terminal region of the Aogrh1 gene locus, 279

allowing for the resulting AoGrh1-EGFP fusion protein to be expressed under control of the 280

native Aogrh1 promoter. 281

Fluorescent microscopy analysis revealed that mDsRed-AoVip36 localized to punctuate 282

structures labeled by AoGrh1-EGFP, but did not clearly colocalize with the ER marker AoClxA-283

EGFP (Fig. 2A). Quantification analysis of colocalization indicated that a higher fraction of 284

mDsRed-AoVip36 overlapped with the Golgi marker than the ER (Fig. 2C). These results 285

supported that AoVip36 predominantly localizes to the Golgi. This finding is consistent with the 286

previously reported localization of mammalian VIP36 in MDCK (31) and HeLa cells (19). In 287

contrast, mDsRed-AoEmp47 localized to tubular structures labeled by AoClxA-EGFP and to 288

punctuate structures labeled by AoGrh1-EGFP (Fig. 2B). Quantification analysis of 289

colocalization suggested that a high fraction of mDsRed-AoEmp47 overlapped with both the ER 290

and Golgi markers (Fig. 2C). It is noteworthy to mention that when co-expressed with AoClxA-291

EGFP, the localization shifted predominantly to the ER, which might be due to an unintentional 292

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

15

interaction between AoClxA-EGFP and mDsRed-AoEmp47. These data suggested that 293

AoEmp47 might cycle between the ER and Golgi. This pattern slightly differed from that of S. 294

cerevisiae Emp47p, which was shown to localize to Golgi localization (29), which is discussed 295

later (See Discussion). 296

297

Effects of AoVip36 and AoEmp47 deletion on endogenous α-amylase secretion. To 298

investigate the function of AoVip36 and AoEmp47, the genes encoding these putative receptors 299

were deleted (Fig. S1B and C in the supplemental material) and α-amylase activities in the 300

culture supernatant of the deletion strains were measured. The analysis revealed that the deletion 301

of Aovip36 reduced the α-amylase activity by approximately 30% compared with the control 302

strain, whereas the deletion of AoEmp47 did not affect the α-amylase activity (Fig. 3A). Deletion 303

of each putative cargo receptor did not affect the growth and the total amount of proteins in the 304

culture supernatant (Fig. 3B and C). SDS-PAGE/Coomassie staining analysis showed that most 305

of protein band patterns from the culture supernatant did not significantly differ among the 306

control and deletion strains (Fig. 3D). These data suggested that the decrease in α-amylase 307

activity of the ΔAovip36 strain was not caused by the defects in growth, expression of secreted 308

proteins and extracellular protein stability. The α-amylase activity is specifically correlated with 309

a protein band of approximately 50 kDa in SDS-PAGE/Coomassie staining analysis as evidenced 310

by the fact that deletion of all three A. oryzae α-amylase genes (amyA, amyB and amyC) 311

encoding the identical amino acid sequences except for two amino acids led to a complete loss of 312

both the activity and the protein band (32). The intensity of the protein band corresponding to α-313

amylase was reduced in the ΔAovip36 strain (Fig. 3D). Taken together, these results suggested 314

that AoVip36 was involved in the secretion of α-amylase in A. oryzae. 315

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

16

316

Effects of AoVip36 and AoEmp47 deletion on carrier-fused heterologous protein secretion. 317

To determine if deletion of the putative cargo receptors affected carrier-fused heterologous 318

protein secretion in A. oryzae, the α-amylase fused form of bovine prochymosin (AmyB-319

proCHY) was used as a carrier-fused heterologous protein model to investigate secretory protein 320

trafficking in the ΔAovip36 and ΔAoemp47 strains. The AmyB-proCHY fusion protein is thought 321

to be cleaved at a Kex2-like protease cleavage site in the Golgi, and the released prochymosin 322

autocatalytically converts into mature CHY after further cleavage of the prosequence (33). We 323

first evaluated the secretion of chymosin on the second day of cultivation to minimize the effects 324

of protease degradation. Immunoblotting analysis of the culture supernatant of the ΔAovip36 and 325

ΔAoemp47 strains showed that the band corresponding to prochymosin was increased compared 326

to the control (Fig. 4A). Notably, the size of the detected band corresponded to that of 327

prochymosin after cleavage from AmyB, suggesting that the secreted protein had passed through 328

the complete secretory pathway and did not enter the supernatant as a result of an unconventional 329

secretion process or leakage due to cell lysis. The enzymatic activity of chymosin was quantified 330

on the fourth day of cultivation, because the majority of prochymosin is converted into active 331

chymosin by this day (3, 10). An assay for milk clotting activity in the culture supernatant 332

revealed that deletion of AoVip36 or AoEmp47 increased the amount of mature CHY in the 333

culture medium by approximately two folds compared to the control cells (Fig. 4B). 334

To confirm that deletion of the putative cargo receptors improved carrier-fused heterologous 335

protein secretion, we examined the effect of their deletion on α-amylase-fused EGFP (AmyB-336

EGFP). Immunoblotting analysis of the culture supernatant after two days of cultivation 337

demonstrated that a band corresponding to the size of AmyB-EGFP fusion protein 338

