disease-associated mutations identify a novel region in human sting … · 2017. 4. 3. · 1...

26
The University of Manchester Research Disease-associated mutations identify a novel region in human STING necessary for the control of type I interferon signaling DOI: 10.1016/j.jaci.2016.10.031 Document Version Accepted author manuscript Link to publication record in Manchester Research Explorer Citation for published version (APA): Melki, I., Rose, Y., Uggenti, C., Van Eyck, L., Frémond, M-L., Kitabayashi, N., Rice, G. I., Jenkinson, E. M., Boulai, A., Jeremiah, N., Gattorno, M., Volpi, S., Sacco, O., Terheggen-Lagro, S. W. J., Tiddens, H. A. W. M., Meyts, I., Morren, M-A., De Haes, P., Wouters, C., ... Crow, Y. J. (2017). Disease-associated mutations identify a novel region in human STING necessary for the control of type I interferon signaling. The Journal of allergy and clinical immunology. https://doi.org/10.1016/j.jaci.2016.10.031 Published in: The Journal of allergy and clinical immunology Citing this paper Please note that where the full-text provided on Manchester Research Explorer is the Author Accepted Manuscript or Proof version this may differ from the final Published version. If citing, it is advised that you check and use the publisher's definitive version. General rights Copyright and moral rights for the publications made accessible in the Research Explorer are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. Takedown policy If you believe that this document breaches copyright please refer to the University of Manchester’s Takedown Procedures [http://man.ac.uk/04Y6Bo] or contact [email protected] providing relevant details, so we can investigate your claim. Download date:24. Jun. 2021

Upload: others

Post on 04-Feb-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

  • The University of Manchester Research

    Disease-associated mutations identify a novel region inhuman STING necessary for the control of type I interferonsignalingDOI:10.1016/j.jaci.2016.10.031

    Document VersionAccepted author manuscript

    Link to publication record in Manchester Research Explorer

    Citation for published version (APA):Melki, I., Rose, Y., Uggenti, C., Van Eyck, L., Frémond, M-L., Kitabayashi, N., Rice, G. I., Jenkinson, E. M., Boulai,A., Jeremiah, N., Gattorno, M., Volpi, S., Sacco, O., Terheggen-Lagro, S. W. J., Tiddens, H. A. W. M., Meyts, I.,Morren, M-A., De Haes, P., Wouters, C., ... Crow, Y. J. (2017). Disease-associated mutations identify a novelregion in human STING necessary for the control of type I interferon signaling. The Journal of allergy and clinicalimmunology. https://doi.org/10.1016/j.jaci.2016.10.031Published in:The Journal of allergy and clinical immunology

    Citing this paperPlease note that where the full-text provided on Manchester Research Explorer is the Author Accepted Manuscriptor Proof version this may differ from the final Published version. If citing, it is advised that you check and use thepublisher's definitive version.

    General rightsCopyright and moral rights for the publications made accessible in the Research Explorer are retained by theauthors and/or other copyright owners and it is a condition of accessing publications that users recognise andabide by the legal requirements associated with these rights.

    Takedown policyIf you believe that this document breaches copyright please refer to the University of Manchester’s TakedownProcedures [http://man.ac.uk/04Y6Bo] or contact [email protected] providingrelevant details, so we can investigate your claim.

    Download date:24. Jun. 2021

    https://doi.org/10.1016/j.jaci.2016.10.031https://www.research.manchester.ac.uk/portal/en/publications/diseaseassociated-mutations-identify-a-novel-region-in-human-sting-necessary-for-the-control-of-type-i-interferon-signaling(5bef9358-218d-4871-90bd-7be28bd825c4).html/portal/gillian.rice.html/portal/emma.jenkinson.htmlhttps://www.research.manchester.ac.uk/portal/en/publications/diseaseassociated-mutations-identify-a-novel-region-in-human-sting-necessary-for-the-control-of-type-i-interferon-signaling(5bef9358-218d-4871-90bd-7be28bd825c4).htmlhttps://www.research.manchester.ac.uk/portal/en/publications/diseaseassociated-mutations-identify-a-novel-region-in-human-sting-necessary-for-the-control-of-type-i-interferon-signaling(5bef9358-218d-4871-90bd-7be28bd825c4).htmlhttps://doi.org/10.1016/j.jaci.2016.10.031

  •  

    Disease-associated mutations identify a novel region in human STING necessary 1

    for the control of type I interferon signaling 2

    3

    Isabelle Melki, MD,a,b,c,d Yoann Rose, BSc,a,b Carolina Uggenti, PhD,a,b Lien Van 4

    Eyck, MD,a,b,l Marie-Louise Frémond, MD,a,b,d Naoki Kitabayashi, BSc,a,b Gillian I 5

    Rice, PhD,e Emma M Jenkinson, PhD,e Anaïs Boulai, BSc,a,b Nadia Jeremiah, PhD,b,p 6

    Marco Gattorno, MD,f Sefano Volpi, MD, PhD,f Olivero Sacco, MD,g Suzanne W.J. 7

    Terheggen-Lagro, MD,h Harm A.W.M. Tiddens, MD, PhD,i,j Isabelle Meyts, MD, 8

    PhD,k,l Marie-Anne Morren, MD,l,m Petra De Haes, MD, PhD,l,m Carine Wouters, 9

    MD, PhD,d,k,l Eric Legius, MD,n,o Anniek Corveleyn, PhD,o Frederic Rieux-Laucat, 10

    PhD,b,p Christine Bodemer, MD, PhD,b,q Isabelle Callebaut, PhD,r Mathieu P Rodero, 11

    PhD,a,b Yanick J Crow, MD, PhD,a,b,e 12

    13 aINSERM UMR 1163, Laboratory of Neurogenetics and Neuroinflammation, Paris, 14

    France 15 bParis Descartes University, Sorbonne-Paris-Cité, Institut Imagine, Paris, France 16 cGeneral Pediatrics, Infectious Disease and Internal Medicine Department, Hôpital 17

    Robert Debré, AP-HP, Paris, France 18 dPediatric Hematology-Immunology and Rheumatology Department, Hôpital Necker-19

    Enfants Malades, AP-HP, Paris, France 20 eManchester Centre for Genomic Medicine, Institute of Human Development, Faculty 21

    of Medical and Human Sciences, Manchester Academic Health Centre, Manchester, 22

    United Kingdom 23 fUO Pediatria 2 Reumatologia, G. Gaslini, Genova, Italy 24 gUO Pneumologia, G. Gaslini, Genova, Italy 25 hPaediatric Pulmonologist, Department of Paediatric Pulmonology Emma Children’s 26

    Hospital AMC DD, Amsterdam, Netherlands 27 iDepartment of Radiology, Erasmus Medical Center, Rotterdam, Netherlands 28 jDepartment of Paediatrics, Respiratory Medicine and Allergology,Erasmus Medical 29

