multimerization- and glycosylation-dependent receptor binding of sars-cov-2 spike proteins ·...

32
1 Multimerization- and glycosylation-dependent receptor binding of SARS- 1 CoV-2 spike proteins 2 3 Kim M. Bouwman 1* , Ilhan Tomris 1* , Hannah L. Turner 2 , Roosmarijn van der 4 Woude 1 , Gerlof P. Bosman 1 , Barry Rockx 3 , Sander Herfst 3 , Bart L. Haagmans 3 , 5 Andrew B. Ward 2 , Geert-Jan Boons 1,3,4,5 , and Robert P. de Vries 1# 6 7 1 Department of Chemical Biology & Drug Discovery, Utrecht Institute for 8 Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG 9 Utrecht, The Netherlands 10 2 Department of Integrative Structural and Computational Biology, The 11 Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 12 92037, USA 13 3 Department of Viroscience, Erasmus University Medical Center, 14 Rotterdam, The Netherlands. 15 4 Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, 16 The Netherlands. 17 5 Complex Carbohydrate Research Center, University of Georgia, 315 18 Riverbend Rd, Athens, GA 30602, USA. 19 6 Department of Chemistry, University of Georgia, Athens, GA 30602, 20 USA. 21 * Contributed equally 22 # for correspondence: [email protected] 23 24 25 preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this this version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558 doi: bioRxiv preprint

Upload: others

Post on 18-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    Multimerization- and glycosylation-dependent receptor binding of SARS-1

    CoV-2 spike proteins 2

    3

    Kim M. Bouwman1*, Ilhan Tomris1*, Hannah L. Turner2, Roosmarijn van der 4

    Woude1, Gerlof P. Bosman1, Barry Rockx3, Sander Herfst3, Bart L. Haagmans3, 5

    Andrew B. Ward2, Geert-Jan Boons1,3,4,5, and Robert P. de Vries1# 6

    7

    1 Department of Chemical Biology & Drug Discovery, Utrecht Institute for 8

    Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584CG 9

    Utrecht, The Netherlands 10

    2 Department of Integrative Structural and Computational Biology, The 11

    Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 12

    92037, USA 13

    3 Department of Viroscience, Erasmus University Medical Center, 14

    Rotterdam, The Netherlands. 15

    4 Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, 16

    The Netherlands. 17

    5 Complex Carbohydrate Research Center, University of Georgia, 315 18

    Riverbend Rd, Athens, GA 30602, USA. 19

    6 Department of Chemistry, University of Georgia, Athens, GA 30602, 20

    USA. 21

    * Contributed equally 22

    # for correspondence: [email protected] 23

    24

    25

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 2

    Receptor binding studies using recombinant SARS-CoV proteins have 26

    been hampered due to challenges in approaches creating spike protein 27

    or domains thereof, that recapitulate receptor binding properties of native 28

    viruses. We hypothesized that trimeric RBD proteins would be suitable 29

    candidates to study receptor binding properties of SARS-CoV-1 and -2. 30

    Here we created monomeric and trimeric fluorescent RBD proteins, 31

    derived from adherent HEK293T, as well as in GnTI mutant cells, to 32

    analyze the effect of complex vs high mannose glycosylation on receptor 33

    binding. The results demonstrate that trimeric fully glycosylated proteins 34

    are superior in receptor binding compared to monomeric and immaturely 35

    glycosylated variants. Although differences in binding to commonly used 36

    cell lines were minimal between the different RBD preparations, 37

    substantial differences were observed when respiratory tissues of 38

    experimental animals were stained. The RBD trimers demonstrated 39

    distinct ACE2 expression profiles in bronchiolar ducts and confirmed the 40

    higher binding affinity of SARS-CoV-2 over SARS-CoV-1. Our results 41

    show that fully glycosylated trimeric RBD proteins are attractive to 42

    analyze receptor binding and explore ACE2 expression profiles in tissues. 43

    44

    45

    46

    47

    48

    49

    50

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 3

    KEYWORDS 51

    Coronavirus spike protein, fluorescent reporter protein, multimerization domain, 52

    glycosylation, ACE2, SARS 53

    54

    INTRODUCTION 55

    SARS-CoV-2 has sparked a society changing pandemic, and additional means 56

    to understand this virus will facilitate counter-measures. SARS coronaviruses 57

    carry a single protruding envelope protein, called spike, that is essential for 58

    binding to and subsequent infection of the host cell. The trimeric spike protein 59

    consists of 3 protomers of 140kD each, containing 60 N-linked glycosylation 60

    sites. The spike is composed of an S1 and S2 domain, in which S2 contains 61

    membrane fusion activity. S1 of coronaviruses can be further divided into N-62

    terminal and C-terminal domains (NTD & CTD), both can contain the receptor-63

    binding domain. In SARS-CoV-1 and -2 this domain is located in the CTD and 64

    referred to as the receptor-binding domain (RBD). The RBD binds to 65

    angiotensin-converting enzyme 2 (ACE2) [1-3], which functions as an entry 66

    receptor for SARS-CoV. After binding and internalization, several proteinases 67

    induce the spike protein into its fusogenic form allowing the fusion of the viral 68

    and target membrane. Although this pathway is known, the details that are of 69

    importance for receptor binding and what differentiates SARS-CoV-2 from its 70

    predecessor SARS-CoV-1, are incompletely understood. It has been shown 71

    that the affinity of the SARS-CoV-2 spike to ACE2 is significantly higher 72

    compared to SARS-CoV-1 [2, 4]. However, how this relates to tissue and cell 73

    tropism remains to be determined. 74

    75

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 4

    Several recombinant protein approaches to create coronavirus spike proteins 76

    have been utilized with success for the development of serological assays [5], 77

    elucidating of spike structures, and isolation of neutralizing antibodies [6, 7]. 78

    However, the conformation of the receptor-binding domain of the spike, which 79

    can be in the “up” and “down” configuration [8-12], is highly variable. This 80

    variability is important in receptor binding as only in the “up” conformation can 81

    RBD bind ACE2. Approaches to control the conformation for vaccine purposes 82

    using stabilization mutations appear to keep the RBD conformation in their 83

    down-state [11, 12], making them non preferred proteins to analyze receptor-84

    binding properties. Also, the multimerization status of the RBD is critical to allow 85

    the analysis of ACE2 interactions. While monomeric RBD proteins can be 86

    efficiently made in large quantities [5], and bind ACE2, they are hardly used in 87

    receptor binding assays to cells and tissues. The majority of studies analyzing 88