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

17

(approximately 73 kDa) was present. Quantitative analysis of the band intensity indicated that a 339

larger amount of AmyB-EGFP was secreted into the culture medium of the ΔAovip36 and 340

ΔAoemp47 strains compared to the control strain, similar to the case of AmyB-proCHY (Fig. 4C). 341

These results indicated that the putative cargo receptors negatively affected carrier-fused 342

heterologous protein secretion. 343

344

Effects of AoVip36 and AoEmp47 deletion on ER retention of carrier-fused heterologous 345

proteins. As AoVip36 and AoEmp47 were found to localize to the ER and Golgi, we reasoned 346

that they might interfere with the normal intracellular trafficking of carrier-fused heterologous 347

proteins. Thus, we examined and compared the retention of fusion proteins in the ER of the wild-348

type, ΔAovip36, and ΔAoemp47 strains. ER-enriched fractions were obtained by centrifuging 349

mycelial extracts at 20,000×g and collecting the pellet. To test the efficiency of this method, the 350

ER-enriched pellet and supernatant isolated from mycelia expressing the ER marker AoClxA-351

EGFP were immunoblotted for GFP. The analysis showed that AoClxA-EGFP was concentrated 352

in the pellet and was only barely detectable in the supernatant (Fig. S2 in the supplemental 353

material). Thus, the 20,000×g pellet was used an ER-enriched fraction in subsequent analyses. 354

ER-enriched fractions were prepared from the wild-type, ΔAovip36, and ΔAoemp47 strains 355

expressing AmyB-proCHY and then analyzed by immunoblotting for CHY. A band 356

corresponding to the predicted size of AmyB-proCHY (approximately 87 kDa) was detected in 357

the ER-enriched fractions of all strains (Fig. 5A, upper blot), indicating that the fusion protein 358

had not been processed by the Kex2-like protease. As the S. cerevisiae homolog of Kex2p 359

resides in the Golgi (34), the detection of intact AmyB-proCHY supported the validity of the ER 360

enrichment process. Quantitative analysis of the immunoblots revealed that the amount of 361

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

18

AmyB-proCHY in the ER-enriched fraction was reduced in the ΔAovip36 and ΔAoemp47 strains 362

compared to that in the wild-type strain (Fig. 5A, lower). 363

Immunoblotting analysis of the ER-enriched fraction of mycelia expressing AmyB-EGFP 364

revealed that a band consistent with the predicted size of the fusion protein (approximately 73 365

kDa) was present in all strains (Fig. 5B, upper blot). However, the amount of AmyB-EGFP in the 366

ER-enriched fraction was decreased in the ΔAovip36 and ΔAoemp47 strains as compared with 367

the wild-type strain (Fig. 5B, lower). These results suggest that AoVip36 and AoEmp47 interfere 368

with secretion of carrier-fused heterologous proteins by increasing the ER retention rate, a 369

conclusion that is consistent with the increased secretion of AmyB-proCHY and AmyB-EGFP in 370

the ΔAovip36 and ΔAoemp47 strains (Fig. 4). 371

372

Secretion of carrier-fused heterologous protein in strains overexpressing AoVip36 and 373

AoEmp47. Fluorescent microscopic observation of the intracellular trafficking of AmyB-EGFP 374

in strains ΔAovip36 and ΔAoemp47 did not detect any differences compared with that in the 375

wild-type strain (data not shown). We speculated that the putative cargo receptors may be 376

expressed at low levels in the wild-type strain, thereby making it difficult to detect differences in 377

the ER retention rate. For this reason, AoVip36 and AoEmp47 were overexpressed in mDsRed-378

tagged forms under control of the amyB promoter in the presence of the inducing substrate 379

dextrin (35), which was added to cultures as a carbon source. The mDsRed-tagged AoVip36 and 380

AoEmp47 expressed under their native promoters complemented the ΔAovip36 and ΔAoemp47 381

strains, respectively, by suppressing the increased CHY production (Fig. S3 in the supplemental 382

material), which indicated that the mDsRed-tagged forms of cargo receptors are functional. 383

AmyB-EGFP was co-expressed in these strains to detect any effects from the overexpression of 384

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

19

the cargo receptors on the intracellular trafficking of heterologous proteins. In the wild-type 385

strain, an intense signal corresponding to AmyB-EGFP was detected at the hyphal tip (Fig. 6A), 386

similar to the localization of the Spitzenkörper, the location where exocytotic secretory 387

components and secretory proteins concentrate in filamentous fungi (23, 36–38). Under 388

overexpression conditions, mDsRed-AoVip36 was detected as punctate structures, whereas the 389

intracellular AmyB-EGFP signal seemed to be decreased and the localization was more 390

prominent at the cell periphery (Fig. 6B). This observation implied that the polarity of secretion 391

may have shifted from the hyphal tip to the cell periphery, likely as a result of increased 392

intracellular trafficking. In contrast to mDsRed-AoVip36, overexpression of mDsRed-AoEmp47 393

significantly increased the intracellular retention of AmyB-EGFP in tubular structures and 394