    Center-Sophia Children’s Hospital, Zuid-Holland, Netherlands 30 kDepartment of Pediatrics, University Hospitals Leuven, Leuven, Belgium 31 lDepartment of Immunology and Microbiology, Childhood Immunology, University 32

    of Leuven, Leuven, Belgium 33 mDepartment of Dermatology, University Hospitals Leuven, Leuven, Belgium 34

  •  

    nDepartment of Human Genetics, KU Leuven, Leuven, Belgium 35 oClinical Department of Human Genetics, University Hospitals, Leuven, Belgium 36 pINSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmunity, 37

    Paris, France 38 qDermatology Department, National reference center for rare skin diseases MAGEC, 39

    Hôpital Necker-Enfants Malades, Paris, France 40 rCNRS UMR7590, Sorbonne Université, Université Pierre et Marie Curie-Paris6–41

    MNHN–IRD–IUC, Paris, France 42

    43

    Corresponding author: Yanick J Crow 44

    INSERM UMR 1163, Laboratory of Neurogenetics and Neuroinflammation, Paris, 45

    France, 24, boulevard du Montparnasse, 75015 Paris, France. Mail: 46

    [email protected], Phone: +331 42 75 44 51, Fax: +331 44 49 50 70 47

    48

    Sources of funding: This study was funded by the European Research Council, the 49

    National Research Agency (France), the Imagine Institute (France), the Research 50

    Foundation Flanders (FWO) and Telethon (Italy). 51

  •  

    Capsule summary 52

    53

    Three new mutations in TMEM173 are reported in patients presenting STING-54

    associated autoinflammation. These mutations confer constitutive activation 55

    independent of ligand stimulation, revealing a novel functionally important region of 56

    the protein. 57

    58

    Key words 59

    60

    Stimulator of interferon genes (STING), TMEM173, interferon, type I 61

    interferonopathy, STING associated vasculopathy with onset in infancy (SAVI) 62

  •  

    Abstract 63

    64

    Gain-of-function mutations in TMEM173 encoding STING (stimulator of interferon 65

    genes) underlie a recently described type I interferonopathy. We report three 66

    individuals variably exhibiting the core features of STING-associated vasculopathy 67

    with onset in infancy (SAVI) including systemic inflammation, destructive skin 68

    lesions and interstitial lung disease. Molecular and in vitro data demonstrate that the 69

    pathology in these patients is due to substitutions at positions 206, 281 and 284 of the 70

    human protein. These mutations confer cGAMP-independent constitutive activation 71

    of type I interferon signaling through TBK1 (TANK binding kinase). Structural 72

    analysis indicates that these three amino acids lie in a discrete region of the protein, 73

    thereby implicating a novel cluster of amino acids in STING as functionally important 74

    in the regulation of type I interferon signaling. 75

    76

    Key Messages 77

    78

    • p.Cys206, p.Arg281 and p.Arg284 are new mutations in TMEM173 identified 79

    in patients with STING-associated vasculopathy with onset in infancy (SAVI). 80

    • These mutations confer constitutive activation independent of ligand 81

    stimulation, thereby revealing a novel, functionally important region of the 82

    protein. 83

    84

    Abbreviations 85

    86

    3D: Three-dimensional; AMP: Adenosine monophosphate; ANA: Antinuclear 87

    antibodies; BSA: Bovine serum albumin; c: Complementary DNA; CBD: cGAMP 88

    binding domain; cGAMP: 2'3'-cyclic GMP-AMP; cGAS: Cyclic GMP-AMP 89

    synthase; cm: Centimeter; CT: Computed tomography; CTD: C-terminal cyclic 90

    dinucleotide-binding domain; DNA: deoxyribonucleic acid; ER: Endoplasmic 91

    reticulum; ERGIC: ER-Golgi intermediate compartment; ExAC: Exome Aggregation 92

    Consortium; GMP: Cyclic guanosine monophosphate; HEK: Human embryonic 93

    kidney; IFN: Interferon; IFNα: Interferon alpha; IFNAR: Interferon alpha receptor; 94

    IFNß: Interferon beta; IRF3: Interferon regulatory factor 3; ISGs: Interferon-95

  •  

    stimulated genes; JAK1: Janus kinase 1; Kg: Kilogram; mAb: Monoclonal antibody; 96

    MAFFT: Multiple alignment using fast fourier transform; P: Patient; pcDNA: plasmid 97

    cDNA; PCR: Polymerase chain reaction; PDB: Protein Data Bank; RCSB: Research 98

    Collaboratory for Structural Bioinformatics; RNA: Ribonucleic acid; p: Protein; 99

    SAVI: STING associated vasculopathy with onset in infancy; SD: Standard deviation; 100

    STAT1: Signal transducer and activator of transcription 1; STING: Stimulator of 101

    interferon genes; TBK1: TANK binding kinase; TBS: Tris-buffered saline; Tm: 102

    Melting temperature; TMEM173: Transmembrane protein 173; TNF: Tumor necrosis 103

    factor; TYK2: Tyrosine kinase 2; WB: Western blot; WT: Wild type. 104

    105

  •  

    Introduction 106

    107

    Gain-of-function mutations in TMEM173 encoding STING (stimulator of interferon 108

    genes) have been described to cause an autoinflammatory syndrome1–8 belonging to a 109

    recently defined class of disorders referred to as the type I interferonopathies9,10. This 110

    phenotype, known as STING associated vasculopathy with onset in infancy (SAVI), 111

    is characterized by early-onset systemic inflammation with fever, a severe skin 112

    vasculopathy leading in some cases to extensive tissue loss, and interstitial lung 113

    disease resulting in pulmonary fibrosis and end-stage respiratory failure1–5,7,8 114

    115

    To date, twenty-three patients from fifteen families have been reported with gain-of-116

    function mutations in STING1–8, a key adaptor molecule in the cytosolic DNA-117

    sensing pathway, expressed in various endothelial and epithelial cell types, as well as 118

    in haematopoetic cells, such as T cells, macrophages and dendritic cells. Once 119

    stimulated, STING induces the transcription of type I interferons (IFN) and the 120

    expression of a set of IFN-stimulated genes (ISGs), thus establishing an antiviral 121

    state11–13. In all of these published cases the heterozygous substitution involves one of 122

    three amino acids at positions 147, 154, 155, and 166, being located either in the 123

    linker connecting the transmembrane domain of STING to the cyclic dinucleotide 124

    domain (CTD) (p.Val147), or within the N-terminal dimerization region of the CTD 125

    (p.Asn154, p.Val155 and p.Gly166)14,15. It has been suggested that these disease-126

    associated residues play non-redundant roles in retaining the protein on the 127

    endoplasmic reticulum (ER), causing STING to constitutively localize to the ER-128

    Golgi intermediate compartment (ERGIC) and activate downstream signaling through 129

    the TANK binding kinase (TBK1) - Interferon regulatory factor 3 (IRF3) axis15,16. 130

    131

    We describe three individuals variably exhibiting the characteristic features of 132