    RBD protein binding to cells and tissues, utilize Fc-tagged proteins [2]. Fc-tags 89

    on recombinant proteins are extremely convenient for mammalian cell 90

    expression and purification systems, which result in dimeric recombinant spikes 91

    that are biologically functional. However, the coronavirus spike is trimeric and 92

    thus Fc tagged spikes do not fully recapitulate native properties. 93

    94

    We hypothesized that fluorescent trimeric RBD proteins would provide 95

    complementary means to study RBD-receptor interactions. In this study, we 96

    compared monomeric and trimeric SARS-CoV RBD with full-length trimeric 97

    spikes expressed in cells producing proteins with either complex or high 98

    mannose glycosylation. The fusion of sfGFP and mOrange2 at the C-terminus 99

    has previously been shown to increase expression yields, protein stability, and 100

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 5

    afford additional means for fluorescent-based experiments [13], and thus are 101

    attractive to be fused to RBD proteins. The resulting proteins were analyzed for 102

    binding to cell culture cells and paraffin-embedded tissues of various hosts 103

    including susceptible and non-susceptible animals. The results demonstrate 104

    that fully glycosylated trimeric SARS-CoV-2 RBD proteins reveal the 105

    differences in ACE2 expression between cell cultures and tissue sections. 106

    These trimeric RBD proteins bind ACE2 efficiently in a species-dependent 107

    manner and can be used to profile ACE2 tissue expression. Finally, we 108

    observed distinct expression of ACE2 in bronchioles of experimental animal 109

    models. 110

    111

    RESULTS 112

    Generation of fluorescent coronavirus receptor-binding domain spike proteins 113

    To create recombinant fluorescent soluble full-length ectodomains, NTDs and 114

    RBDs, we cloned these open reading frames (ORF) in plasmids with and 115

    without the GCN4 trimerization domain, fused to either sfGFP or mOrange (Fig 116

    1A). The monomeric and trimeric RBDs were efficiently expressed in both HEK 117

    293T as well as GnTI cells (data not shown), with an increased yield up to 2- to 118

    5-fold when fused to a C-terminal sfGFP (Fig 1B). Expression yields of the 119

    mOrange2 fusions were comparable to that of sfGFP fusions (data not shown). 120

    To illustrate the expression yields of SARS spike proteins or domains thereof 121

    we measured the fluorescence in the cell culture supernatant (Fig 1C). The 122

    wild-type full-length ectodomains were difficult to express even with the addition 123

    of sfGFP or mOrange2 fusion (Fig 1B). To increase yields for the full-length 124

    ectodomain we introduced the 2P and additional hexapro mutations [14], and 125

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 6

    analyzed the fluorescence in cell culture supernatants five days post-126

    transfection after incubation at 33 or 37°C. Although we did not observe a large 127

    increase in yields, we were able to purify sufficient protein to compare full-length 128

    ectodomain trimers vs monomeric and trimeric RBD and NTD proteins. 129

    130

    Spike RBD domains in frame with a C-terminal GCN4 and fluorescent reporter 131

    protein display multimeric features on gel and maintain antigenicity 132

    After purification, all RBD proteins were analyzed on gel under non- and 133

    reducing conditions (Fig 2A). Without reducing agent, monomeric RBD proteins 134

    revealed dimeric fractions which could be reduced to a single monomeric form. 135

    The NTD trimers were reduced under non-reducing conditions, thus solely by 136

    SDS. The trimeric RBD variants, on the other hand, revealed dimers and trimers 137

    that could be reduced. Besides, the NTD of prototypical γ-coronavirus IBV-M41 138

    and influenza A virus HA PR8 as control proteins were included. Finally, we 139

    determined the extent of N-glycosylation maturation on purified proteins 140

    expressed in either GnTI or 293T by subjecting the monomeric and trimeric 141

    proteins to PNGaseF and EndoH treatment (Fig S1). Upon PNGaseF 142

    treatment, all N-glycans were trimmed whereas 293T derived proteins were 143

    insensitive to Endo H, as expected. Surprisingly trimeric RBDs derived from 144

    GnTI cells were partially resistant to Endo H treatment, whereas the monomers 145

    derived from GnTI cells where fully deglycosylated using Endo H. 146

    147

    Fluorescent multimeric Spike RBD proteins maintain antigenicity 148

    Next, we examined the antigenicity of the SARS-CoV-1 and -2 proteins using 149

    serum collected from macaques 21 days post-infection with SARS-CoV-2 [15]. 150

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 7

    Both SARS-CoV-2 RBD monomers and trimers derived from 293T cells were 151

    efficiently recognized, indicating proper folding (Fig 2B). As expected, SARS-152

    CoV-1 RBD proteins were poorly recognized, and the negative controls M41 153

    NTD and PR8 HA displayed baseline binding identical to pre-infection serum. 154

    The NTD trimers were likewise not recognized by the serum (not shown), 155

    indicating that the majority of antibodies in naïve animals after infection are 156

    directed against the SARS-CoV RBD [16]. Similar results were obtained using 157

    GnTI-derived proteins, with the RBD trimer being less efficiently recognized by 158

    the macaque serum than its monomeric counterpart. This is in line with recent 159

    observations that insect cell-derived proteins are less well bound by serum 160

    antibodies [5], indicating the importance of mature N-glycans. 161

    162

    RBD fused to GCN4 and sfGFP fold as trimers with three RBD and sfGFP 163

    molecules divided by the trimerization coiled-coil. 164

    To determine whether the fluorescent RBD trimers are indeed structured in a 165

    trimeric manner we subjected these proteins to negative stain single-particle 166

