Increased lethality in Inuenza and SARS-CoV-2 co ...

27
Increased lethality in Inァuenza and SARS-CoV-2 co- infection is prevented by inァuenza immunity but not SARS-CoV-2 immunity Hagit Achdout Israel Institute for Biological Research Einat B. Vitner Israel Institute for Biological Research https://orcid.org/0000-0001-8578-8551 Boaz Politi Israel Institute for Biological Research Sharon Melamed Israel Institute for Biological Research Yfat Yahalom-Ronen Israel Institute for Biological Research Hadas Tamir Israel Institute for Biological Research Noam Erez Israel Institute for Biological Research Roy Avraham Israel Institute for Biological Research Lilach Cherry Israel Institute for Biological Research Eヲ Makdasi Israel Institute for Biological Research Didi Gur Israel Institute for Biological Research Moshe Aftalion Israel Institute for Biological Research Yaron Vagima Israel Institute for Biological Research Nir Paran Israel Institute for Biological Research https://orcid.org/0000-0001-5143-9660 Tomer Israely ( [email protected] ) Israel Institute for Biological Research https://orcid.org/0000-0003-0246-4477

Transcript of Increased lethality in Inuenza and SARS-CoV-2 co ...

Increased lethality in In�uenza and SARS-CoV-2 co-infection is prevented by in�uenza immunity but notSARS-CoV-2 immunityHagit Achdout 

Israel Institute for Biological ResearchEinat B. Vitner 

Israel Institute for Biological Research https://orcid.org/0000-0001-8578-8551Boaz Politi 

Israel Institute for Biological ResearchSharon Melamed 

Israel Institute for Biological ResearchYfat Yahalom-Ronen 

Israel Institute for Biological ResearchHadas Tamir 

Israel Institute for Biological ResearchNoam Erez 

Israel Institute for Biological ResearchRoy Avraham 

Israel Institute for Biological ResearchLilach Cherry 

Israel Institute for Biological ResearchE� Makdasi 

Israel Institute for Biological ResearchDidi Gur 

Israel Institute for Biological ResearchMoshe Aftalion 

Israel Institute for Biological ResearchYaron Vagima 

Israel Institute for Biological ResearchNir Paran 

Israel Institute for Biological Research https://orcid.org/0000-0001-5143-9660Tomer Israely  ( [email protected] )

Israel Institute for Biological Research https://orcid.org/0000-0003-0246-4477

Letter

Keywords: SARS-CoV-2, in�uenza, vaccination, lethality

Posted Date: January 13th, 2021

DOI: https://doi.org/10.21203/rs.3.rs-136702/v1

License: This work is licensed under a Creative Commons Attribution 4.0 International License.  Read Full License

Version of Record: A version of this preprint was published at Nature Communications on October 5th,2021. See the published version at https://doi.org/10.1038/s41467-021-26113-1.

1

Increased lethality in Influenza and SARS-CoV-2 co-infection is prevented by

influenza immunity but not SARS-CoV-2 immunity

-Yfat Yahalom, 1*, Sharon Melamed1*Boaz Politi, 1*Einat. B. Vitner*, 1Hagit Achdout1

Didi ,1, Efi Makdasi1Lilach Cherry, 1, Roy Avraham1Noam Erez ,1Hadas Tamir, 1Ronen2

¶1Tomer Israely ,1Nir Paran, 2, Yaron Vagima2Moshe Aftalion ,2Gur 3

4

Affiliations: 5

Ziona, -Infectious diseases, Israel institute for Biological Research, Nessof Departments16

7410001, Israel 7

Molecular Genetics, Israel Institute for Biological Department of Biochemistry and28

Research, Ness-Ziona, 7410001, Israel. 9

10

*These authors contributed equally to this work 11

¶To whom correspondence should be addressed: [email protected] 12

13

2

Abstract 14

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause for the 15

ongoing COVID-19 pandemic1. The continued spread of SARS-CoV-2 along with the 16

imminent flu season increase the probability of influenza-SARS-CoV-2 dual infection 17

which might result in a severe disease. In this study, we examined the disease outcome of 18

influenza A virus (IAV) and SARS-CoV-2 co-infection in K18-hACE2 mice. Our data 19

indicates that IAV-infected mice are more susceptible to develop severe disease upon co-20

infection with SARS-CoV-2 two days post influenza infection. This co-infection results in 21

severe morbidity and nearly uniform fatality as compared to the non-fatal influenza disease, 22

or the partial fatality of SARS-CoV-2 alone. Co-infection was associated with elevated 23

influenza viral load in respiratory organs. Remarkably, prior immunity to influenza, but 24

not to SARS-CoV-2, prevented the severe disease and mortality. These data provide an 25

experimental support that flu intervention by prior vaccination may be valuable in reducing 26

the risk of sever Flu - SARS-CoV-2 comorbidity, and highlight the importance of 27

vaccination. 28

3

Main 29

COVID-19 pandemic presents with a broad spectrum of severity ranging from 30

asymptomatic presentation to severe pneumonia. Several risk factors for severe COVID-31