aggregates, where mDsRed-AoEmp47 was also predominantly localized (Fig. 6C). These 395

contrasting observations between the two putative cargo receptors suggested that overexpression 396

of AoVip36 enhanced, AmyB-EGFP trafficking, whereas overexpression of AoEmp47 promoted 397

the ER retention of AmyB-EGFP. 398

To confirm this speculation, the effect of overexpressing the two putative cargo receptors 399

on AmyB-EGFP secretion was examined. Immunoblotting of the culture supernatant revealed 400

that overexpression of mDsRed-AoVip36 significantly enhanced AmyB-EGFP secretion, 401

whereas overexpression of mDsRed-AoEmp47 impaired the secretion (Fig. 6D). Overexpression 402

of each cargo receptor did not affect the growth (Fig. S4A in the supplemental material), 403

indicating that these secretion effects were not due to the altered growth rate. Based on these 404

findings, we concluded that the overexpression of AoEmp47 promoted the intracellular retention 405

of AmyB-EGFP, whereas overexpressing AoVip36 alleviated the retention and even stimulated 406

the trafficking of AmyB-EGFP. 407

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

20

DISCUSSION 408

409

Although ER-Golgi cargo receptors are important for ER-Golgi trafficking, no reports prior 410

to the present study have examined the relationship between heterologous protein secretion and 411

cargo receptors. This study has provided evidence that two putative lectin-type cargo receptors 412

affect the intracellular trafficking of heterologous proteins in filamentous fungi. By genetically 413

deleting the putative receptors AoVip36 and AoEmp47, we succeeded in increasing the secretion 414

of carrier-fused heterologous proteins in A. oryzae. 415

The mDsRed-tagged form of AoVip36 predominantly localized to the Golgi (Fig. 2A), 416

similar to mammalian VIP36 (19, 31), suggesting that AoVip36 functions as a cargo receptor by 417

cycling between the Golgi and ER. We found that deletion of AoVip36 impaired the secretion of 418

α-amylase (Fig. 3), whereas AoVip36 overexpression increased the secretion of AmyB-EGFP 419

(Fig. 6D). Together, these results indicate that AoVip36 promotes the anterograde transport of 420

both endogenous and the EGFP-fused form of α-amylase, a property that is in agreement with 421

the fact that overexpression of mammalian VIP36 promotes secretion of the secretory 422

glycoprotein clusterin (18). It was also previously shown that mutation of the carbohydrate-423

binding domain of mammalian VIP36 leads to the loss of lectin activity, but does not result in 424

increased clusterin secretion (18). These results, taken together with our present findings, suggest 425

that the promotion of anterograde transport of VIP36 depends on its lectin activity. As the 426

affinity of mammalian VIP36 to high-mannose glycan is reportedly high in the pH range of 6.5 - 427

7.0, which corresponds to the pH of the ER, but is reduced sharply in the acidic conditions of 428

late-Golgi (39), VIP36 likely binds to glycoproteins in the ER and then transports and releases 429

them at the late-Golgi. Because A. oryzae α-amylase is a glycoprotein (40) and is also N-430

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

21

glycosylated when fused to heterologous proteins, it is assumed that the secretion-promoting 431

effect of AoVip36 might depend on its lectin activity. 432

In contrast to the anterograde transport-promoting function of AoVip36, deletion of AoVip36 433

increased heterologous protein secretion in A. oryzae (Fig. 3), providing evidence that this 434

putative cargo receptor has a protein retention function. In mammals, the glycoprotein α1-435

antitrypsin, which was identified as a retention target of VIP36, exhibits increased intracellular 436

transport upon silencing of VIP36 (19). The interacting complex between VIP36 and α1-437

antitrypsin was shown to repeatedly cycle from the Golgi to the ER, and was also found to co-438

immunoprecipitate with the ER molecular chaperone BiP (19). VIP36 also forms a complex with 439

BiP independently of its N-glycan binding activity (20), and overproduction of a lectin-deficient 440

form of VIP36 decreases clusterin secretion (18). These observations suggest that VIP36 also 441

negatively interferes with protein transport in an N-glycan-independent manner. Consistent with 442

this speculation, deletion of AoVip36 and AoEmp47 alleviated the ER retention of the 443

heterologous proteins chymosin and EGFP in A. oryzae (Fig. 5). The fusion proteins AmyB-444

proCHY and AmyB-EGFP induce UPR (10; data not shown), suggesting that they may be 445

abnormally folded. We speculate that misfolded AmyB-heterologous protein fusions might bind 446

to the chaperone BiP and subsequently form a protein complex with AoVip36, which then 447

recycles back to the ER, as demonstrated with the ER retention (Fig. 5). This hypothesis conflicts 448

with the fact that overexpression of AoVip36 leads to increased secretion of AmyB-EGFP (Fig. 449

6D). To explain these apparently conflicting findings, it is possible that the retention effect might 450

be saturated at a certain concentration of overexpressed AoVip36 when BiP molecules are fully 451

engaged. After exceeding this threshold concentration, AoVip36 overexpression would still 452

promote anterograde transport, leading to induction of the intracellular transport of glycoproteins. 453