    EM. The EM data revealed that the RBD proteins form stable trimers that 167

    resemble known spike structures (Fig. 2C). Initially, 58,018 individual particles 168

    were picked, placed into a stack, and submitted to reference-free two-169

    dimensional (2C) classification. From the initial 2D classes, particles that did 170

    not resemble RBD were removed, resulting are final particle stacks of 32,152 171

    particles, which were then subject to Relion 2D classification. All resultant 172

    classes demonstrated evident and distinct trimeric RBD, GCN4, and three 173

    sfGFP protein structures that could be identified in the EM images. From the 174

    EM images, we generated a model in which we took the crystal structures of 175

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 8

    sfGFP, the GCN4 trimerization domain (PDB:2O7H), and the SARS-CoV-2 176

    RBD (PDB: 6XM4) to demonstrate the likely structure of our RBD trimer (Fig 177

    2D). 178

    179

    Fluorescent multimeric Spike RBD proteins bind cell lines in an ACE2 180

    dependent manner and similar to the full-length ectodomain. 181

    To determine the biological activity of our RBD proteins we stained A549 and 182

    VERO cells that are reported to support SARS-CoV replication, with the latter 183

    being more susceptible [17]. However, A549 cells were bound by all our RBD 184

    proteins with a slight increase in intensity from monomeric GnTI derived RBD 185

    proteins to trimeric 293T derived RBDs (Fig 3). Trimeric 293T RBD binding was 186

    efficiently blocked using 4μM recombinant ACE2 whereas 400nM ACE2 pre-187

    incubation was not sufficient to prevent binding of fully glycosylated trimeric 188

    RBD proteins to cells completely. SARS-CoV-2 RBD proteins bound slightly 189

    more intensely to A549 cells compared to the same SARS-CoV-1 RBD 190

    proteins. A similar pattern was observed for VERO-E6 cells, however, the fully 191

    glycosylated SARS-CoV-2 RBD trimer bound markedly stronger compared to 192

    the other RBD preparations (Fig S2A). Importantly, the full-length ectodomain 193

    also bound efficiently to A549 cells (Fig S2B). We did not observe any binding 194

    of the trimeric NTD domains to A549 cells (Fig S2B). MDCK cells, derived from 195

    canine kidney, served as negative controls, to which we indeed did not observe 196

    any binding with any of the indicated proteins (Fig S2C). 197

    198

    Fluorescent RBD proteins reveal strong binding to bronchioles in both ferret 199

    and Syrian golden hamster lungs. 200

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 9

    Ferrets are a susceptible animal model for SARS-CoV-2 [18, 19] and closely 201

    related minks are easily infected on farms [20]. Formalin-fixed, paraffin-202

    embedded tissue slides, will most closely resemble the complex membrane 203

    structures to which spike proteins need to bind. First, ACE2 expression was 204

    assessed using an ACE2 antibody which allowed for comparisons with SARS-205

    CoV-RBD protein binding localization. Here, ACE2 expression was observed in 206

    bronchiolar epithelium cells of ferret lungs (Fig 4A), while the antibody failed to 207

    bind mouse lungs. Comparably, the SARS-CoV RBD protein preparations did 208

    not stain mouse lungs. In ferret lung tissues, we observed only minimal binding 209

    of monomeric RBD proteins derived from GnTI cells (Fig 4B). The fully 210

    glycosylated counterparts showed a slight increase in fluorescent intensity, 211

    which was further increased when fully glycosylated trimeric RBDs were 212

    applied. In all cases, SARS-CoV-2 displayed a higher avidity compared to 213

    SARS-CoV-1. A similar trend of binding intensities was observed for 214

    monomeric, trimeric, and different N-glycosylated SARS-CoV-RBD proteins 215

    fused to mOrange2 (Fig S3A). Again specific binding was seen to the epithelium 216

    of terminal bronchioles and, to a much lower extent, to alveoli and endothelium. 217

    The results were confirmed using horseradish peroxidase readout with a 218

    hematoxylin counterstain (Fig S3B), which output is enzyme driven and purely 219

    qualitative, however, we did observe similar differences in staining intensities. 220

    Here, very minimal staining using the SARS-CoV NTD domains was observed 221

    (Fig S3B), which we did not detect using a fluorescent readout (Fig S3C). To 222

    determine if the binding was ACE2 dependent we pre-incubated trimeric RBD 223

    proteins with recombinant ACE2. While 4 μM was sufficient to block binding to 224

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 10

    cell culture cells (Fig 4C), 8μM was needed to prevent all detectable binding to 225

    ferret lung tissue. 226

    227

    To confirm our observations of different binding on tissues, we quantified the 228

    intensities of the ACE2 antibody and SARS-CoV-1 and -2 RBD proteins, except 229

    for the monomeric GnTI derived proteins as these were almost at the 230

    background (Fig 4D). As expected a noteworthy trend was observed of 231

    increasing binding strength from SARS-CoV1 GnTI derived monomers to 232

    SARS-CoV-2 fully glycosylated RBD trimers. Interestingly multimerization 233

    appears to be more important for strong ACE2 interaction to tissue compared 234

    to the glycosylation status. 235

    236

    Finally, we applied our RBD proteins on the lung tissue of Syrian hamsters, 237

    which are currently the preferred small animal model and known to be highly 238

    susceptible to SARS-CoV-1 and -2 [21-23]. Here again, trimeric fully 239

    glycosylated RBD proteins were superior in binding compared to their 240

    monomeric counterparts. Interestingly, SARS-CoV-2 bound with an even 241

    higher intensity to terminal bronchioles compared to SARS-CoV-1. Importantly, 242

    all binding could be inhibited by pre-incubation of the RBD proteins with 243

    recombinant ACE2, indicating that binding intensity differences are likely ACE2 244

    related. 245

    246

    DISCUSSION 247

    In this report, we describe the generation of fluorescent trimeric RBD proteins 248

    derived from SARS-CoV-1 and 2. These proteins can be efficiently expressed 249

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 11

    in adherent cells and purified directly from the cell culture supernatant using the 250

    increased protein yields provided by the introduction of a C-terminal sfGFP or 251

    mOrange fusion. The fluorescent RBD trimers were superior in receptor binding 252

    on tissues compared to their monomeric RBD counterparts. 253

    254

    Mature N-glycosylation on the spike protein appears to be important for the avid 255

    binding of trimeric RBD proteins to ACE2 in different assays. There is a wealth 256

    of information on site-specific N- and O- glycan conformations on spike proteins 257

    [24-26], with some disagreement on the nature of the RBD N-glycans. In our 258

    protein preparations, they appear to be complex as the majority could not be 259

    cleaved by EndoH (Fig S1). Furthermore, the N-glycosylation of the NTD 260

    domain has been shown to affect the conformation of the RBD in full-length 261

    spikes [27], however similar analyses on the RBD N-glycans are lacking. 262

    263

    Currently, most knowledge of ACE2 expression is based on genomic and 264

    transcriptomic data [28]. However, these analyses are limited as it does not 265

    determine expression biochemically on epithelial cells that make contact to the 266

    outside world [29]. Several groups have transfected variants of ACE2 in cells 267

    to analyze transduction by SARS-CoV-pseudo viruses, although informative 268

    these studies do not provide information on the natural expression pattern of 269