19 disease, such age, sex, and obesity were identified2. Whether co-infection with other 32

pathogens may impact disease severity, are yet to be elucidated. IAV infection is one of 33

the leading causes of respiratory infections in the United States resulting in respiratory 34

illness3. Complications involving secondary infections with pathogens, mostly bacteria, 35

significantly exacerbate the risk of severe flu disease4,5. However, while most of the 36

research in the field of secondary infections following IVA involves bacteria, the secondary 37

effect of infection with viruses is a less explored area. 38

To delineate the interplay between IAV and SARS-CoV-2 infections, we employed 39

transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) by the 40

human cytokeratin 18 promoter (K18-hACE2) which represent a susceptible SARS-CoV-41

2 murine model6. Mice were infected with a non-lethal dose of mouse adapted IAV 42

(A/Puerto Rico/8/1934 H1N1 (PR8)) and were subsequently infected with SARS-CoV-2 43

to mimic co-infection. While the terms ‘co-infection’ and ‘superinfection’ are often 44

interchanges, the use of ‘co-infection’ herein after refers to a sequential infection with 2 45

viruses within a very short time, with the second infection occurring prior to elimination 46

of the first virus. 47

First, the outcome of SARS-CoV-2 infection was tested at two days post influenza infection 48

(dpIi), a pre-symptomatic stage of flu. At this stage, mice does not present any 49

manifestations but the viral titer of IAV in the lungs is high7. IAV-infected mice, started 50

losing weight 5 dpIi and exhibited maximal morbidity at 9-10 dpIi (75% of initial body 51

4

weight) (Fig. 1a). At eleven dpIi mice began to gain weight, and returned to their initial 52

body weight by 18 dpIi. Remarkably, mice infected with SARS-CoV-2 two days post IAV-53

infection, exhibited an earlier and increased weight loss compared to IAV infection alone. 54

Moreover, all of the co-infected mice died by 5-7 days post SARS-CoV-2 infection (dpSi), 55

compared to no-death or only 38% lethality of the IAV- and SARS-CoV-2 infected mice, 56

respectively (Fig. 1b p****<0.0001). 57

Next, we tested co-infection with SARS-CoV-2 at five days post IAV infection; the early-58

symptomatic stages. IAV infected and co-infected mice started to lose weight at 6-7 dpIi 59

(Fig. 1c). However, while the IAV infected mice reached maximal weight loss 8-9 dpIi 60

(84% of initial weight), the co-infected mice continued to lose weight until 10 dpIi (73% 61

of initial body weight). Also, the recovery period of the co-infected mice was prolonged 62

compared to IAV infected mice. While IAV infected mice reached 91% of initial body 63

weight at 10 dpIi and returned to their initial body weight by 11 dpIi, the co-infected mice 64

reached 91% of initial body weight only 16 dpIi and returned to baseline weight only 22 65

dpIi (Fig. 1c). Moreover, co-infection of SARS-CoV-2 at 5 dpIi results in 70% lethality 66

rate in comparison to 43% among mice infected with SARS-CoV-2 alone (p=0.08) (Fig. 67

1d). 68

Finally, we tested co-infection when SARS-CoV-2 was administered during late-69

symptomatic stage of flu disease, in which maximal morbidity was detected (8 dpIi, Fig. 70

1e,f). At this time point, IAV is effectively cleared from the lungs7. Interestingly, SARS-71

CoV-2 infection at 8 dpIi had no effect on the body weight of the mice, nor on their survival 72

rate. These results suggest that co-infection per se (infection with SARS-CoV-2, whilst the 73

IAV is still present in the organs) results in a more severe disease. This may result from 74

5

the adaptive immunity to IAV which already takes place at 8 dpIi that contributes in 75

avoiding influenza-disease exacerbations. 76

In the human population, co-infection is more likely to occur during the asymptomatic 77

period, when the patient does not feel sick and is still active. Also, at this stage, as 78

represented by two dpIi, co-infection results in the most severe and fatal disease. Therefore, 79

we chose to focus on this stage, and hereafter co-infection refers to infection with IAV 80

followed by infection with SARS-CoV-2 two days later. 81

To correlate between increased morbidity and mortality observed in co-infected mice and 82

viral load, SARS-CoV-2 PFU and IAV viral load were quantified in the lungs and nasal 83

turbinates (Fig. 2a). A significant increase in IAV viral RNA was observed in the lungs 84