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

22

The α-amylase activity in the culture supernatant was not significantly changed by 454

overexpression of AoVip36 (Fig. S4B in the supplemental material), which suggests that the 455

overexpression effect is mainly on the secretion of AmyB-EGFP but not endogenous α-amylase. 456

The observed localization of AoEmp47 in the ER and Golgi (Fig. 2B) is consistent with that 457

of S. cerevisiae Emp47p, which cycles between these two organelles (28, 29). When co-458

expressed with AoClxA-EGFP, mDsRed-AoEmp47 shifted the localization to the ER (Fig. 2B). 459

This observation is similar to the shift of AoEmp47 localization to a tubular structure when 460

overexpressed with AmyB-EGFP (Fig. 6C), suggesting that the overexpression and/or co-461

expression of AmyB-EGFP inhibited the ER exit of AoEmp47. Although AoClxA-EGFP 462

expressed from the native gene locus was shown to be functional (23), we hypothesize that 463

expression of the EGFP fusion of AoClxA might give some negative effect such as the ER stress 464

and inhibition of ER exit, leading to the increase in the retention of AoEmp47 in the ER. 465

Deletion of AoEmp47 increased the secretion of heterologous proteins and reduced their ER 466

retention (Figs. 4 and 5), whereas AoEmp47 overexpression led to the intracellular retention and 467

reduced secretion of AmyB-EGFP (Fig. 6C and D), suggesting that AoEmp47 functions to retain 468

heterologous proteins in the ER. These results are in clear contrast to the observations that 469

deletion of yeast Emp47p impairs the secretion of certain glycoproteins (28), and that the 470

mammalian homolog of AoEmp47, ERGIC-53, mediates the ER export of a subset of 471

glycoproteins, including α1-antitrypsin (17). The present study of cargo receptors in A. oryzae is 472

the first to reveal that an Emp47 homolog has a retention effect for the intracellular transport of 473

secretory proteins. Overexpression of AoEmp47 reduced the α-amylase activity in the culture 474

supernatant (Fig. S4B in the supplemental material). This raises the possibility that the secretion 475

of endogenous α-amylase could be inhibited by AoEmp47 overexpression. However, it remains 476

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

23

unknown whether the interaction of AoEmp47 with target proteins involves N-glycan binding via 477

lectin-like domains. This key question regarding the molecular basis of AoEmp47 interaction 478

with target cargo proteins, and how this interaction results in protein retention in the ER, 479

warrants further investigation. 480

A. oryzae possesses an inherently high potential for protein secretion, which is an attractive 481

property for recombinant protein production. However, before A. oryzae can be used as a cost-482

effective protein secretion host, its capacity for heterologous protein production needs to be 483

markedly improved. The present study has provided the first evidence that the transport of 484

proteins from the ER to Golgi represents a bottleneck in the secretory pathway of A. oryzae and 485

selectively hinders heterologous protein transport. Moreover, our findings suggest that modifying 486

the cargo receptor involved in the trafficking of intracellular proteins can improve heterologous 487

protein secretion. Further investigation on the interaction mechanism between cargo receptors 488

and their target proteins might provide useful information for not only the optimization of A. 489

oryzae, but also for other eukaryotic hosts, for protein production and contribute to the general 490

knowledge of intracellular vesicular trafficking. 491

492

493

ACKNOWLEDGEMENTS 494

H-D.H. was financially supported by an Ajinomoto ASEAN Scholarship. This study was also 495

supported by a Grant-in-Aid for Scientific Research (B) to K. Kitamoto from the Ministry of 496

Education, Culture, Sports, Science and Technology, Japan. 497

498

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

24

REFERENCES 499

500

1. Punt PJ, van Biezen N, Conesa A, Albers A, Mangnus J, van den Hondel C. 2002. 501

Filamentous fungi as cell factories for heterologous protein production. Trends Biotechnol. 502

20:200–206. 503

2. Yoon J, Maruyama J, Kitamoto K. 2011. Disruption of ten protease genes in the 504

filamentous fungus Aspergillus oryzae highly improves production of heterologous 505

proteins. Appl. Microbiol. Biotechnol. 89:747–759. 506

3. Yoon J, Aishan T, Maruyama J, Kitamoto K. 2010. Enhanced production and secretion 507

of heterologous proteins by the filamentous fungus Aspergillus oryzae via disruption of 508

vacuolar protein sorting receptor gene Aovps10. Appl. Environ. Microbiol. 76:5718–5727. 509

4. Yoon J, Kikuma T, Maruyama J, Kitamoto K. 2013. Enhanced production of bovine 510

chymosin by autophagy deficiency in the filamentous fungus Aspergillus oryzae. PLoS 511

One 8:e62512. 512

5. Tsuchiya K, Nagashima T, Yamamoto Y, Gomi K, Kitamoto K, Kumagai C, Tamura 513

G. 1994. High level secretion of calf chymosin using a Glucoamylase-prochymosin fusion 514

gene in Aspergillus oryzae. Biosci. Biotechnol. Biochem. 58:895-899. 515

6. Nyyssönen E, Keränen S. 1995. Multiple roles of the cellulase CBHI in enhancing 516

production of fusion antibodies by the filamentous fungus Trichoderma reesei. Curr. 517