    STING-associated autoinflammation. However, in contrast to earlier reports, our data 133

    show that the pathology in these patients is due to substitutions at positions 206, 281 134

    and 284 of the human protein, thus implicating a novel region of STING as 135

    functionally important in the regulation of type I IFN signaling. 136

  •  

    Methods 137

    138

    Patient and study approval 139

    The study was approved by the Comité de Protection des Personnes (ID-140

    RCB/EUDRACT: 2014-A01017-40) and undertaken with written informed parental 141

    consent. 142

    143

    Genetic analysis 144

    DNA was extracted from whole blood samples using standard methods. Whole-145

    exome sequencing was performed on genomic DNA from patient 2 (P2) and her 146

    mother using SureSelect Human All Exon kit (Agilent Technologies) for targeted 147

    enrichment and Illumina HiSeq2000 for sequencing. Sanger sequencing was 148

    performed on DNA from patient 1 (P1), patient 3 (P3) and their parents, and from P2 149

    and her mother to confirm the variant found by exome sequencing (Primers in Table 150

    EII). Variant frequency data in controls was derived from the Exome Aggregation 151

    Consortium (ExAC)17. 152

    153

    Structural analysis 154

    The experimental 3D structures of STING were extracted from the Research 155

    Collaboratory for Structural Bioinformatics (RCSB) Protein Data Bank (PDB) and 156

    analyzed using Chimera (https://www.cgl.ucsf.edu/chimera/). 157

    158

    Construct generation 159

    Site-directed mutagenesis was performed to generate the desired TMEM173 variants 160

    by using the Q5® Site-Directed Mutagenesis Kit (E0554S from NEB) following the 161

    manufacturer’s instructions. pMSCV-hygro(+) (Addgene) carrying wild type (WT) 162

    TMEM173 gene was used as a template for the PCR reactions18. This plasmid carries 163

    the haplotype most frequently recorded in the general population (p.Arg71, p.Gly230, 164

    p.Arg232, p.Arg293)19. Mutagenic primer sequences were designed using the NEBase 165

    Changer software (http://nebasechanger.neb.com/). Cycling parameters are indicated 166

    in Table EI. NEB 5-alpha Competent E. coli were transformed with the newly 167

    synthesized pcDNA, and colonies were screened for the presence of the desired 168

    variants (primers in Table EII). 169

    170

  •  

    Cell culture procedures 171

    Human embryonic kidney (HEK) 293T cells were grown in 96-well plates at 37°C in 172

    5% CO2 in DMEM (GIBCO #31966-021) supplemented with 10% (v/v) fetal bovine 173

    serum (GIBCO #10270), 100 U/ml penicillin and 100 mg/ml streptomycin (GIBCO 174

    #15070-063). At 70% confluency, HEK293T cells were co-transfected with 60 ng of 175

    vector pMSCV- hygro(+) either empty or encoding the TMEM173 variants, 40 ng of 176

    IFNß promoter-driven firefly luciferase reporter plasmid (IFNß-pGL3) and 1.4 ng of 177

    constitutively expressed renilla luciferase reporter plasmid (pRL-TK) by using 178

    TransIT-293 (Mirus #MIR2700 from Euromedex). 24 hours later, cells were 179

    stimulated by transfecting 1.3-12 µg/ml 2’3’-cGAMP_STING ligand (tlrl-nacga23, 180

    InvivoGen) using lipofectamine 2000 (#11668027 from Thermo Fischer). 24 hours 181

    after cGAMP stimulation, cells were lysed with passive lysis buffer 5X (#E194A 182

    from Promega) containing protease and phosphatase inhibitors. One third of the lysate 183

    for each condition was used for the luciferase assay, and the remaining material for 184

    protein analysis by western blotting (WB). 185

    186

    Luciferase assay 187

    The IFNß-pGL3 plasmid was used to measure IFNß promoter activity, and the renilla 188

    reporter control plasmid to normalize for transfection efficiency. Luciferase assays 189

    were performed using the Dual-Glo® Luciferase Assay System (E2940 from 190

    Promega) following the manufacturer’s protocol. Luminescence was acquired on a 191

    FLUOstar OPTIMA microplate reader (BMG LABTECH). Firefly luciferase activity 192

    was normalized against renilla luciferase activity. 193

    194

    Western blot 195

    Bolt LDS Sample Buffer (4X) (#B0008 Novex Life Technologies) and Bolt Sample 196

    Reducing agent (10X) (#B0009 Novex Life Technologies) were added to protein 197

    lysates, and samples were resolved on 4-12% Bis-Tris Plus gels (# NW04122BOX 198

    Invitrogen) transferred to nitrocellulose membrane (#IB23002 Invitrogen). When the 199

    phosphorylation status of the protein was investigated, membranes were blocked in 5 200

    % BSA in TBS, and primary phospho-antibodies were incubated overnight in the 201

    blocking solution. Otherwise, membranes were blocked with 5 % milk in TBS and 202

    primary antibodies were incubated overnight in the blocking solution. Proteins were 203

  •  

    blotted with monoclonal anti-STING (R&D MAB7169), anti-Phospho IRF3 Rabbit 204

    mAb (Ser396, #4947 Cell Signaling), anti-IRF3 Rabbit mAb (#11904 Cell Signaling) 205

    and anti-cofilin Rabbit mAb (#5175 Cell Signaling). Primary antibodies were detected 206

    with horseradish peroxidase–conjugated secondary antibody (#7074S and 7076S, Cell 207

    Signaling). All western blot images were captured and quantified with a ChemiDoc 208

    MP imager and Image Lab software (Bio-Rad Laboratories, Hercules, CA) after 209

    adding Clarity Western ECL substrate (Biorad). 210

    211

    STAT1 phosphorylation assay staining 212

    Ficolled PBMCs were treated or not with ruxolitinib 1 µM for 45 minutes at 37°C. 213

    Cells were fixed and permeabilized according to PerFix EXPOSE Kit from Beckman 214

    Coulter (#B26976) recommendations. Cells were stained for pSTAT1 PE-anti-STAT1 215

    pY701, and cell surface marker (PE-CyTM 7-CD19, APC-CD3, BV421-CD8) for 1 216

    hour at room temperature. Flow cytometry analysis was performed on a Gallios 217

    Beckman Coulter flow cytometer. Results were analyzed using Kaluza software v1.3. 218

    219

    Results 220

    221

    Clinical presentation (Table I, Fig 1) 222

    223

    P1 (F1058) is a 7-year-old female born to non-consanguineous parents of white 224