(4.6 -fold increase) and in the nasal turbinates (11-fold increase) of co-infected mice 85

compared to IAV- infected mice (Fig. 2b,c), which coincide with the exacerbated disease. 86

In contrast, the level of SARS-CoV-2 was reduced in the co-infected mice compared to 87

SARS-CoV-2- infected mice both in the lung and in the nasal turbinates (Fig. 2d,e), 88

suggesting a significant role for IAV in the observed severe disease. This finding is in 89

accordance with previous evidence of pathogenic competition between respiratory viruses, 90

such as influenza and seasonal coronaviruses8,9. A possible explanation for the reduced 91

SARS-CoV-2 viral load might be the induction of innate immune response activated by 92

IAV infection prior to the infection with SARS-CoV-2, and inhibiting establishment of 93

infection and replication10,11,12. It is yet unclear what mechanism allows IAV to evade such 94

antiviral immunity. In addition to innate immunity mechanisms underlying the reduced 95

SARS-CoV-2 viral load, IAV infection may also interfere with SARS-CoV-2 infection 96

through super-infection exclusion. It is yet to be determined whether a mechanism similar 97

6

to that previously shown for inhibiting influenza super-infection by neuraminidase (NA) is 98

also applied here13. Notably, though SARS-CoV-2 viral load was reduced in the co-infected 99

mice, the remaining levels were sufficient to trigger the lethal outcome of the co-infection. 100

Taken together, these results suggest that the increased morbidity and mortality detected in 101

the co-infected mice are associated with higher levels of IAV in the respiratory system, 102

rather than that of SARS-CoV-2. 103

To elaborate on the host response to IAV, SARS-CoV-2, and co-infection of IAV and 104

SARS-CoV-2, we assessed the expression of immune-related genes in the lungs at 4 and 2 105

days post IAV and SARS-CoV-2 infection, respectively (Fig. 2a). Overall, in lungs of 106

SARS-CoV-2 infected mice, no alterations in mRNA levels of the tested genes were 107

observed compared to uninfected mice (Fig. 3), most likely due to low infection dose 108

(10pfu/mouse) and short time post infection (2 days). Upon infection with IAV, all of the 109

tested genes were over-expressed (Fig. 3). Remarkably, IAV and SARS-CoV-2 co-110

infection resulted in a significantly higher elevation of gene expression compared to that 111

exhibited upon IAV infection alone, indicating a robust induction of the immune system 112

that may lead to the exacerbated disease. 113

Then, we examined whether pre-existing immunity to SARS-CoV-2 prevents the severe 114

manifestations of co-infected mice. Efficient immunity to SARS-CoV-2 was induced by 115

infection of mice with a non-lethal dose of SARS-CoV-2. This infection induced 116

neutralizing antibodies against SARS-CoV-2 (data not shown), and rescued mice from 117

SARS-CoV-2 challenge (Fig. 4a). However, while pre-existing immunity to SARS-CoV-118

2 completely prevented the lethality caused by SARS-CoV-2 infection, amounting to 33%, 119

it had no effect on the morbidity and lethality caused by IAV and SARS-CoV-2 co-120

7

infection (90% compared to 86% lethality in pre-existing SARS-CoV-2 immunity) (Fig. 121

4a,b). This data further supports the notion that the severe manifestations of the co-122

infection are not the result of SARS-CoV-2 replication. 123

To determine whether pre-existing immunity to IAV can prevent the co-infection 124

manifestations, mice were vaccinated intramuscular (i.m.) with IAV 30 days prior to viral 125

infection with IAV and SARS-CoV-2. Pre-exposure to IAV alleviated morbidity observed 126

upon infection with IAV (Fig. 4d), and had no effect on the survival rate upon SARS-CoV-127

2 infection (50% compared to 67% survival, respectively. Fig. 4c,d, p=0.64). Remarkably, 128

pre-existing immunity to IAV prevented the severe manifestations and fatality caused by 129

IAV and SARS-CoV-2 co-infection. Neither weight loss nor increased lethality were 130

detected in the co-infected mice that were vaccinated to IAV compared to co-infected mice 131

without immune background (Fig. 4 c,d). Altogether, our data suggest that IAV- SARS-132

CoV-2 co-infection results in a severe and lethal disease in susceptible mice. Severe 133

manifestations are associated with robust induction of innate immunity and elevated IAV 134

viral load in the respiratory organs. Importantly, prior immunity to Flu but not to SARS-135

CoV-2 prevented disease and death. Based on these results, we suggest that flu 136

intervention, by prior vaccination, may prove valuable in reducing the risk of severe Flu - 137

SARS-CoV-2 comorbidity. 138

139

8

Methods 140

Cells 141

Vero E6 (ATCC® CRL-1586TM) were obtained from the American Type Culture 142

Collection (Summit Pharmaceuticals International, Japan). Madin-Darby Canine Kidney 143