Genet. 28:71–79. 518

7. Nakajima K, Asakura T, Maruyama J, Morita Y, Oike H, Shimizu-ibuka A, 519

Sorimachi H, Arai S, Abe K. 2006. Extracellular production of neoculin, a sweet-tasting 520

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

25

heterodimeric protein with taste-modifying activity, by Aspergillus oryzae. Appl. Environ. 521

Microbiol. 72:3716–3723. 522

8. Gouka RJ, Punt PJ. 1997. Glucoamylase gene fusions alleviate limitations for protein 523

production in Aspergillus awamori at the transcriptional and (post) translational level. 524

Appl. Environ. Microbiol. 63:488–497. 525

9. Guillemette T, van Peij NNME, Goosen T, Lanthaler K, Robson GD, van den Hondel 526

C a MJJ, Stam H, Archer DB. 2007. Genomic analysis of the secretion stress response 527

in the enzyme-producing cell factory Aspergillus niger. BMC Genomics 8:158. 528

10. Ohno A, Maruyama J, Nemoto T, Arioka M, Kitamoto K. 2011. A carrier fusion 529

significantly induces unfolded protein response in heterologous protein production by 530

Aspergillus oryzae. Appl. Microbiol. Biotechnol. 92:1197–1206. 531

11. Lombraña M, Moralejo FJ, Pinto R, Martín JF, Lombran M, Martı JF. 2004. 532

Modulation of Aspergillus awamori thaumatin secretion by modification of bipA gene 533

expression. Appl. Environ. Microbiol. 70:5145–5152. 534

12. Moralejo F, Watson AJ, Jeenes DJ, Archer DB, Martin JF. 2001. A defined level of 535

protein disulfide isomerase expression is required for optimal secretion of thaumatin by 536

Aspergillus awamori. Mol. Genet. Genomics 266:246–253. 537

13. Vembar SS, Brodsky JL. 2008. One step at a time: endoplasmic reticulum-associated 538

degradation. Nat. Rev. Mol. Cell Biol. 9:944–957. 539

14. Valkonen M, Ward M, Wang H, Penttila M. 2003. Improvement of foreign protein 540

production in Aspergillus niger var. awamori by constitutive induction of the unfolded-541

protein response. Appl. Environ. Microbiol. 69:6979–6986. 542

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

26

15. Spang A. 2009. On vesicle formation and tethering in the ER-Golgi shuttle. Curr. Opin. 543

Cell Biol. 21:531–536. 544

16. Dancourt J, Barlowe C. 2010. Protein sorting receptors in the early secretory pathway. 545

Annu. Rev. Biochem. 79:777–802. 546

17. Hauri HP, Kappeler F, Andersson H, Appenzeller C. 2000. ERGIC-53 and traffic in 547

the secretory pathway. J. Cell Sci. 113:587–596. 548

18. Hara-Kuge S, Ohkura T, Ideo H, Shimada O, Atsumi S, Yamashita K. 2002. 549

Involvement of VIP36 in intracellular transport and secretion of glycoproteins in polarized 550

Madin-Darby canine kidney (MDCK) cells. J. Biol. Chem. 277:16332–16339. 551

19. Reiterer V, Nyfeler B, Hauri H-P. 2010. Role of the lectin VIP36 in post-ER quality 552

control of human alpha1-antitrypsin. Traffic 11:1044–1055. 553

20. Nawa D, Shimada O, Kawasaki N, Matsumoto N, Yamamoto K. 2007. Stable 554

interaction of the cargo receptor VIP36 with molecular chaperone BiP. Glycobiology 555

17:913–921. 556

21. Machida M, Asai K, Sano M, Tanaka T, Kumagai T, Terai G, Kusumoto K-I, Arima 557

T, Akita O, Kashiwagi Y, Abe K, Gomi K, Horiuchi H, Kitamoto K, Kobayashi T, 558

Takeuchi M, Denning DW, Galagan JE, Nierman WC, Yu J, Archer DB, Bennett 559

JW, Bhatnagar D, Cleveland TE, Fedorova ND, Gotoh O, Horikawa H, Hosoyama A, 560

Ichinomiya M, Igarashi R, Iwashita K, Juvvadi PR, Kato M, Kato Y, Kin T, 561

Kokubun A, Maeda H, Maeyama N, Maruyama J, Nagasaki H, Nakajima T, Oda K, 562

Okada K, Paulsen I, Sakamoto K, Sawano T, Takahashi M, Takase K, Terabayashi 563

Y, Wortman JR, Yamada O, Yamagata Y, Anazawa H, Hata Y, Koide Y, Komori T, 564

Koyama Y, Minetoki T, Suharnan S, Tanaka A, Isono K, Kuhara S, Ogasawara N, 565

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

27

Kikuchi H. 2005. Genome sequencing and analysis of Aspergillus oryzae. Nature 566