    European ethnicity. She was delivered at term weighing 3.2kg. She presented at the 225

    age of 3 months with feeding difficulties and respiratory distress necessitating oxygen 226

    therapy (Table I). Computed tomography (CT) scan of the lungs at age 1.5 years 227

    revealed extensive ground glass abnormalities, and there was interstitial fibrosis on 228

    lung biopsy without signs of vasculitis (Fig 1, A). Telangiectatic skin lesions on the 229

    cheeks and nose were noted from the age of 4 months (Fig 1, B). She has not 230

    experienced any involvement of the digits. Limited skin biopsy did not reveal any 231

    sign of vasculitis. Elevated transaminases and hepatomegaly prompted a liver biopsy 232

    at age 2 years, when the presence of necrotizing granulomatous hepatitis was 233

    suggested (intralobular granulomas characterized by necrotizing zones with mixed 234

    inflammatory infiltrates). She has demonstrated no further evidence of liver disease. 235

    She had a gastrostomy fitted at age 2 years because of poor weight gain, and 236

    subsequently suffered recurrent skin infections with poor healing around the entry 237

  •  

    site. In light of a continued oxygen requirement and raised inflammatory markers she 238

    was treated with immunosuppressants (steroids, methotrexate and anti-TNFα therapy) 239

    starting at age 23 months, but with limited efficacy. She remains oxygen dependent 240

    with a severe deficit of lung function and markedly limited exercise tolerance. 241

    242

    P2 (F1125) is a 25-year-old female born to non-consanguineous parents of white 243

    European ethnicity. There is no family history of note. She demonstrated intra-uterine 244

    growth retardation (birth weight at term -3 SD), with subsequent failure to thrive 245

    (adult weight / height of 143 cm and 34 kg) requiring a gastrostomy to be placed by 246

    age 1 year until puberty. She suffered recurrent bacterial infections of the upper 247

    respiratory tract, leading to a severe septicemia at the age of five months (due to 248

    Pseudomonas aeruginosa) and palatal and nasal septum necrosis. Screening for 249

    primary immune deficiencies was negative. A widespread livedo was noted from 250

    birth, which worsened gradually with an impressive purpuric aspect in patches on 251

    both cheeks and buttocks, and a marked dark purple marbling on the limbs (Fig 1, C 252

    and D). When last seen at the age of 25 years she had lost a large part of the external 253

    nares, and demonstrated extreme acrocyanosis of the hands and feet. She has never 254

    experienced fevers, and systemic markers of inflammation and autoantibody titers 255

    have been consistently normal until recent antinuclear antibodies (ANA) were 256

    detected at a low level (1/160). Lung function is normal. She is not currently treated. 257

    258

    P3 (F1802) is a 15-year-old male born to non-consanguineous parents of white 259

    European ethnicity. A fixed erythema of the cheeks was noted shortly after birth. 260

    Telangiectasia of the cheeks, buttocks and limbs were observed for the first time at 261

    age 10 years. From the age of 14 years he has suffered recurrent acrocyanosis 262

    moderately influenced by cold temperature, with painful acral erosions that heal as 263

    atrophic scars on the ear helices and digits. Lesional gluteal skin biopsies revealed 264

    dilated capillaries without obvious vasculitis or thrombosis. He has been treated with 265

    nifedipin, pentoxyfillin and acetyl salicylic acid, with limited efficacy. He has never 266

    experienced fevers or other features of systemic disease. Blood analysis, including 267

    markers of inflammation and autoantibody profile, has been consistently normal. 268

    Recent chest CT revealed no evidence of interstitial lung disease. Lung function 269

    testing and a six minute exercise tolerance test were normal. 270

    271

  •  

    Identification of three new variants in STING 272

    273

    A summary of the molecular data relating to these three patients is given in Figure 2 274

    and Table I. A multiple sequence alignment of STING proteins from various species 275

    is provided in Supplementary Figure E1. 276

    277

    P1 (F1058) was observed to carry a c.842G>A (p.Arg281Gln) variant in exon 7 of 278

    TMEM173 (Fig 2, A) which has not been recorded on more than 120,000 control 279

    alleles on the ExAC database. Both parents were WT at this nucleotide, and 280

    microsatellite testing confirmed that the substitution had arisen de novo. The arginine 281

    at position 281 is conserved in mouse, but is an alanine in Xenopus tropicalis and a 282

    glutamate in Danio rerio. We note a similarly limited degree of conservation of the 283

    valine at position 147, substitution of which for a leucine has been previously shown 284

    to act as a gain-of-function mutation1. 285

    286

    P2 (F1125) was observed to carry a c.850A>G (p.Arg284Gly) variant in exon 7 of 287

    TMEM173 (Fig 2, A) which has not been recorded on more than 120,000 control 288

    alleles on the ExAC database. The mother was WT for this variant. DNA was not 289

    available from the father. The arginine is conserved to Nematostella vectensis. 290

    291

    P3 (F1802) was observed to carry a c.617G>A (p.Cys206Tyr) variant in exon 6 of 292

    TMEM173 (Fig 2, A) which has not been recorded on more than 120,000 control 293

    alleles on the ExAC database. Both parents were WT at this nucleotide, and 294

    microsatellite testing confirmed that the substitution had arisen de novo. The cysteine 295

    at this position is generally well conserved, except in Danio rerio where it is replaced 296

    by an alanine. 297

    298

    3D structural analysis reveals a new mutation hotspot in STING 299

    300

    In order to gain further insight into the effect of the three STING substitutions that we 301

    observed in the patients described above, we examined the experimental 3D structures 302

    of the CTD of STING (Fig 2, B). p.Cys206 is buried within the 3D structure, whilst 303

    p.Arg281 and p.Arg284 lie on the surface of the protein. However, these three amino 304

    acids are in close proximity and are aligned, with p.Arg284 located in between 305

  •  

    p.Cys206 and p.Arg281 (Fig 2, C and D). The substitution of the cysteine for a 306

    tyrosine is predicted to induce a steric clash, which might locally destabilize the 3D 307

    structure of the helix bundle and affect the positioning of the 281 / 284 arginine 308

    residues. 309

    310

    p.Cys206, p.Arg281 and p.Arg284 mutations in STING induce over-activation of 311

    IFNß transcription 312

    313

    In order to determine if these mutations were gain-of-function, we used a luciferase-314

    based IFNß transcription reporter assay to measure their impact on type I IFN 315

    induction1,15,16. As previously shown, the p.Val155Met and p.Asn154Ser mutants 316

    induce reporter activity in the absence of ligand (Fig 2, E)1,2,15. We observed a 317

    similarly robust activation with patient-associated substitutions at p.Cys206Tyr, 318

    p.Arg281Gln and p.Arg284Gly (Fig 2, E), supporting the hypothesis that these 319

    variants are constitutively active. 320

    321

    To investigate whether these three variants lie within a novel functional cluster in 322

    STING, we went on to mutate other residues spatially close to the amino acids at 206, 323