(MDCK) cells (ATCC® CCL-34™) were kindly provided by Dr. Michal Mandelboim 144

(Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-145

Hashomer, Israel). Cells were maintained in Dulbecco’s Modified Eagle’s Medium 146

(DMEM) supplemented with 10% fetal bovine serum (FBS), MEM non-essential amino 147

acids, 2 nM L-Glutamine, 100 Units/ml Penicillin, 0.1 mg/ml streptomycin and 12.5 148

Units/ml Nystatin (Biological Industries, Israel). Cells were cultured at 37°C, 5% CO2 at 149

95% air atmosphere. 150

Viruses 151

SARS-CoV-2, isolate Human 2019-nCoV ex China strain BavPat1/2020 was kindly 152

provided by Prof. Dr. Christian Drosten (Charité, Berlin) through the European Virus 153

Archive – Global (EVAg Ref-SKU: 026V-03883). Virus stocks were propagated (4 154

passages) and tittered on Vero E6 cells. The virus was stored at -80°C until use. 155

Strain A/Puerto Rico/8/1934 (H1N1) influenza A virus (IAV, PR8) was a kind gift from 156

Dr. Michal Mandelboim. The virus was propagated in embryonated chicken eggs as 157

previously described14. HAU was determined with chicken erythrocytes, and virus titers 158

were determined by a plaque assay on MDCK cell monolayers. The virus was stored at -159

80°C until use. 160

161

162

9

Animal experiments 163

All animal experiments involving SARS-CoV-2 were conducted in a BSL3 facility. 164

Treatment of animals was in accordance with regulations outlined in the U.S. Department 165

of Agriculture (USDA) Animal Welfare Act and the conditions specified in the Guide for 166

Care and Use of Laboratory Animals (National Institute of Health, 2011). Animal studies 167

were approved by the local IIBR ethical committee for animal experiments (protocols 168

number M-29-20, M-40-20 and M-41-20). Female K18-hACE2 transgenic mice (The 169

Jackson Laboratory) 6-8 weeks old were maintained at 20−22°C and a relative humidity of 170

50 ± 10% on a 12hrs light/dark cycle. Animals were fed with commercial rodent chow 171

(Koffolk Inc.) and provided with tap water ad libitum. Prior to infection, mice were kept in 172

groups of 10. Mice were randomly assigned to experimental groups. 173

For infection, the viruses were diluted in phosphate buffered saline (PBS) supplemented 174

with 2% FBS (Biological Industries, Israel). Anesthetized animals (Ketamine 75 mg/kg, 175

Xylazine 7.5 mg/kg in PBS) were infected by 20µl intranasal (i.n.) instillation (PR8 176

80pfu/mouse, SARS-CoV-2 10pfu/mouse). 177

Animal’s immunization: for SARS-CoV-2 immunization, i.n. instillation of 2pfu/mouse 178

SARS-CoV-2 was performed. For IAV immunization, mice were vaccinated 179

intramuscularly (i.m.) with 106pfu/mouse. Immunized mice were infected 30 days post 180

immunization. 181

Determination of viral load in organs 182

Viral loads were determined at 2 days post SARS-CoV-2 infection, or 4 days post influenza 183

infection. In the co-infected group, viral load was determined 4 days post IAV infection, 184

which is 2 days post SARS-CoV-2 infection. Each group of mice included 10 mice. K18-185

10

hACE2 mice were sacrificed, and lungs and nasal turbinates (n.t.) were harvested and 186

stored in -80°C until further processing. Organs were processed for titration in 1.5 mL of 187

ice-cold PBS as previously described15. Part of the processed tissue samples was used 188

immediately for RNA extraction for IAV viral RNA determination and for gene expression, 189

while the other part was kept in -80°C until further processing for viral titration (used for 190

SARS-CoV-2 pfu). 191

SARS-CoV-2 viral load was determined using pfu assay16. Briefly, serial dilutions of 192

extracted organs from mice infected with SARS-CoV-2 or co-infected with IAV and 193

SARS-CoV-2, were prepared in MEM containing 2% FCS, and used to infect Vero E6 194

monolayers in duplicates (200µl/well). Plates were incubated for 1 hour at 37ºC to allow 195

viral adsorption. Then, 2ml/well of overlay (MEM containing 2% FBS and 0.4% tragacanth 196

(Merck, Israel)) was added to each well and plates were incubated at 37°C, 5% CO2 for 48 197

hours. The media was then aspirated and the cells were fixed and stained with 1ml/well of 198

crystal violet solution (Biological Industries, Israel). The number of plaques in each well 199

was determined, and SARS-CoV-2 PFU (plaque forming unit) titer was calculated. 200