438:1157–1161. 567

22. Maruyama J, Kitamoto K. 2008. Multiple gene disruptions by marker recycling with 568

highly efficient gene-targeting background (DeltaligD) in Aspergillus oryzae. Biotechnol. 569

Lett. 30:1811–1817. 570

23. Kimura S, Maruyama J, Watanabe T, Ito Y, Arioka M, Kitamoto K. 2010. In vivo 571

imaging of endoplasmic reticulum and distribution of mutant α-amylase in Aspergillus 572

oryzae. Fungal Genet. Biol. 47:1044–1054. 573

24. Kitamoto K. 2002. Molecular biology of the Koji molds. Adv. Appl. Microbiol. 51:129–574

153. 575

25. Nemoto T, Watanabe T, Mizogami Y, Maruyama J, Kitamoto K. 2009. Isolation of 576

Aspergillus oryzae mutants for heterologous protein production from a double proteinase 577

gene disruptant. Appl. Microbiol. Biotechnol. 82:1105–1114. 578

26. Bolte S, Cordelières FP. 2006. A guided tour into subcellular colocalization analysis in 579

light microscopy. J. Microsc. 224:213–232. 580

27. Sato K, Nakano A. 2003. Oligomerization of a cargo receptor directs protein sorting into 581

COPII-coated transport vesicles. Mol. Biol. Cell 14:3055–3063. 582

28. Sato K, Nakano A. 2002. Emp47p and its close homolog Emp46p have a tyrosine-583

containing endoplasmic reticulum exit signal and function in glycoprotein secretion in 584

Saccharomyces cerevisiae. Mol. Biol. Cell 13:2518–2532. 585

29. Schröder S, Schimmöller F, Singer-Krüger B, Riezman H. 1995. The Golgi-586

localization of yeast Emp47p depends on its di-lysine motif but is not affected by the ret1-587

1 mutation in alpha-COP. J. Cell Biol. 131:895–912. 588

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

28

30. Pantazopoulou A, Peñalva MA. 2009. Organization and dynamics of the Aspergillus 589

nidulans Golgi during apical extension and mitosis. Mol. Biol. Cell 20:4335–4347. 590

31. Füllekrug J, Scheiffele P, Simons K. 1999. VIP36 localisation to the early secretory 591

pathway. J. Cell Sci. 112:2813–2821. 592

32. Nemoto T, Maruyama J, Kitamoto K. 2012. Contribution ratios of amyA, amyB, amyC 593

genes to high-level α-amylase expression in Aspergillus oryzae. Biosci. Biotechnol. 594

Biochem. 76:1477–1483. 595

33. Nemoto T, Maruyama J, Kitamoto K. 2009. Improvement of heterologous protein 596

production in Aspergillus oryzae by RNA interference with α-Amylase genes. Biosci. 597

Biotechnol. Biochem. 73:2370–2373. 598

34. Redding K, Holcomb C, Fuller RS. 1991. Immunolocalization of Kex2 protease 599

identifies a putative late Golgi compartment in the yeast Saccharomyces cerevisiae. J. Cell 600

Biol. 113:527–538. 601

35. Tsuchiya K, Tada S, Gomi K, Kitamoto K, Kumagai C, Jigami Y, Tamura G. 1992. 602

High level expression of the synthetic human lysozyme gene in Aspergillus oryzae. Appl. 603

Microbiol. Biotechnol. 38:109–114. 604

36. Khalaj V, Brookman JL, Robson GD. 2001. A study of the protein secretory pathway of 605

Aspergillus niger using a glucoamylase-GFP fusion protein. Fungal Genet. Biol. 32:55–65. 606

37. Taheri-talesh N, Horio T, Araujo-baza L, Dou X, Espeso EA, Pen MA, Osmani SA, 607

Oakley BR. 2008. The tip growth apparatus of Aspergillus nidulans 19:1439–1449. 608

38. Hayakawa Y, Ishikawa E, Shoji J-Y, Nakano H, Kitamoto K. 2011. Septum-directed 609

secretion in the filamentous fungus Aspergillus oryzae. Mol. Microbiol. 81:40–55. 610

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

29

39. Kamiya Y, Yamaguchi Y, Takahashi N, Arata Y, Kasai K-I, Ihara Y, Matsuo I, Ito Y, 611

Yamamoto K, Kato K. 2005. Sugar-binding properties of VIP36, an intracellular animal 612

lectin operating as a cargo receptor. J. Biol. Chem. 280:37178–37182. 613

40. Minobe S, Nakajima H, Ito N, Funakoshi I, Yamashina I. 1979. Structure of a major 614

oligosaccharide of Taka-amylase A. J. Biochem. 86:1851–1854. 615

616

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

30

TABLE 1 Strain list 617

Strain name Host Genotype References 618

NSlD1 NSPlD-1 niaD- sC- adeA- argB::adeA- ΔligD::argB ΔpyrG::adeA pgEpG[pyrG] Yoon et al., 2011 619

NSR-ΔlD-2 NSAR1 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB Maruyama et al., 2008 620

NS-CaG-1 NSR-ΔlD-2 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB AoclxA-egfp::adeA Kimura et al., 2010 621