    ACE2 in susceptible hosts [30, 31]. A detailed understanding of the difference 270

    between animal species and cell-specific expression of ACE2 at the molecular 271

    level is essential, as this can provide valuable knowledge on potential hosts 272

    that can be susceptible to SARS-like coronaviruses. Here we demonstrate that 273

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 12

    our fluorescent trimeric RBD proteins are complementary to study ACE2 274

    expression and SARS-CoV receptor binding dynamics. 275

    276

    An intriguing observation was the abundant expression of ACE2 in terminal 277

    bronchioles both in the ferret and Syrian hamster lungs, which are used as 278

    experimental animal models. SARS-CoV-RBD protein binding correlated 279

    perfectly with the staining of the ACE2 antibody and a ferret infection study [18]. 280

    In another ferret infection study, however, the infection was observed in nasal 281

    epithelium and many type I pneumocytes and fewer type II pneumocytes in the 282

    lungs, with few bronchial epithelial cells expressing viral antigen [19]. Some 283

    studies have been performed with anti-ACE2 antibodies, but these stainings 284

    appear not to correlate with infection patterns [32, 33]. We observed a similar 285

    trend indicating that several other factors may be important to infect cells and 286

    that infection could, in turn, regulate ACE2 expression. 287

    288

    MATERIAL AND METHODS 289

    Expression and purification of coronavirus spike proteins for binding studies 290

    Recombinant SARS-CoV-1 and -2 envelope proteins and their subunits were 291

    cloned using Gibson assembly from cDNAs encoding codon-optimized open 292

    reading frames of full-length SARS-CoV-1 and -2 spikes (A kind gift of Rogier 293

    Sanders, Amsterdam Medical Centre, The Netherlands). The pCD5 expression 294

    vector as described previously [34], was adapted to clone the SARS-1 295

    (GenBank: MN908947.3) and 2 (GenBank: MN908947.3), ectodomains 296

    (SARS-2 15-1213, SARS-1 15-1195), N-terminal S1 (SARS-2 15-318, SARS-1 297

    15-305, M41 19 to 272 ) and RBDs (SARS-2 319-541, SARS-1 306-527) 298

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 13

    sequences coding for a secretion signal sequence, a GCN4 trimerization 299

    domain (RMKQIEDKIEEIESKQKKIENEIARIKK) followed by a seven amino 300

    acid cleavage recognition sequence (ENLYFQG) of tobacco etch virus (TEV), 301

    a super folder GFP [13], or mOrange2 [35] and the Twin-Strep 302

    (WSHPQFEKGGGSGGGSWSHPQFEK); IBA, Germany). Alongside we 303

    expressed infectious bronchitis virus M41 spike RBD (GenBank: AY851295.1) 304

    [36], ) and influenza A virus PR8 HA (GenBank: NP_040980) [13]. The viral 305

    envelope proteins were purified from cell culture supernatants after expression 306

    in HEK293T or HEK293GnTI(-) cells as described previously [34]. In short, 307

    transfection was performed using the pCD5 expression vectors and 308

    polyethyleneimine I. The transfection mixtures were replaced at 6 h post-309

    transfection by 293 SFM II expression medium (Gibco), supplemented with 310

    sodium bicarbonate (3.7 g/L), Primatone RL-UF (3.0 g/L), glucose (2.0 g/L), 311

    glutaMAX (Gibco), valproic acid (0,4 g/L) and DMSO (1,5%). At 5 to 6 days after 312

    transfection, tissue culture supernatants were collected. 313

    314

    For ACE2 inhibition studies, ACE2 was expressed in a highly identical fashion, 315

    ACE2 (Addgene Plasmid #145171) was cloned into a pCD5 expression vector 316

    with SacI and BamHI restriction enzymes. The adapted pCD5 expression 317

    vector with an N-terminal HA leader (MKTIIALSYIFCLVFA) peptide, ACE2, and 318

    Twin-Strep (WSHPQFEKGGGSGGGSWSHPQFEK); IBA, Germany) was 319

    purified from HEK293T cell culture supernatant. 320

    321

    Determining expression yield 322

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 14

    We measure fluorescence in the cell culture supernatant when applicable using 323

    a polarstar fluorescent reader with excitation and emission wavelengths of 480 324

    nm and 520 nm for sfGFP and 520nm and 550nm for mOrange2, respectively. 325

    Spike protein expression was confirmed by western blotting using a StrepMAB-326

    HRP classic antibody. Proteins are purified using a single-step with strepTactin 327