IAV viral RNA was determined using Real-time RT-PCR (see below). 201

Quantitative Real-Time RT-PCR 202

RNA was extracted by Viral RNA mini kit (Qiagen, Germany) as per manufacturesr’s 203

instructions. IAV viral RNA load in the lung and nasal turbinates (n.t.) were determined by qRT-204

PCR. Real-time RT-PCR was conducted with SensiFAST™ Probe Lo-ROX One-Step Kit (Bioline, 205

78005) and analyzed with the 7500 Real Time PCR System (Applied Biosystems). The PFU 206

Equivalent per organ (pfuE/organ) were calculated from standard curve generated from virus 207

stocks. qPCR primers and probes for the detection of PR8: PR8-PA-FW: 208

11

CGGTCCAAATTCCTGCTGA; PR8-PA-RW:CATTGGGTTCCTTCCATCCA; PR8-PA-Probe: 209

CCAAGTCATGAAGGAGAGGGAATACCGCT 210

Total RNA extracted from lungs of mice infected with IAV or SARS-CoV-2, or co-211

infected, at 2 dpSi and 4 dpIi was used to measure differently-expressed genes by qRT-212

PCR using the corresponding specific primers printed on 96 well plates (Custom TaqMan 213

Array Plates, Applied BiosystemsTM) as previously described15. Briefly, 1 microgram 214

cDNA was synthesized out of the RNA using Verso cDNA Synthesis Kit (Thermo Fisher 215

Scientific, Waltham, MA, USA) according to the manufacturer’s instructions. Samples 216

were subjected to qPCR with TaqMan® Fast Advanced Master Mix (7500 Real Time PCR 217

System, Applied Biosystems, Thermo Fisher Scientific). The housekeeping gene gapdh 218

was used to normalize fold change of each gene as compared to mock-infected control at 219

the same time point and was calculated as ∆∆CT. 220

221

12

Acknowledgment 222

The authors would like to thank Prof. Dr. Christian at the Charité Universitätsmedizin, 223

Institute of Virology, Berlin, Germany for providing the SARS-CoV-2 BavPat1/2020 224

strain. We thank Dr. Shai Weiss for safety advisory. We thank Dr. Michal Mandelboim 225

for MDCK cells and Influenza virus A/Puerto Rico/8/34 H1N1 (PR8). 226

227

Author contributions 228

H.A, E.B.V, B.P, S.M, T.I designed the research; H.A, E.B.V, B.P, S.M, R.A, H.T, 229

Y.Y.R, N.E, L.C.M, Y.V, D.G, E.M, N.P, and T.I, performed the experiments. H.A, 230

E.B.V. N.P, T.I wrote the manuscript. All authors discussed results and commented on 231

the manuscript before submission. R.A. is supported by the Israel Science Foundation 232

(grant 521/18). E.B.V is supported by the Katzir Foundation. 233

234

Competing interests 235

The author declare no conflict of interest. 236

237

13

References 238

1. https://www.who.int/emergencies/diseases/novel-coronavirus-2019. World Health 239

Organization Coronavirus Disease (COVID-19) pandemic. (2020). 240

2. Regina, J., et al. Epidemiology, risk factors and clinical course of SARS-CoV-2 241

infected patients in a Swiss university hospital: An observational retrospective 242

study. PLOS ONE 15, e0240781 (2020). 243

3. https://www.cdc.gov/flu/about/burden/index.html. Centers for Disease Control 244

and Prevention. Influenza (Flu). (2020). 245

4. McCullers, J.A. The co-pathogenesis of influenza viruses with bacteria in the 246

lung. Nature Reviews Microbiology 12, 252-262 (2014). 247

5. Falsey, A.R., et al. Bacterial complications of respiratory tract viral illness: a 248

comprehensive evaluation. The Journal of infectious diseases 208, 432-441 249

(2013). 250

6. Oladunni, F.S., et al. Lethality of SARS-CoV-2 infection in K18 human 251

angiotensin-converting enzyme 2 transgenic mice. Nature Communications 11, 252

6122 (2020). 253

7. Blazejewska, P., et al. Pathogenicity of different PR8 influenza A virus variants in 254

mice is determined by both viral and host factors. Virology 412, 36-45 (2011). 255

8. Nickbakhsh, S., et al. Virus–virus interactions impact the population dynamics of 256

influenza and the common cold. Proceedings of the National Academy of Sciences 257