NS-Grh1G-1 NSR-ΔlD-2 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB Aogrh1-egfp::adeA This study 622

S-CaGRVp NS-CaG-1 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB AoclxA-egfp::adeA pEpARVpN[PamyB::Aovip36-mdsred::TamyB::niaD] This study 623

S-CaGREp NS-CaG-1 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB AoclxA-egfp::adeA pEpAREpN[PamyB::Aoemp47-mdsred::TamyB::niaD] This study 624

S-G1GRVp NS-Grh1G-1 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB Aogrh1-egfp::adeA pEpARVpN[PamyB::Aovip36-mdsred::TamyB::niaD] This study 625

S-G1GREp NS-Grh1G-1 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB Aogrh1-egfp::adeA pEpAREpN[PamyB::Aoemp47-mdsred::TamyB::niaD] This study 626

NSlDVp-1 NSPlD-1 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA ΔAovip36::pyrG This study 627

NSlDEp-1 NSPlD-1 niaD- sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA ΔAoemp47::pyrG This study 628

SlD-aG-2 NSlD-1 niaD-::pNamyBEGFP[PamyB::amyB-egfp::TagdA::niaD] sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA pgEpG[pyrG] This study 629

SlDVp-aG-2 NSlDVp-1 niaD-::pNamyBEGFP[PamyB::amyB-egfp::TagdA::niaD] sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA ΔAovip36::pyrG This study 630

SlDEp-aG-2 NSlDEp-1 niaD-::pNamyBEGFP[PamyB::amyB-egfp::TagdA::niaD] sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA ΔAoemp47::pyrG This study 631

SlD-AKC NSlD-1 niaD-::pgAKCN[PamyB::amyB-(kex2)-proCHY::TamyB::niaD] sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA pgEpG[pyrG] This study 632

SlDVp-AKC NSlDVp-1 niaD-::pgAKCN[PamyB::amyB-(kex2)-proCHY::TamyB::niaD] sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA pgEpG[pyrG] This study 633

ΔAovip36::pyrG 634

SlDEp-AKC NSlDEp-1 niaD-::pgAKCN[PamyB::amyB-(kex2)-proCHY::TamyB::niaD] sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA pgEpG[pyrG] This study 635

ΔAoemp47::pyrG 636

lD-aGRVp SlD-aG-2 niaD-::pNamyBegfp[PamyB::amyB-egfp::TagdA::niaD] sC- adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA This study 637

pgEpG[pyrG] pNRVp[PamyB::Aovip36-mdsred::TamyB::sC] 638

lD-aGREp SlD-aG-2 niaD-::pNamyBegfp[PamyB::amyB-egfp::TagdA::niaD] sC-adeA- ΔargB::adeA- ΔligD::argB ΔpyrG::adeA This study 639

pgEpG[pyrG] pNRVp[PamyB::Aoemp47-mdsred::TamyB::sC] 640

641

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

31

Figure legends 642

643

FIG 1 Comparison of domain organization between putative cargo receptors in A. oryzae and 644

their known homologs. (A) AoVip36 and Homo sapiens VIP36 (Accession number 645

NP_006807.1). (B) AoEmp47 and S. cerevisiae Emp47p (Accession number CAA60953.1). 646

Signal sequence (SS) was predicted by SignalP 4.0 (http://www.cbs.dtu.dk/services/SignalP/). 647

Coiled-coil domain (CCD) was predicted by COILS (http://embnet.vital-648

it.ch/software/COILS_form.html). Transmembrane region (TM) was predicted by TMHMM 649

server 2.0 (http://www.cbs.dtu.dk/services/TMHMM/). Lectin-like domain was predicted by 650

Search for Conserved Domains in The National Center for Biotechnology Information 651

(http://www.ncbi.nlm.nih.gov/Structure/cdd/wrpsb.cgi). (C) Tyrosine-containing motifs and 652

dilysine motifs of Emp47 and AoEmp47 are boxed and indicated in italic and bold characters, 653

respectively. 654

655

FIG 2 Subcellular localizations of two putative cargo receptors. mDsRed-tagged AoVip36 or 656

AoEmp47 was co-expressed with the EGFP-tagged version of the ER marker AoClxA-EGFP or 657

the Golgi marker AoGrh1-EGFP. Approximately 105 conidia were inoculated in 100 μl CD+Met 658

and incubated for 18 h at 30°C. (A) AoVip36 localized exclusively to the Golgi. Representative 659

fluorescence microscopy images of the apical area of hyphae expressing AoClxA-EGFP or 660

AoGrh1-EGFP with mDsRed-AoVip36. Arrowheads indicate the punctuate structures labeled by 661

both AoGrh1-EGFP and mDsRed-AoVip36 (Bars = 5 μm). (B) AoEmp47 localized to both the 662

ER and Golgi. Representative fluorescence microscopy images of the apical regions of hyphae 663

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

32

expressing AoClxA-EGFP or AoGrh1-EGFP with mDsRed-AoEmp47. Arrowheads indicate 664

punctuate structures labeled by both AoGrh1-EGFP and mDsRed-AoEmp47 (Bars = 5 μm). (C) 665