    281 and 284. Specifically, we changed p.Asp205 to p.Asp205Tyr and p.Asp205His, 324

    p.Leu265 to p.Leu265Tyr, p.Phe269 to p.Phe269Tyr, p.Gln273 to p.Gln273Ala and 325

    p.Gln273Leu, and p.Glu282 to p.Glu282Ala and p.Glu282Asp (Fig 3, A and B). All 326

    these amino acids are exposed at the surface of the protein, except p.Leu265 which is 327

    buried and in contact with p.Cys206. In contrast to our patient-associated variants, 328

    substitution of any of these residues, except for the aspartate at position 205, behaved 329

    in a manner similar to WT (Fig 3, C and Table EIII). In contrast, substitution of the 330

    p.Asp205 resulted in an upregulation of IFNß reporter activity as for the gain-of-331

    function variants (Fig 3, C and Table EIII). 332

    333

    Altogether, these results suggest the identification of a novel region implicated in 334

    STING regulation, involving at least four amino acids. The fact that all of the 335

    different substitutions tested at these residues (two, four, three and three at p.Asp205, 336

    p.Cys206, p.Arg281 and p.Arg284 respectively) result in increased IFNß activity 337

    indicates that subtle conformational changes are sufficient to disrupt the normal 338

    control of the pathway. The specificity of this effect was emphasized by the fact that 339

  •  

    four other amino acids, p.Leu265, p.Phe269, p.Gln273 and p.Glu282, located in close 340

    proximity in the same tertiary structure did not induce a gain-of-function when 341

    substituted. 342

    343

    STING gain-of-function induced by p.Cys206Tyr, p.Arg281Gln and 344

    p.Arg284Gly is independent of cGAMP binding 345

    346

    While all previously described STING-associated mutations are located close to the 347

    cGAMP binding site, the three novel mutations described here lie on the surface of 348

    the protein, without direct contact with this binding site (Fig 2, B). In order to 349

    determine if the gain of function observed in association with our new mutations 350

    resulted from ligand independent constitutive activation, or reflected an increased 351

    sensitivity to low levels of endogenous cGAMP, we generated double mutants 352

    involving the arginine at 232 which is essential for cGAMP binding14,15. For every 353

    previously and newly described mutation we observed a similarly robust upregulation 354

    of signaling, suggesting that despite clearly distinct localization in the tertiary 355

    structure they all confer ligand-independent constitutive activation (Fig 4, A). 356

    357

    Furthermore, using an antibody against phosphorylated residue p.Ser396 of IRF3, we 358

    recorded increased phosphorylation of IRF3 in cells expressing the three patient-359

    associated mutations, comparable with the previously described p.Val155Met. 360

    Consistent with our luciferase results, this increased IRF3 phosphorylation was not 361

    impaired by introducing a second mutation at p.Arg232Ala, which abrogates cGAMP 362

    binding and IRF3 phosphorylation in the WT state (Fig 4, B and E2, A and D). 363

    364

    STING gain-of-function induced by p.Cys206Tyr, p.Arg281Gln and 365

    p.Arg284Gly is independent of the membrane fusion pathway 366

    367

    Recently, it has been reported that a STING-dependent but cGAS-independent 368

    pathway can induce type I IFN expression in response to membrane fusion of 369

    enveloped RNA viruses20–22. In this model, the arginine at 169 in human STING was 370

    shown to be important for stimulation of IFN expression by liposomes but not 371

    cGAMP, demonstrating that activation by these two stimuli can be mechanistically 372

    separated. p.Arg169 is surface exposed, located just outside the STING dimerization 373

  •  

    domain and facing away from the cGAMP-binding pocket. To determine if the 374

    arginine at position 169 was crucial in the constitutive activation that we observed 375

    with our patient-associated mutations, we substituted an alanine for the wild-type 376

    arginine at 169 (p.Arg169Ala) and expressed the double-mutants in our in vitro 377

    system. We observed no effect of the 169 substitution with mutations at 206, 281 and 378

    284, or with the previously described mutations at 154 and 155, indicating that human 379

    disease-associated constitutive activation of STING is not dependent on this 380

    alternative pathway (Fig 5, A). A similar result was obtained with the double mutant 381

    p.Asp205His and p.Arg169Ala (Fig E2, B and E). Consistent with these data, 382

    constitutively activated constructs bearing the second mutation p.Arg169Ala 383

    demonstrated phosphorylated IRF3 at p.Ser396 in the basal state (Fig 5, B and E2, B 384

    and E). 385

    386

    Constitutive activation of newly identified disease-causing STING mutants is 387

    dependent on IRF3 phosphorylation 388

    389

    Phosphorylation of STING at p.Ser366 by TBK1 is critical for the recruitment and 390

    activation of IRF3 by STING and the subsequent activation of IFN signaling23,24. To 391

    investigate whether SAVI-associated variants were dependent on TBK1-mediated 392

    STING phosphorylation and IRF3 recruitment, we introduced a mutation of serine to 393

    alanine at position 366 (p.Ser366Ala) into our mutant constructs. In all cases, this 394

    substitution led to an abolition of IFNß activation despite similar expression of single 395

    and double STING mutants (Fig 6, A and B). This was associated with loss of IRF3 396

    phosphorylation, indicating that patient-associated mutations are dependent on 397

    phosphorylation at residue 366 for subsequent downstream activation of IRF3 (Fig 6, 398

    B and Fig E2, C and D) and transcription of ISGs. 399

    400

    STING constitutive activation leads to increased phosphorylation of STAT1 in T 401

    and B cells of P3 and normalize after ruxolitinib treatment in vitro 402

    403

    Type I interferons bind to a specific IFNα receptor (IFNAR), thereby activating Janus 404

    kinase 1 (JAK1) and Tyrosine kinase 2 (TYK2), which then induce phosphorylation 405

    of Signal transducer and activator of transcription 1 (STAT1). We investigated 406

    STING constitutive activation in CD4, CD8 and CD19 cells from P3, and observed 407

  •  

    increased levels of STAT1 phosphorylation compared to a control (Fig 7). Consistent 408

    with published data, this upregulation normalized after 45 minutes of treatment with 409

    ruxolitinib in vitro (Fig 7)1,5. 410

    411

    Discussion 412

    413

    Since its identification in 2008 as an adaptor molecule in the cytosolic DNA-sensing 414

    pathway, STING has emerged as a central player in antiviral immunity, 415

    autoinflammation and cancer11,13. Thus, an understanding of the mechanism of action 416

    and control of STING in health and disease is of considerable scientific and medical 417

    importance. 418

    419

    Here we describe three novel variants seen in association with a phenotype consistent 420

    with previously described cases of STING-associated autoinflammation. In two of 421

    these patients we were able to show that the amino acid substitutions arose de novo, 422

    whilst in the third the mother was WT but DNA was unavailable from the father. 423