116, 27142-27150 (2019). 258

9. Wolff, G.G. Influenza vaccination and respiratory virus interference among 259

Department of Defense personnel during the 2017–2018 influenza season. 260

Vaccine 38, 350-354 (2020). 261

10. Rigby, R.E., Wise, H.M., Smith, N., Digard, P. & Rehwinkel, J. PA-X 262

antagonises MAVS-dependent accumulation of early type I interferon messenger 263

RNAs during influenza A virus infection. Scientific Reports 9, 7216 (2019). 264

11. Felgenhauer, U., et al. Inhibition of SARS-CoV-2 by type I and type III 265

interferons. The Journal of biological chemistry 295, 13958-13964 (2020). 266

14

12. McAfee, M.S., Huynh, T.P., Johnson, J.L., Jacobs, B.L. & Blattman, J.N. 267

Interaction between unrelated viruses during in vivo co-infection to limit 268

pathology and immunity. Virology 484, 153-162 (2015). 269

13. Huang, I.-C., et al. Influenza A Virus Neuraminidase Limits Viral Superinfection. 270

Journal of Virology 82, 4834-4843 (2008). 271

14. Vagima, Y., et al. Influenza virus infection augments susceptibility to respiratory 272

Yersinia pestis exposure and impacts the efficacy of antiplague antibiotic 273

treatments. Scientific Reports 10, 19116 (2020). 274

15. Israely, T., et al. Differential Response Following Infection of Mouse CNS with 275

Virulent and Attenuated Vaccinia Virus Strains. Vaccines 7(2019). 276

16. Yahalom-Ronen, Y., et al. A single dose of recombinant VSV-∆G-spike vaccine 277

provides protection against SARS-CoV-2 challenge. Nature Communications 11, 278

6402 (2020). 279

280

281

15

0 2 4 6 8 10 12 14 16 18 20 22 24

70

80

90

100

110

120

% o

f in

itia

l boody w

eig

ht

***

Day of infection

IAV

0 1 2

0 5 10 15 20

0

20

40

60

80

100

Perc

ent surv

ival

****

****

0 5 10 15 20 25

70

80

90

100

110

120

% o

f in

itia

l boody w

eig

ht

***

0 5

Day of infection

0 5 10 15 20

0

20

40

60

80

100

Perc

ent surv

ival

ns

**

0 4 8 12 16 20 24

70

80

90

100

110

120

% o

f in

itia

l boody w

eig

ht

0 8

Day of infection

0 5 10 15 20

0

20

40

60

80

100

Perc

ent surv

ival

*

ns

Days post IAV infection Days post SARS-CoV-2 infection

SARS-CoV-2IAV IAV & SARS-CoV-2

a

c

e

b

d

f

1

282

Fig. 1: Morbidity and mortality in K18-hACE2 mice infected with SARS-CoV-2 following 283

influenza infection 284

K18-hACE2 mice were infected with IAV (80 pfu/mice, i.n), followed by infection with SARS-285

CoV-2 (10 pfu/mice, i.n) at two (a,b), five (c,d) or eight (e,f) dpIi. Percent body weight loss 286

following infection (a,c,e). Red arrow represents IAV infection. Blue arrow represents SARS-CoV-287

2 infection. Error bars represent standard errors (SE). p.values are indicated in the figure as 288

asterisks, and were calculated by Student’s t-test using GraphPad Prism 8.4.3. ***p<0.0002 at 4-7 289

dpIi (a), and at 9-16 dpIi (c) IAV infection compared to co-infection. Survival curves (b,d,f): 290

16

*p=0.04; **p=0.0026; ****p<0.0001 IAV or SARS-CoV-2 infection compared to co-infected. ns, 291

not significant, Log-rank (Mantel-Cox). Figure shows one representative experiment out of 4 (a,b), 292

2 (c,d), 1 (e,f) performed. Figure shows: IAV infected group consists of 10 (a,b), 11 (c,d) or 8 (e,f) 293

mice. SARS-CoV-2 infected group consists of 13 (a,b) or 14 (c-f) mice. Co-infection group consists 294

of 11 (a,b), 10 (c,d) or 7 (e,f) mice. 295

296

17

Day

SARS-CoV-2 infection

0

Day

IAV infection

IAV

0 4 2

sacrificed SARS-2

4 2

sacrificed

SARS-C

oV-2

Co-in

fect

ion

101

102

103

104

105

SA

RS

-CoV

-2 (

pfu

/lu

ng)

SARS-C

oV-2

Co-in

fect

ion

101

102

103

104

105

106

SA

RS

-Co

V-2

(p

fu/n

.t.)

IAV

Co-in

fect

ion

106

107

108

109

PR

8 (

pfu

E/lung)

**

IAV

Co-in

fect

ion

102

103

104

105

106

107

108

PR

8 (

pfu

E/n

.t.)