    sepharose beads in batch format. Purified proteins were pre-treated (were 328

    indicated) with PNGase F or Endo H (New England Biolabs, USA) according to 329

    the manufacturer’s protocol before analysis by Western blotting using Strep-330

    MAb-HRP classic antibody. 331

    332

    Antigen ELISA 333

    Plates were coated with 2 μg/mL spike protein in PBS for 16 hours at 4°C, 334

    followed by blocking by 3% bovine serum albumin (BSA, VWR, 421501J) in 335

    phosphate-buffered saline-Tween 0,1% (PBS-T 0,1%). After the block, the 336

    proteins were detected using a 21-days post-infection macaque serum and 337

    serum of the same monkey before infection. Serum starting dilution was 1:100 338

    and diluted 1:1 for 5 times and incubated at RT for 2 hrs. Next, wells were 339

    treated with goat-α-human HRP secondary antibody for 1 hour at room 340

    temperature. Serum spike binding antibodies were detected using ODP and 341

    measured in a plate reader (Polarstar Omega, BMG Labtech) at 490 nm. 342

    343

    Negative stain electron microscopy structural analysis 344

    SARS-CoV2-RBD-GCN4-sfGFP in 10mM Tris, 150mM NaCl at 4°C was 345

    deposited on 400 mesh copper negative stain grids and stained with 2% uranyl 346

    formate. The grid was imaged on a 200KeV Tecnai F20 electron microscope 347

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 15

    and a 4k x 4k TemCam F416 camera. Micrographs were collected using 348

    Leginon [37] and then uploaded to Appion [38]. Particles were picked using 349

    DoGPicker [39], stacked. 2D processing was undertaken using Relion. Images 350

    showing Trimeric RBD, a GCN4 connector, and three sfGFPs. 351

    352

    Immunofluorescent cell staining 353

    Vero-E6, A549, and MDCK cells grown on coverslips were analyzed by 354

    immunofluorescent staining. Cells were fixed with 4% paraformaldehyde in 355

    PBS for 25 min at RT after which permeabilization was performed using 0,1% 356

    Triton in PBS. Subsequently, the coronavirus spike proteins were applied at 357

    50μg/ml for 1 h at RT. Primary Strep-MAb classic chromeo-488 (IBA) and 358

    secondary Alexa-fluor 488 goat anti-mouse (Invitrogen) were applied 359

    sequentially with PBS washes in between. DAPI (Invitrogen) was used as 360

    nuclear staining. Samples were imaged on a Leica DMi8 confocal microscope 361

    equipped with a 10x HC PL Apo CS2 objective (NA. 0.40). Excitation was 362

    achieved with a Diode 405 or white light for excitation of Alexa488 and 363

    Alexa555, a pulsed white laser (80MHz) was used at 488 nm and 549 nm, and 364

    emissions were obtained in the range of 498-531nm and 594-627 nm 365

    respectively. Laser powers were 10 – 20% with a gain of a maximum of 200. 366

    LAS Application Suite X was used as well as ImageJ for the addition of the 367

    scale bars. 368

    369

    Tissue staining 370

    Sections of formalin-fixed, paraffin-embedded ferret, Syrian hamster, and 371

    mouse lungs were obtained from the Department of Veterinary Pathobiology, 372

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 16

    Faculty of Veterinary Medicine, Utrecht University and the department of 373

    Viroscience, Erasmus University, The Netherlands, respectively. Tissue 374

    sections were rehydrated in a series of alcohol from 100%, 96% to 70%, and 375

    lastly in distilled water. Tissues slides were boiled in citrate buffer pH 6.0 for 10 376

    min at 900 kW in a microwave for antigen retrieval and washed in PBS-T three 377

    times. Endogenous peroxidase activity was blocked with 1% hydrogen peroxide 378

    for 30 min. Tissues were subsequently incubated with 3% BSA in PBS-T 379

    overnight at 4 °C. The next day, the purified viral spike proteins (50μg/ml) were 380

    added to the tissues for 1 h at RT. With rigorous washing steps in between the 381

    proteins were detected with mouse anti-strep-tag- antibodies (IBA) and goat 382

    anti-mouse IgG antibodies (Life Biosciences). Where indicated recombinant 383

    RBD and ACE2 proteins were pre-incubated O/N at 4 °C before application on 384

    lung tissues or cells. 385

    386

    Quantifying and plotting fluorescent image data 387

    Image quantification was performed by measuring the intensity of gray pixels 388

    of the stained ferret and Syrian hamster lung tissue slides with ImageJ version 389

    1.52p. For stained tissues, background correction was performed by 390

    subtracting the average signal intensity of the antibody control from the images 391

    stained with ACE2 antibody, SARS-CoV1, and SARS-CoV2 recombinant 392

    proteins. Regions of interest were set by highlighting the area using the Image 393

    -> Adjust -> Threshold setting. The 8-bit images were checked for saturation by 394

    plotting the distribution of gray values with Analyze -> Histogram. Subsequent 395

    analysis was performed by quantifying the mean ± standard deviation of 396

    alveoli/cells stained with distinct proteins in various images (Analyze -> 397

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 17

    Measure). Plotting means ± standard deviation values were performed in 398

    GraphPad Prism v8.0.1. A 3D Surface plot was generated for several staining 399

    conditions with Analyze -> 3D Surface Plot. 400

    401

    ACKNOWLEDGEMENTS 402

    R.P.dV is a recipient of an ERC Starting Grant from the European Commission 403

    (802780) and a Beijerinck Premium of the Royal Dutch Academy of Sciences. 404

    Electron microscopic imaging work in the Ward lab was supported by Bill and 405

    Melinda Gates Foundation OPP1170236. SH was funded by NIH/NIAID 406

    (contract number HHSN272201400008C). The authors would like to thank 407

    Rogier Sanders and Phillip Brouwer of the Amsterdam Medical Centre for 408

    sharing the SARS-CoV 1 & 2 open reading frames, Andrea Gröne, Darryl 409

    Leydekkers, and Hélène Verheije from the Division of Pathology, Department 410

    Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht 411

    University, for sharing tissues, IBV M41 plasmids and editing of the manuscript. 412

    413

    REFERENCES 414

    415

    1. Chen X, Li R, Pan Z, Qian C, Yang Y, You R, et al. Human monoclonal 416 antibodies block the binding of SARS-CoV-2 spike protein to angiotensin 417 converting enzyme 2 receptor. Cell Mol Immunol. 2020. Epub 2020/04/22. doi: 418 10.1038/s41423-020-0426-7. PubMed PMID: 32313207. 419 2. Wang Q, Zhang Y, Wu L, Niu S, Song C, Zhang Z, et al. Structural and 420 Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell. 2020. 421 Epub 2020/04/11. doi: 10.1016/j.cell.2020.03.045. PubMed PMID: 32275855; 422 PubMed Central PMCID: PMCPMC7144619. 423 3. Letko M, Marzi A, Munster V. Functional assessment of cell entry and 424 receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. 425 Nature Microbiology. 2020;5(4):562-9. doi: 10.1038/s41564-020-0688-y. 426 4. Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. 427 Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. 428 Cell. 2020;181(2):281-92 e6. Epub 2020/03/11. doi: 429