    These substitutions, involving evolutionarily well-conserved residues, have not been 424

    described in publically available databases comprising more than 120,000 alleles. As 425

    for other molecularly confirmed cases of STING-associated autoinflammation, all 426

    three substitutions were associated with the in vitro induction of IFNß reporter 427

    activity. Considering these observations, it is very likely that the variants we describe 428

    represent pathogenic gain-of-function mutations. 429

    430

    In contrast to all previously published cases of type I interferonopathy related to 431

    mutations in STING, the three variants reported here do not lie in the linker region 432

    connecting the N-terminal transmembrane domain of STING to the CTD or in the 433

    first a-helix (a5) of the CTD involved in dimerization. The function of the arginine 434

    residues at 281 and 284 is currently undefined. However, these residues are exposed 435

    at the surface of the molecule, so that they are unlikely to involve the intramolecular 436

    interaction of the STING dimer. A recent survey of STING mutations in the COSMIC 437

    database led to the demonstration that a substitution of the arginine at 284 to 438

    methionine (p.Arg284Met) was able to induce an IFNß reporter to a significantly 439

    greater extent than WT STING25. Alternative substitutions of arginine 284 for lysine 440

    or threonine (p.Arg284Lys, p.Arg284Thr) also represented hyperactive mutants. The 441

  •  

    results of these experiments were further supported by demonstrating increased IFNß 442

    in tissue culture supernatants, and an enhanced ability to inhibit viral replication. Of 443

    note, the mutation identified in P2 affects this same residue, substituting the arginine 444

    for a glycine. Thus, multiple types of amino acid substitution at 284 confer a gain-of-445

    function, indicating a high degree of specificity for the function of an arginine at this 446

    position. 447

    448

    Similar to the arginine at 284, we found that different substitutions of the arginine at 449

    281 and the cysteine at 206 also resulted in constitutive activation. It is of note that 450

    these three amino acids are aligned at the 3D level, with the arginine at 284 located in 451

    between the cysteine at 206 and the arginine at 281. The substitution of the cysteine 452

    for a tyrosine at 206 is predicted to lead to steric hindrance and locally disturb the 453

    fold, which might affect the positioning of the arginine at 284. However, substituting 454

    other less bulky amino acids at 206 had the same effect, highlighting the specificity of 455

    the WT cysteine at this position. 456

    457

    In order to define the extent of a putative functionally important region involving the 458

    amino acids that we identified through human disease genetics, we went on to mutate 459

    a selection of other residues predicted to lie in close proximity with these disease-460

    associated mutations - specifically, p.Leu265, p.Phe269, p.Gln273 and p.Glu282. We 461

    also derived a number of conservative and non-conservative substitutions at Asp205, 462

    which lies in contact with the p.Arg284. Substitutions at these residues behaved in a 463

    similar manner to WT protein, except for those at position 205, where we observed 464

    enhanced signaling in our in vitro assay similar to the patient-associated mutations. Of 465

    note, constitutive activation has been described previously at the equivalent residue in 466

    murine STING12,26. 467

    468

    In regards of cGAMP-dependency, the involvement of a cGAMP-independent 469

    signaling pathway and TBK1 downstream signaling, our data did not suggest any 470

    difference in the behavior of variants at 206, 281 and 284 compared to previously 471

    described disease-associated mutations at 147, 154 and 155. Recently, Dobbs et al. 472

    suggested that the latter mutations may disrupt the retention of STING in the ER, 473

    resulting in translocation to the ERGIC and activation of TBK1 / IRF3 in the absence 474

    of cGAMP binding15. Furthermore, these mutations appeared to result in continued 475

  •  

    activation, presumed due to a failure of STING degradation. Our data clearly 476

    implicate a novel region of STING as important in type I IFN signaling, leading us to 477

    speculate that the surface-exposed arginine residues at 281 and 284 may also be 478

    involved in ER retention, or in binding to a negative regulator of STING25. 479

    480

    Our results indicate that mutations at residues 206, 281 and 284 of human STING are 481

    responsible for the disabling autoinflammation observed in the three patients that we 482

    describe. Given the favorable effects seen with JAK1/2 inhibition in other patients 483

    with STING gain-of-function mutations, these patients may similarly benefit from 484

    such a therapeutic approach5. Alternative potential treatment strategies might include 485

    ER exit blocking agents, palmitoylation inhibitors or TBK1 antagonists15,27–29. 486

    Considering that a p.Ser366Ala substitution abrogated the constitutive activation seen 487

    with every patient-associated mutation tested in our in vitro model, TBK1 blockade 488

    might be relevant to all SAVI patients. 489

    490

    A major aspect of the pathology due to mutations in TMEM173 appears to relate to a 491

    dysfunction of endothelium consequent upon an upregulation of type I IFN signaling. 492

    The explanation for variable expression observed between, and indeed within 493

    families, will require further study, with such observations likely reflecting 494

    differential environmental exposures and / or modifying genetic factors. Indeed, 495

    whilst the cGAS-STING cytosolic DNA signaling pathway is essential for the 496

    induction of an effective anti-viral response, human genetics is teaching us about the 497

    risk of inflammatory disease associated with variation in this system. Polymorphisms 498

    in STING, and other molecules involved in cytosolic nucleic acid recognition, can 499

    confer differences in signaling to a type I IFN response - a non-binary situation where 500

    a balance is struck between anti-viral priming and the risk of autoinflammation19,30. 501

    Elucidation of the mechanisms of STING regulation is thus of great importance in 502

    understanding both microbial pathogenesis and inflammation. The findings presented 503

    here suggest a previously unappreciated aspect of the control of STING in this 504

    context. 505

  •  

    Acknowledgements 506

    Y.J.C. acknowledges the European Research Council (GA 309449: Fellowship), and a 507

    state subsidy managed by the National Research Agency (France) under the 508

    "Investments for the Future" program bearing the reference ANR-10-IAHU-01. FRL 509

    acknowledges a state subsidy managed by the National Research agency referenced as 510

    Lumugene (ANR-14-CE14-0026-01). IM acknowledges the Programme Santé-511

    Sciences MD/PhD of Imagine Institute. S.V. and M.G. gratefully acknowledge the 512

    financial support of Telethon, Italy (Grant no. # GGP15241A). L.V.E is funded by a 513

    research grant from the Research Foundation Flanders (FWO). We thank Nicolas 514

    Manel for WT STING, p.Val155Met STING and IFNß-pGL3 plasmids. The authors 515

    would like to thank the Exome Aggregation Consortium and the groups that provided 516

    exome variant data for comparison. A full list of contributing groups can be found at 517

    http://exac.broadinstitute.org/about. 518

    519

    Conflict of interest 520

    The authors declare no conflict of interest. 521

  •  

    References 522

    523

    1. Liu, Y. et al. Activated STING in a vascular and pulmonary syndrome. N. 524

    Engl. J. Med. 371, 507–518 (2014). 525

    2. Jeremiah, N. et al. Inherited STING-activating mutation underlies a familial 526

    inflammatory syndrome with lupus-like manifestations. J. Clin. Invest. 124, 5516–527

    5520 (2014). 528

    3. Munoz, J. et al. Stimulator of Interferon Genes-Associated Vasculopathy With 529

    Onset in Infancy: A Mimic of Childhood Granulomatosis With Polyangiitis. JAMA 530