2

b

***

*ns

c

d e

a

297

Fig. 2: Increased IAV and decreased SARS-CoV-2 viral load in co-infected mice 298

(a) Schematic time lines, K18-hACE2 mice were sacrificed at 4 dpIi and 2 dpSi. IAV viral RNA 299

load was measured in lungs (b) and nasal turbinates (n.t), (c) and the pfu Equivalent per organ 300

(pfuE/organ) were calculated. SARS-CoV-2 viral load was determined by pfu in lungs (d) and in 301

nasal turbinates (e). 302

Each symbol represents one mouse (10 mice each group). Lines represent mean. Error bars 303

represent SE. *p=0.0212; **p=0.0062; ***p=0.0008, ns, not significant, Student’s t-test. 304

18

0

10

20

30

40

C1ra, C1rb

***

*

*

0

50

100

150

C3

*

*

*

0

5

10

15

20

C1s1

*

*

***

0

2

4

6

8

H2-eb1

*

ns

***

0

20

40

60

80

H2-k1

**

*

**

0

2

4

6

8

10

Cd2

**

ns

****

0

5

10

15

20

25

Cd3e

**

*

**

0

10

20

30

Cd8a

*

ns

***

0

5

10

15

20

25

Fas

**

**

***

0

10

20

30

40

50

Cd38

*

*

**

0

10

20

30

40

50

Gzma

*

*

**

0

5

10

15

20

25

Prf1

ns

*

**

0

20

40

60

80

Il-1a

*

*

**

0

50

100

150

Il6

*

*

*

0

50

100

150

200

Ccl5

**

*

**

0

5

10

15

20

25

Ccl8

*

ns***

0

20

40

60

80

Ccl12

*

**

0

500

1000

1500

2000

Cxcl10

*

*

*

0

100

200

300

400

Ifi44

*

*

*

0

200

400

600

800

Ifit1

*

*

*

0

50

100

150

200

250

Ifit3

*

**

**

0

10

20

30

40

50

Ifi27l2a

*

ns

**

0

200

400

600

Irf7

*

**

**

0

10

20

30

40

Mmp3

*

*

***

0

100

200

300

Mmp8

*

*

*

0

5

10

15

Mmp9

ns

*

**

0

50

100

150

200

250

Timp1

*

*

**

0

50

100

150

200

250

Slfn4

*

***

ns

0

200

400

600

Isg15

*

*

*

0

10

20

30

40

50

Zbp1

*

*

***

IAVSARS-CoV-2 PR8 & SARS-CoV-2

Fold

change

com

pare

d n

aiv

e m

ice

3

305

Fig. 3: A panel of increased inflammatory-related genes in the lungs of co-infected mice 306

Expression of various inflammatory related genes in the lungs. RNA were isolated from lungs of 307

K18-hACE2 mice 4 dpIi or 2 dpSi and analyzed by quantitative real-time RT-PCR. Each symbol 308

represents one mouse (4 per each group). Y axis represents fold change of infected compared to 309

19

naïve mice. Colum height represent mean. Error bars represent SE. *p<0.05; **p<0.005; 310

***p<0.0005 Student’s t-test. The genes tested can be divided to different groups: complement 311

system (C1ra, C1rb, C3 and C1s1); antigen presentation (H2-eb1 and H2-k1); recruitment and 312

activation of immune cells (Cd2, Cd3e, Cd8a, Fas, Cd38, Gzma, Prf1); interleukins (Il) (Il-1a and 313

Il6); chemokines (Ccl5, Ccl8, Ccl12, Cxcl10); interferon (Ifi44, Ifit1, Ifit3, Ifi27l2a, Irf7); matrix 314

metalloproteinase (Mmp) and tissue damage (Mmp3, Mmp8, Mmp9, Timp1); and a members of 315

the Schlafen (Slfn) family, Slfn4 and the ubiquitin-like modifier Isg15 and Z-DNA binding 316

protein 1 (Zbp1). 317

318

20

Symbol SARS-CoV-2 Pre-immune

Infection with n

Yes SARS-CoV-2 7

No IAV 7

No SARS-CoV-2 9

Yes IAV & SARS-CoV-2 14

No IAV & SARS-CoV-2 9

0 5 10 15 20 250

50

100

Days post SARS-CoV-2 infection

Perc

ent su

rviv

al

******

**

0 5 10 16 2170

80

90

100

110

Days post IAV infection

% o

f in

itia

l w

eig

ht

0 5 10 15 20 250

50

100

Days post SARS-CoV-2 infection

Perc

ent su

rviv

al

***

ns **

***

0 5 10 16 2170

80

90

100

110

Days post IAV infection

% o

f in

itia

l w

eig

htSymbol

IAV Pre-immune

Infection with n

Yes IAV 5

No IAV 7

Yes IAV & SARS-CoV-2 9

No SARS-CoV-2 9

Yes SARS-CoV-2 8 No IAV & SARS-CoV-2 9

a b

c d

4

319

Fig. 4: Prior immunity to IAV, but not to SARS-CoV-2, rescue co-infected K18-hACE2 mice 320