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 18

    10.1016/j.cell.2020.02.058. PubMed PMID: 32155444; PubMed Central 430 PMCID: PMCPMC7102599. 431 5. Amanat F, Stadlbauer D, Strohmeier S, Nguyen THO, Chromikova V, 432 McMahon M, et al. A serological assay to detect SARS-CoV-2 seroconversion 433 in humans. Nature Medicine. 2020. doi: 10.1038/s41591-020-0913-5. 434 6. Yuan M, Wu NC, Zhu X, Lee CD, So RTY, Lv H, et al. A highly 435 conserved cryptic epitope in the receptor-binding domains of SARS-CoV-2 436 and SARS-CoV. Science. 2020. Epub 2020/04/05. doi: 437 10.1126/science.abb7269. PubMed PMID: 32245784; PubMed Central 438 PMCID: PMCPMC7164391. 439 7. Brouwer PJM, Caniels TG, van der Straten K, Snitselaar JL, Aldon Y, 440 Bangaru S, et al. Potent neutralizing antibodies from COVID-19 patients 441 define multiple targets of vulnerability. Science. 2020. Epub 2020/06/17. doi: 442 10.1126/science.abc5902. PubMed PMID: 32540902; PubMed Central 443 PMCID: PMCPMC7299281. 444 8. Xu C, Wang Y, Liu C, Zhang C, Han W, Hong X, et al. Conformational 445 dynamics of SARS-CoV-2 trimeric spike glycoprotein in complex with receptor 446 ACE2 revealed by cryo-EM. bioRxiv. 2020:2020.06.30.177097. doi: 447 10.1101/2020.06.30.177097. 448 9. McCallum M, Walls AC, Corti D, Veesler D. Closing coronavirus spike 449 glycoproteins by structure-guided design. bioRxiv. 2020:2020.06.03.129817. 450 doi: 10.1101/2020.06.03.129817. 451 10. Ke Z, Oton J, Qu K, Cortese M, Zila V, McKeane L, et al. Structures, 452 conformations and distributions of SARS-CoV-2 spike protein trimers on intact 453 virions. bioRxiv. 2020:2020.06.27.174979. doi: 10.1101/2020.06.27.174979. 454 11. Juraszek J, Rutten L, Blokland S, Bouchier P, Voorzaat R, Ritschel T, 455 et al. Stabilizing the Closed SARS-CoV-2 Spike Trimer. bioRxiv. 456 2020:2020.07.10.197814. doi: 10.1101/2020.07.10.197814. 457 12. Hsieh C-L, Goldsmith JA, Schaub JM, DiVenere AM, Kuo H-C, 458 Javanmardi K, et al. Structure-based Design of Prefusion-stabilized SARS-459 CoV-2 Spikes. bioRxiv. 2020:2020.05.30.125484. doi: 460 10.1101/2020.05.30.125484. 461 13. Nemanichvili N, Tomris I, Turner HL, McBride R, Grant OC, van der 462 Woude R, et al. Fluorescent Trimeric Hemagglutinins Reveal Multivalent 463 Receptor Binding Properties. J Mol Biol. 2019;431(4):842-56. Epub 464 2019/01/01. doi: 10.1016/j.jmb.2018.12.014. PubMed PMID: 30597163; 465 PubMed Central PMCID: PMCPMC6397626. 466 14. Hsieh CL, Goldsmith JA, Schaub JM, DiVenere AM, Kuo HC, 467 Javanmardi K, et al. Structure-based design of prefusion-stabilized SARS-468 CoV-2 spikes. Science. 2020. Epub 2020/07/25. doi: 469 10.1126/science.abd0826. PubMed PMID: 32703906. 470 15. Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM, Oude 471 Munnink BB, et al. Comparative pathogenesis of COVID-19, MERS, and 472 SARS in a nonhuman primate model. Science. 2020. Epub 2020/04/19. doi: 473 10.1126/science.abb7314. PubMed PMID: 32303590; PubMed Central 474 PMCID: PMCPMC7164679. 475 16. Yuan M, Liu H, Wu NC, Lee C-CD, Zhu X, Zhao F, et al. Structural 476 basis of a shared antibody response to SARS-CoV-2. Science. 477 2020:eabd2321. doi: 10.1126/science.abd2321. 478

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 19

    17. Harcourt J, Tamin A, Lu X, Kamili S, Sakthivel SK, Murray J, et al. 479 Severe Acute Respiratory Syndrome Coronavirus 2 from Patient with 2019 480 Novel Coronavirus Disease, United States. Emerg Infect Dis. 2020;26(6). 481 Epub 2020/03/12. doi: 10.3201/eid2606.200516. PubMed PMID: 32160149. 482 18. Kim Y-I, Kim S-G, Kim S-M, Kim E-H, Park S-J, Yu K-M, et al. Infection 483 and Rapid Transmission of SARS-CoV-2 in Ferrets. Cell Host & Microbe. 484 2020;27(5):704-9.e2. doi: 10.1016/j.chom.2020.03.023. 485 19. Richard M, Kok A, de Meulder D, Bestebroer TM, Lamers MM, Okba 486 NMA, et al. SARS-CoV-2 is transmitted via contact and via the air between 487 ferrets. Nat Commun. 2020;11(1):3496. Epub 2020/07/10. doi: 488 10.1038/s41467-020-17367-2. PubMed PMID: 32641684; PubMed Central 489 PMCID: PMCPMC7343828. 490 20. Oreshkova N, Molenaar RJ, Vreman S, Harders F, Oude Munnink BB, 491 Hakze-van der Honing RW, et al. SARS-CoV-2 infection in farmed minks, the 492 Netherlands, April and May 2020. Euro Surveill. 2020;25(23). Epub 493 2020/06/20. doi: 10.2807/1560-7917.ES.2020.25.23.2001005. PubMed PMID: 494 32553059. 495 21. Imai M, Iwatsuki-Horimoto K, Hatta M, Loeber S, Halfmann PJ, 496 Nakajima N, et al. Syrian hamsters as a small animal model for SARS-CoV-2 497 infection and countermeasure development. Proceedings of the National 498 Academy of Sciences. 2020;117(28):16587-95. doi: 499 10.1073/pnas.2009799117. 500 22. Roberts A, Vogel L, Guarner J, Hayes N, Murphy B, Zaki S, et al. 501 Severe Acute Respiratory Syndrome Coronavirus Infection of Golden Syrian 502 Hamsters. Journal of Virology. 2005;79(1):503-11. doi: 10.1128/jvi.79.1.503-503 511.2005. 504 23. Sia SF, Yan L-M, Chin AWH, Fung K, Choy K-T, Wong AYL, et al. 505 Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature. 506 2020;583(7818):834-8. doi: 10.1038/s41586-020-2342-5. 507 24. Shajahan A, Supekar NT, Gleinich AS, Azadi P. Deducing the N- and 508 O- glycosylation profile of the spike protein of novel coronavirus SARS-CoV-2. 509 Glycobiology. 2020. Epub 2020/05/05. doi: 10.1093/glycob/cwaa042. PubMed 510 PMID: 32363391; PubMed Central PMCID: PMCPMC7239183. 511 25. Watanabe Y, Allen JD, Wrapp D, McLellan JS, Crispin M. Site-specific 512 glycan analysis of the SARS-CoV-2 spike. Science. 2020;369(6501):330-3. 513 Epub 2020/05/06. doi: 10.1126/science.abb9983. PubMed PMID: 32366695; 514 PubMed Central PMCID: PMCPMC7199903. 515 26. Zhang Y, Zhao W, Mao Y, Chen Y, Wang S, Zhong Y, et al. Site-516 specific N-glycosylation Characterization of Recombinant SARS-CoV-2 Spike 517 Proteins. bioRxiv. 2020:2020.03.28.013276. doi: 10.1101/2020.03.28.013276. 518 27. Henderson R, Edwards RJ, Mansouri K, Janowska K, Stalls V, Kopp 519 M, et al. Glycans on the SARS-CoV-2 Spike Control the Receptor Binding 520 Domain Conformation. bioRxiv. 2020. Epub 2020/07/09. doi: 521 10.1101/2020.06.26.173765. PubMed PMID: 32637959; PubMed Central 522 PMCID: PMCPMC7337389. 523 28. Lukassen S, Chua RL, Trefzer T, Kahn NC, Schneider MA, Muley T, et 524 al. SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in 525 bronchial transient secretory cells. EMBO J. 2020:e105114. Epub 2020/04/05. 526 doi: 10.15252/embj.20105114. PubMed PMID: 32246845. 527