    Dermatol. 151, 872–877 (2015). 531

    4. Omoyinmi, E. et al. Stimulator of interferon genes-associated vasculitis of 532

    infancy. Arthritis Rheumatol. Hoboken NJ 67, 808 (2015). 533

    5. Frémond, M.-L. et al. Efficacy of the Janus Kinase 1/2 Inhibitor Ruxolitinib in 534

    the Treatment of Vasculopathy Associated with TMEM173-Activating Mutations in 535

    three children. J. Allergy Clin. Immunol. (2016). doi:10.1016/j.jaci.2016.07.015 536

    6. König, N. et al. Familial chilblain lupus due to a gain-of-function mutation in 537

    STING. Ann. Rheum. Dis. (2016). doi:10.1136/annrheumdis-2016-209841 538

    7. Picard, C. et al. Severe Pulmonary Fibrosis as the First Manifestation of 539

    Interferonopathy (TMEM173 Mutation). Chest 150, e65-71 (2016). 540

    8. Clarke, S. L. N. et al. Interstitial Lung Disease Caused by STING-associated 541

    Vasculopathy with Onset in Infancy. Am. J. Respir. Crit. Care Med. 194, 639–642 542

    (2016). 543

    9. Crow, Y. J. Type I interferonopathies: a novel set of inborn errors of 544

    immunity. Ann. N. Y. Acad. Sci. 1238, 91–98 (2011). 545

    10. Crow, Y. J. & Casanova, J.-L. STING-associated vasculopathy with onset in 546

    infancy--a new interferonopathy. N. Engl. J. Med. 371, 568–571 (2014). 547

    11. Ishikawa, H. & Barber, G. N. STING is an endoplasmic reticulum adaptor that 548

    facilitates innate immune signalling. Nature 455, 674–678 (2008). 549

    12. Burdette, D. L. et al. STING is a direct innate immune sensor of cyclic di-550

    GMP. Nature 478, 515–518 (2011). 551

    13. Barber, G. N. STING: infection, inflammation and cancer. Nat. Rev. Immunol. 552

    15, 760–770 (2015). 553

    14. Zhang, X. et al. Cyclic GMP-AMP containing mixed phosphodiester linkages 554

    is an endogenous high-affinity ligand for STING. Mol. Cell 51, 226–235 (2013). 555

  •  

    15. Dobbs, N. et al. STING Activation by Translocation from the ER Is 556

    Associated with Infection and Autoinflammatory Disease. Cell Host Microbe 18, 557

    157–168 (2015). 558

    16. Liu, S. et al. Phosphorylation of innate immune adaptor proteins MAVS, 559

    STING, and TRIF induces IRF3 activation. Science 347, aaa2630 (2015). 560

    17. Consortium, E. A. et al. Analysis of protein-coding genetic variation in 60,706 561

    humans. bioRxiv 30338 (2015). doi:10.1101/030338 562

    18. Gentili, M. et al. Transmission of innate immune signaling by packaging of 563

    cGAMP in viral particles. Science 349, 1232–1236 (2015). 564

    19. Yi, G. et al. Single nucleotide polymorphisms of human STING can affect 565

    innate immune response to cyclic dinucleotides. PloS One 8, e77846 (2013). 566

    20. Holm, C. K. et al. Virus-cell fusion as a trigger of innate immunity dependent 567

    on the adaptor STING. Nat. Immunol. 13, 737–743 (2012). 568

    21. Olagnier, D. & Hiscott, J. Breaking the barrier: membrane fusion triggers 569

    innate antiviral immunity. Nat. Immunol. 13, 713–715 (2012). 570

    22. Holm, C. K. et al. Influenza A virus targets a cGAS-independent STING 571

    pathway that controls enveloped RNA viruses. Nat. Commun. 7, 10680 (2016). 572

    23. Tanaka, Y. & Chen, Z. J. STING Specifies IRF3 Phosphorylation by TBK1 in 573

    the Cytosolic DNA Signaling Pathway. Sci Signal 5, ra20-ra20 (2012). 574

    24. Konno, H., Konno, K. & Barber, G. N. Cyclic dinucleotides trigger ULK1 575

    (ATG1) phosphorylation of STING to prevent sustained innate immune signaling. 576

    Cell 155, 688–698 (2013). 577

    25. Tang, E. D. & Wang, C.-Y. Single amino acid change in STING leads to 578

    constitutive active signaling. PloS One 10, e0120090 (2015). 579

    26. Shu, C., Yi, G., Watts, T., Kao, C. C. & Li, P. Structure of STING bound to 580

    cyclic di-GMP reveals the mechanism of cyclic dinucleotide recognition by the 581

    immune system. Nat. Struct. Mol. Biol. 19, 722–724 (2012). 582

    27. Mukai, K. et al. Activation of STING requires palmitoylation at the Golgi. 583

    Nat. Commun. 7, 11932 (2016). 584

    28. Hasan, M. et al. Cutting Edge: Inhibiting TBK1 by Compound II Ameliorates 585

    Autoimmune Disease in Mice. J. Immunol. Baltim. Md 1950 195, 4573–4577 (2015). 586

    29. Hasan, M. & Yan, N. Therapeutic potential of targeting TBK1 in autoimmune 587

    diseases and interferonopathies. Pharmacol. Res. (2016). 588

    doi:10.1016/j.phrs.2016.04.008 589

  •  

    30. Shigemoto, T. et al. Identification of loss of function mutations in human 590

    genes encoding RIG-I and MDA5: implications for resistance to type I diabetes. J. 591

    Biol. Chem. 284, 13348–13354 (2009). 592

    31. Ouyang, S. et al. Structural analysis of the STING adaptor protein reveals a 593

    hydrophobic dimer interface and mode of cyclic di-GMP binding. Immunity 36, 594

    1073–1086 (2012). 595

    32. Katoh, K. & Standley, D. M. MAFFT multiple sequence alignment software 596

    version 7: improvements in performance and usability. Mol. Biol. Evol. 30, 772–780 597

    (2013). 598

    33. Robert, X. & Gouet, P. Deciphering key features in protein structures with the 599

    new ENDscript server. Nucleic Acids Res. 42, W320-324 (2014). 600

    601

  •  

    Figure Legends 602

    603

    TABLE I. Clinical and molecular data. 604

    ANA: Antinuclear Antibodies; ANCA: Anti-neutrophilic cytoplasmic antibodies; F: 605

    Female; M: Male 606

    607

    FIG 1. Clinical and radiological phenotypes. A. High resolution chest CT scan of 608

    P1 demonstrating evolution of bilateral alveolar and interstitial disease with ground-609

    glass lesions and interlobular septa thickening (at ages 3, 5 and 8). Improvement of 610

    interstitial lung disease between ages 3 and 5 years was observed after oral steroid 611

    treatment. B. Telangiectatic skin lesions on the cheeks and nose of P1. C. Purpuric 612

    aspect to patches on both cheeks of P2 at age 1.5 years. D. Widespread livedo and 613

    marked dark purple marbling on the limbs of P2 at age 1.5 years. y: years. 614

    615

    FIG 2. Constitutively activated mutations of TMEM173. A. Schematic 616

    representation of the TMEM173 protein highlighting the four transmembrane domains 617