K18-hACE2 mice were immunized to SARS-CoV-2 by infection with 2pfu/mouse SARS-CoV-2 321

(a,b), or immunized to IAV by 106pfu/mouse IAV (i.m.) (c,d). Thirty days post immunization, pre-322

immunized and non-immunized mice were infected i.n. with the indicated virus. Survival curves 323

(a,c): **p=0.0024; ***P<0.0006. ns, not significant, Log-rank (Mantel-Cox). Percent of weight 324

loss following infection (b,d). Error bars represent SE. Figure legend is shown in tables. n, number 325

of mice in each group. Dashed lines (b,d) represent 1 or 2 survived mice out of 9 or 14, respectively. 326

Figures

Figure 1

Morbidity and mortality in K18-hACE2 mice infected with SARS-CoV-2 following in�uenza infection K18-hACE2 mice were infected with IAV (80 pfu/mice, i.n), followed by infection with SARS CoV-2 (10pfu/mice, i.n) at two (a,b), �ve (c,d) or eight (e,f) dpIi. Percent body weight loss following infection (a,c,e).

Red arrow represents IAV infection. Blue arrow represents SARS-CoV-2 infection. Error bars representstandard errors (SE). p.values are indicated in the �gure as asterisks, and were calculated by Student’s t-test using GraphPad Prism 8.4.3. ***p<0.0002 at 4-7 dpIi (a), and at 9-16 dpIi (c) IAV infection comparedto co-infection. Survival curves (b,d,f):*p=0.04; **p=0.0026; ****p<0.0001 IAV or SARS-CoV-2 infectioncompared to co-infected. ns, 291 not signi�cant, Log-rank (Mantel-Cox). Figure shows one representativeexperiment out of 4 (a,b), 292 2 (c,d), 1 (e,f) performed. Figure shows: IAV infected group consists of 10(a,b), 11 (c,d) or 8 (e,f) 293 mice. SARS-CoV-2 infected group consists of 13 (a,b) or 14 (c-f) mice. Co-infection group consists 294 of 11 (a,b), 10 (c,d) or 7 (e,f) mice.

Figure 2

Increased IAV and decreased SARS-CoV-2 viral load in co-infected mice (a) Schematic time lines, K18-hACE2 mice were sacri�ced at 4 dpIi and 2 dpSi. IAV viral RNA load was measured in lungs (b) and nasalturbinates (n.t), (c) and the pfu Equivalent per organ (pfuE/organ) were calculated. SARS-CoV-2 viral loadwas determined by pfu in lungs (d) and in nasal turbinates (e). Each symbol represents one mouse (10

mice each group). Lines represent mean. Error bars represent SE. *p=0.0212; **p=0.0062; ***p=0.0008, ns,not signi�cant, Student’s t-test.

Figure 3

A panel of increased in�ammatory-related genes in the lungs of co-infected mice Expression of variousin�ammatory related genes in the lungs. RNA were isolated from lungs of K18-hACE2 mice 4 dpIi or 2dpSi and analyzed by quantitative real-time RT-PCR. Each symbol represents one mouse (4 per each

group). Y axis represents fold change of infected compared to naïve mice. Colum height represent mean.Error bars represent SE. *p<0.05; **p<0.005; 310 ***p<0.0005 Student’s t-test. The genes tested can bedivided to different groups: complement 311 system (C1ra, C1rb, C3 and C1s1); antigen presentation (H2-eb1 and H2-k1); recruitment and 312 activation of immune cells (Cd2, Cd3e, Cd8a, Fas, Cd38, Gzma,Prf1); interleukins (Il) (Il-1a and  Il6); chemokines (Ccl5, Ccl8, Ccl12, Cxcl10); interferon (I�44, I�t1, I�t3,I�27l2a, Irf7); matrix metalloproteinase (Mmp) and tissue damage (Mmp3, Mmp8, Mmp9, Timp1); and amembers of  the Schlafen (Slfn) family, Slfn4 and the ubiquitin-like modi�er Isg15 and Z-DNA binding 3protein 1 (Zbp1).

Figure 4

Prior immunity to IAV, but not to SARS-CoV-2, rescue co-infected K18-hACE2 mice K18-hACE2 mice wereimmunized to SARS-CoV-2 by infection with 2pfu/mouse SARS-CoV-2 (a,b), or immunized to IAV by106pfu/mouse IAV (i.m.) (c,d). Thirty days post immunization, preimmunized and non-immunized micewere infected i.n. with the indicated virus. Survival curves (a,c): **p=0.0024; ***P<0.0006. ns, notsigni�cant, Log-rank (Mantel-Cox). Percent of weight loss following infection (b,d). Error bars representSE. Figure legend is shown in tables. n, number of mice in each group. Dashed lines (b,d) represent 1 or 2survived mice out of 9 or 14, respectively.