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 20

    29. Hikmet F, Méar L, Uhlén M, Lindskog C. The protein expression profile 528 of ACE2 in human tissues. bioRxiv. 2020:2020.03.31.016048. doi: 529 10.1101/2020.03.31.016048. 530 30. Conceicao C, Thakur N, Human S, Kelly JT, Logan L, Bialy D, et al. 531 The SARS-CoV-2 Spike protein has a broad tropism for mammalian ACE2 532 proteins. bioRxiv. 2020:2020.06.17.156471. doi: 10.1101/2020.06.17.156471. 533 31. Tang Y-D, Li Y-M, Sun J, Zhang H-L, Wang T-Y, Sun M-X, et al. Cell 534 entry of SARS-CoV-2 conferred by angiotensin-converting enzyme 2 (ACE2) 535 of different species. bioRxiv. 2020:2020.06.15.153916. doi: 536 10.1101/2020.06.15.153916. 537 32. Hikmet F, Mear L, Edvinsson A, Micke P, Uhlen M, Lindskog C. The 538 protein expression profile of ACE2 in human tissues. bioRxiv. 539 2020:2020.03.31.016048. doi: 10.1101/2020.03.31.016048. 540 33. Hamming I, Timens W, Bulthuis ML, Lely AT, Navis G, van Goor H. 541 Tissue distribution of ACE2 protein, the functional receptor for SARS 542 coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 543 2004;203(2):631-7. Epub 2004/05/14. doi: 10.1002/path.1570. PubMed PMID: 544 15141377; PubMed Central PMCID: PMCPMC7167720. 545 34. de Vries RP, de Vries E, Bosch BJ, de Groot RJ, Rottier PJ, de Haan 546 CA. The influenza A virus hemagglutinin glycosylation state affects receptor-547 binding specificity. Virology. 2010;403(1):17-25. doi: 548 10.1016/j.virol.2010.03.047. PubMed PMID: 20441997. 549 35. Shaner NC, Lin MZ, McKeown MR, Steinbach PA, Hazelwood KL, 550 Davidson MW, et al. Improving the photostability of bright monomeric orange 551 and red fluorescent proteins. Nat Methods. 2008;5(6):545-51. Epub 552 2008/05/06. doi: 10.1038/nmeth.1209. PubMed PMID: 18454154; PubMed 553 Central PMCID: PMCPMC2853173. 554 36. Bouwman KM, Parsons LM, Berends AJ, de Vries RP, Cipollo JF, 555 Verheije MH. Three Amino Acid Changes in Avian Coronavirus Spike Protein 556 Allow Binding to Kidney Tissue. J Virol. 2020;94(2). Epub 2019/11/07. doi: 557 10.1128/JVI.01363-19. PubMed PMID: 31694947; PubMed Central PMCID: 558 PMCPMC6955270. 559 37. Potter CS, Chu H, Frey B, Green C, Kisseberth N, Madden TJ, et al. 560 Leginon: a system for fully automated acquisition of 1000 electron 561 micrographs a day. Ultramicroscopy. 1999;77(3):153-61. doi: 562 https://doi.org/10.1016/S0304-3991(99)00043-1. 563 38. Lander GC, Stagg SM, Voss NR, Cheng A, Fellmann D, Pulokas J, et 564 al. Appion: An integrated, database-driven pipeline to facilitate EM image 565 processing. Journal of Structural Biology. 2009;166(1):95-102. doi: 566 10.1016/j.jsb.2009.01.002. 567 39. Voss NR, Yoshioka CK, Radermacher M, Potter CS, Carragher B. DoG 568 Picker and TiltPicker: Software tools to facilitate particle selection in single 569 particle electron microscopy. Journal of Structural Biology. 2009;166(2):205-570 13. doi: 10.1016/j.jsb.2009.01.004. 571 572

    573

    FIGURE LEGENDS 574

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 21

    Figure 1. Expression of coronavirus spike proteins using sfGFP and 575

    mOrange2 fusions 576

    (A) Spike expression plasmids with and without GCN4 trimerization 577

    motif fused with either sfGFP or mOrange2 578

    Schematic representation of the used spike expression cassette. The spike 579

    open reading frame is under the control of a CMV-promotor and was cloned in 580

    frame with DNA sequences coding for the CD5 signal peptide. At the C-581

    terminus a GCN4 trimerization domain followed by sfGFP or mOrange2, and a 582

    TEV cleavable Strep-tag II. 583

    (B) Expression analyses of non and sfGFP fused monomeric and 584

    trimeric RBD proteins and the full-length spike 585

    Denatured samples of the cell culture supernatant were subjected to SDS-586

    PAGE and western blot analyzes stained with an anti-Streptag-HRP antibody. 587

    (C) Quantification 588

    sfGFP emission was directly measured in the supernatants. 589

    (D) Full-length spike adaptation to the hexapro variant. 590

    Full-length spike expression vectors containing the wildtype, 2P or Hexapro 591

    were expressed at 33 or 37C and fluorescence was measured in cell culture 592

    supernatant 4 days post transfection. 593

    594

    595

    Figure 2. Molecular analyses 596

    (A) SDS-PAGE analyses of purified RBD proteins 597

    500 ng of purified proteins were loaded on a gel with or without preheating for 598