Graph representing mean Mander’s coefficient quantifying fraction of cargo receptors 666

overlapping with the ER and Golgi (n = 3 biological replicates, error bars = standard errors,). 667

668

FIG 3 Effect of cargo receptor gene deletion on α-amylase secretion. Approximately 105 conidia 669

of the wild-type, ΔAovip36, or ΔAoemp47 strains were cultivated in 20 ml 5×DPY (pH 5.5) at 670

30°C. (A) α-Amylase activities in the culture supernatant on the second day of cultivation were 671

quantified (n = 3 biological replicates, error bars = standard errors, *P<0.05 unpaired t-test). (B) 672

Wet mycelia weight of the control and deletion strains on the second day of cultivation. (C) Total 673

amount of proteins in the culture supernatants of the control and deletion strains on the second 674

day of cultivation. (D) SDS-PAGE/Coomassie staining of 10 µl of culture supernatant on the 675

second day of cultivation. The arrow indicates the band predicted to correspond to α-amylase. 676

677

FIG 4 Deletion of AoVip36 or AoEmp47 increased the secretion of carrier-fused heterologous 678

proteins in A. oryzae. Conidia of the wild-type, ΔAovip36, or ΔAoemp47 strains expressing 679

AmyB-proCHY or AmyB-EGFP were cultivated in 20 ml 5×DPY (pH 5.5) at 30°C. (A) 680

Immunoblotting of 10 μl culture supernatants of 107

conidia expressing AmyB-proCHY on the 681

second day of cultivation using anti-chymosin antibody. The arrow indicates the estimated 682

position of mature chymosin. (B) Milk clotting activity in the culture supernatant of 105 conidia 683

expressing AmyB-proCHY on the fourth day of cultivation (n = 3 biological replicates, error 684

bars = standard errors, *P<0.05 unpaired t-test). (C) Representative immunoblot image of 10 μl 685

culture supernatants of 107

conidia expressing AmyB-EGFP on the second day of cultivation 686

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

33

using anti-GFP antibody. The arrow indicates the estimated position of the AmyB-EGFP fusion 687

protein. The bar graph shows the quantitative analysis of the immunoblot results (n = 3 688

biological replicates, error bars = standard errors, *P<0.05 unpaired t-test). 689

690

FIG 5 Cargo receptor protein deletion alleviated ER retention of carrier-fused heterologous 691

proteins. Approximately 107 conidia of the wild-type, ΔAovip36, or ΔAoemp47 strains 692

expressing AmyB-proCHY or AmyB-EGFP were cultivated in 20 ml 5×DPY (pH 5.5) for 2 days 693

at 30°C. The ER-enriched fractions with 2 μg proteins were immunoblotted with the indicated 694

antibodies. (A) Representative immunoblot image of the ER-enriched fraction of the strain 695

expressing AmyB-proCHY for CHY. The arrow indicates the estimated position of AmyB-696

proCHY. The bar graph shows the quantitative analysis of the immunoblotting results (n = 3 697

biological replicates, error bars = standard errors, *P<0.05 unpaired t-test). (B) Representative 698

immunoblot image of the ER-enriched fraction of the strain expressing AmyB-EGFP for GFP. 699

The arrow indicates the estimated position of fusion AmyB-EGFP. The bar graph shows the 700

quantitative analysis of the immunoblotting results (n = 3 biological replicates, error bars = 701

standard errors, *P<0.05 unpaired t-test). 702

703

FIG 6 Effect of cargo receptor overexpression on the intracellular trafficking of AmyB-EGFP. 704

An N-terminally mDsRed-tagged version of AoVip36 or AoEmp47 was overexpressed under 705

control of the amyB promoter (P-amyB) with AmyB-EGFP. Approximately 105 conidia were 706

inoculated in 100 μl CD+Met and incubated for 24 h at 30°C. (A) Representative fluorescence 707

microscopy image of the apical region of hyphae expressing AmyB-EGFP (bars = 5 μm). (B) 708

Representative fluorescence microscopy images of the apical region of a hyphae overexpressing 709

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

34

mDsRed-AoVip36 with AmyB-EGFP (bars = 5 μm). AoVip36 overexpression shifted the 710

intracellular AmyB-EGFP signal to the cell periphery. (C) Representative fluorescence 711

microscopy images of the apical region of hyphae overexpressing mDsRed-AoEmp47 with 712

AmyB-EGFP. AoEmp47 overexpression promoted intracellular retention of AmyB-EGFP in ER-713

like tubular structures. The arrowhead indicates an abnormal aggregate containing both AmyB-714

EGFP and mDsRed-AoEmp47. (D) Representative immunoblot image of the culture supernatant 715

of the wild-type, mDsRed-AoVip36, or mDsRed-AoEmp47 overexpressing strains for GFP. 716

Overexpression of mDsRed-AoVip36 improved AmyB-EGFP secretion, whereas overexpression 717

of mDsRed-AoEmp47 impaired AmyB-EGFP secretion. The arrow indicates the estimated 718

position of AmyB-EGFP. 719

720

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from

on March 31, 2019 by guest

http://aem.asm

.org/D

ownloaded from