    30 minutes at 98C in the presence of a reducing agent. 599

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 22

    (B) Antigenic analyses using 21-day post-infection macaque serum 600

    2μg/ml SARS-CoV-RBD proteins were coated on 96-well plates. Proteins were 601

    detected with macaque serum for 2hrs at RT. An anti-human IgG-HRP was 602

    used to detect RBD specific antibodies. Both 293T and GnTI derived proteins 603

    were analyzed. As negative controls, IBV-M41-NTD and HA-PR8D were 604

    included. 605

    (C) Negative stain EM of trimeric RBD fused to sfGFP 606

    Negative-stain 2D class averages of soluble RBD proteins demonstrate that 607

    they are well-folded trimers. The C-terminal helices and fusion proteins are 608

    visible in some class averages. 609

    (D) Structural model 610

    Based on the crystal structure of a SARS-CoV-2 RBD in the up conformation, 611

    with a GNC4 trimerization domain with 3 sfGFP domains added. 612

    613

    Figure 3. Binding of RBD proteins to A549 cells 614

    SARS-CoV-RBD proteins were applied at 50μg/ml onto A549 cells and where 615

    indicated pre-incubated with recombinant ACE2 protein. SARS-CoV proteins 616

    were detected using an anti-Streptag and a goat-anti-mouse antibody 617

    sequentially. sfGFP fused SARS-CoV-RBD proteins were applied from GnTI 618

    monomers to 293T trimers, left to right. Scalebar is 50μm. 619

    620

    Figure 4. Binding of SARS-COV-RBD proteins and ACE2 antibody to 621

    mouse and ferret lung tissue slides 622

    (A) ACE2 antibody, antibody control, and SARS-CoV-1 and -2 on 623

    mouse lung. Scalebar is 100μm. 624

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 23

    (B) SARS-CoV-RBD fluorescent protein localization in ferret lung tissue. 625

    SARS-CoV-RBD proteins were applied at 50μg/ml and detected using an anti-626

    Streptag and goat-anti-mouse antibodies sequentially. DAPI was used as a 627

    nucleic stain. Scalebar is 100μm. 628

    (C) SARS-CoV-RBD trimer produced in HEK 293T cells pre-incubated with 629

    recombinant ACE2 before application on ferret lung tissue slides. 630

    (D) Quantification 631

    The intensity of gray pixels of stained ferret lung tissue slides with ImageJ 632

    version 1.52p. 633

    634

    Figure 5. Binding of SARS-CoV-RBD proteins to Syrian hamster tissue 635

    slides 636

    SARS-COV-RBD proteins (50μg/ml) were applied to Syrian hamster lung tissue 637

    slides and detected using additional anti-Streptag and goat-anti-mouse 638

    antibodies sequentially. Where indicated SARS-CoV-RBD proteins were pre-639

    incubated with recombinant ACE2 protein. 640

    Scalebar is 100μM. 641

    642

    643

    644

    645

    646

    647

    648

    649

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 24

    Figure 1. 650

    651

    652

    653

    654

    655

    656

    657

    658

    659

    660

    661

    662

    663

    664

    665

    666

    667

    668

    669

    670

    671

    672

    673

    674

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 25

    Figure 2. 675

    676

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 26

    Figure 3 677

    678

    679

    680

    681

    682

    683

    684

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 27

    Figure 4. 685

    686

    687

    688

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 28

    689

    Figure 5 690

    691

    692

    693

    694

    695

    696

    697

    698

    699

    700

    701

    702

    703

    704

    705

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 29

    706

    Supplemental Figure 1. 707

    708

    709

    710

    711

    712

    713

    714

    715

    716

    717

    718

    719

    720

    721

    722

    723

    Supplemental figure 1. Glycosylation analyses of the SARS-CoV protein 724

    preparations. 725

    0.5μg of protein was subjected without or with PNGase F or EndoH for 1hr and 726

    subjected to SDS-PAGE and western blot analyzes. 727

    728

    729

    730

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 30

    Supplemental Figure 2. 731

    732

    Supplemental figure 2. Binding of RBD proteins to cell lines 733

    (A) Protein binding of RBD proteins observed on VERO E6 cells. 734

    Proteins were applied 50μg/ml and where indicated pre-incubated with 735

    recombinant ACE2 protein. Spike proteins were detected using anti-strep and 736

    goat-anti-mouse antibodies. Scalebar is 5μm. 737

    (B) Binding of full-length SARS-CoV-2 ectodomain, IBV-M41, antibodies 738

    only, and NTD spike proteins to A549 cells. Proteins were applied 50μg/ml 739

    and detected using anti-strep and goat-anti-mouse antibodies. Scalebar is 740

    50μm. 741

    (C) Non-binding of RBD trimers to MDCK cells. 742

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 31

    743

    744

    Supplemental Figure 3. 745

    746

    Supplemental figure 3. Binding of RBD proteins to tissues 747

    (A) Binding of RBD proteins fused to mOrange2 to ferret lung tissues 748

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558

  • 32

    Proteins were applied 50μg/ml and detected using anti-strep and goat-anti-749

    mouse antibodies. Scalebar is 100μm. 750

    (B) Binding of RBD proteins fused to sfGFP proteins to ferret lung tissues, 751

    using HRP as a readout. 752

    Identical experiment to (A) but using an HRP readout using anti-strep and goat-753

    anti-mouse antibodies. Scalebar is 100μm. 754

    (C) Control staining on ferret lung tissues using HRP as readout. 755

    M41, NTD trimers of SARS-CoV-1 and -2 and antibodies only. Proteins were 756

    applied 50μg/ml and detected using anti-strep and goat-anti-mouse antibodies. 757

    Scalebar is 100μm. 758

    (D) Lack of NTD binding to ferret lung tissue using fluorescence 759

    Proteins were applied 50μg/ml and detected using anti-strep and goat-anti-760

    mouse antibodies. Scalebar is 100μm. 761

    (E) Control stainings to Syrian hamster tissues, antibodies only and M41 762

    Proteins were applied 50μg/ml and where indicated pre-incubated with 763

    recombinant ACE2 protein. Scalebar is 100μm. 764

    765

    preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for thisthis version posted September 4, 2020. ; https://doi.org/10.1101/2020.09.04.282558doi: bioRxiv preprint

    https://doi.org/10.1101/2020.09.04.282558