    (TM1, 2, 3 and 4), the cGAMP binding domain (CBD) and the C terminal tail domain 618

    (CTD), as well as four phosphorylation sites (P) at p.S345, p.S358, p.S366 and 619

    p.S379. In black, previously described mutations in exon 5 (p.V147L; p.V147M; 620

    p.N154S; p.V155M). In red, 3 novel mutations in exons 6 and 7 (p.C206Y: P3; 621

    p.R281Q: P1; p.R284G: P2). B. Ribbon representation of the 3D structure of the 622

    STING CTD dimer in complex with cyclic-di-GMP (PDB reference 4ef4), indicating 623

    the position of the three novel amino acids found mutated in this study, as well as 624

    amino acids previously reported with mutations and involved in the CTD dimer 625

    interface, at the bottom of the ligand binding site. C. Focus on the 3D aligned 626

    positions of p.C206, p.R281 and p.R284. D. Surface representation of the 3D 627

    structure of the STING CTD dimer (orthogonal view relative to panel B), showing the 628

    accessibility of the three amino acids with patient-associated substitutions (p.C206, 629

    p.R281 and p.R284). E. IFNß-Luc reporter assay. HEK293T cells were transfected 630

    with IFNß-Firefly Luc, Renilla Luciferase and indicated human STING plasmids 631

    (EV: Empty vector; WT: Wild type STING; p.N154S p.V155M; p.C206Y; p.R281Q; 632

    p.R284G) and stimulated with increasing doses of cGAMP (none; 4 µg/µl; 12 µg/µl). 633

    Luciferase activity was determined by Dual-luc assay 24 hours post-stimulation. 634

    Mean and SDs shown, representative of 5 different experiments in triplicate. 635

  •  

    636

    FIG 3. Analysis of substitutions conferring constitutive activation of STING. A. 637

    and B. Localization of amino acids on surface and ribbon representations of the 638

    STING dimer 3D structure (PDB reference 4ef4)31. Colored amino acids in which 639

    mutations confer constitutive activation: p.D205 in green, p.C206 in yellow, p.R281 640

    in red and p.R284 in blue. Amino acids where substitutions did not confer constitutive 641

    activation are shown in grey. C. IFNß-Luc reporter assay. HEK293T cells were 642

    transfected with IFNß-Firefly Luc, Renilla Luciferase and indicated human STING 643

    plasmid (EV: Empty vector; WT: Wild type STING; p.N154A; p.V155M; p.D205Y 644

    or H; p.C206Y, F, L or S; p.R281Q, K or V; p.R284G, H or K) and stimulated with 645

    increasing doses of cGAMP (none; 4 µg/µl; 12 µg/µl). Luciferase activity was 646

    determined by Dual-luc assay 24 hour post-stimulation. Mean and SDs shown, 647

    representative of 3 different experiments in triplicate. 648

    649

    FIG 4. Constitutive activation conferred by p.C206Y, p.R281Q and p.R284G is 650

    independent of cGAMP binding. A. IFNß-Luc reporter assay. HEK293T cells were 651

    transfected with IFNß-Firefly Luc, Renilla Luciferase and indicated human STING 652

    plasmid (EV: Empty vector; WT: Wild type STING; p.V155M; p.C206Y; p.R281Q; 653

    p.R284G with or without a second mutation p.R232A) and stimulated with increasing 654

    doses of cGAMP (none; 4 µg/µl; 12 µg/µl). Luciferase activity was determined by 655

    Dual-luc assay 24 hours post-stimulation. Mean and SDs shown, representative of 4 656

    different experiments in triplicate. B. Immunoblot (IB) analysis of STING, 657

    phosphorylated IRF3 (p.S396), total IRF3 and cofilin in whole cell lysates of 658

    HEK293T cells transfected with empty vector (EV), wild type (WT) human STING 659

    with or without cGAMP stimulation, p.V155M, p.C206Y, p.R281Q and p.R284G 660

    (unstimulated), either alone or in combination with a second mutation at p.R232A. Of 661

    note, in HEK293T cells transfected with WT plasmids, the amount of STING was 662

    greater at basal state than upon stimulation with cGAMP, which is consistent with a 663

    model of STING degradation, possibly through an autophagic-mediated mechanism24. 664

    665

    FIG 5. Constitutive activation conferred by p.C206Y, p.R281Q and p.R284G 666

    mutants is independent of non-canonical pathway STING engagement. A. IFNß-667

    Luc reporter assay. HEK293T cells were transfected with IFNß-Firefly Luc, Renilla 668

    Luciferase, and indicated human STING plasmid (EV: Empty vector; WT: Wild type 669

  •  

    STING; p.N154S; p.V155M; p.C206Y; p.R281Q; p.R284G with or without a second 670

    mutation p.R169A) and stimulated with increasing doses of cGAMP (none; 4 µg/µl; 671

    12 µg/µl). Luciferase activity was determined by Dual-luc assay 24 hours post-672

    stimulation. Mean and SDs shown, representative of 4 different experiments in 673

    triplicate. B. Immunoblot (IB) analysis of STING, IRF3 phosphorylated at p.S396, 674

    total IRF3 and cofilin in whole cell lysate of HEK293T cells transfected with empty 675

    vector (EV), wild type (WT) human STING with or without cGAMP stimulation, 676

    p.N154S, p.V155M, p.C206Y, p.R281Q and p.R284G unstimulated, with or without 677

    double mutant p.R169A. 678

    679

    FIG 6. Activation of newly identified disease-causing STING mutants relies on 680

    IRF3 phopshorylation. A. IFNß-Luc reporter assay. HEK293T cells were 681

    transfected with IFNß-Firefly Luc, Renilla Luciferase, and indicated human STING 682

    plasmid (EV: Empty vector; WT: Wild type STING; p.V155M; p.C206Y; p.R281Q; 683

    p.R284G with or without a second mutation p.S366A) and stimulated with cGAMP 684

    (12 µg/µl) or not. Luciferase activity was determined by Dual-luc assay 24 hours 685

    post-stimulation. Mean and SDs shown, representative of 3 different experiments in 686

    triplicate. B. Immunoblot (IB) analysis of STING, IRF3 phosphorylated at p.S396, 687

    total IRF3 and cofilin in whole cell lysate of HEK293T cells transfected with empty 688

    vector (EV), wild type (WT) human STING with or without cGAMP stimulation, 689

    p.V155M, p.C206Y, p.R281Q and p.R284G unstimulated, with or without a second 690

    mutation p.S366A. 691

    692

    FIG 7. STAT1 phosphorylation status in CD4, CD8 and CD19 cells before and 693

    after treatment with ruxolitinib in vitro. Increased basal STAT1 phosphorylation 694

    was observed in CD4, CD8 and CD19 cells from P3, which normalized after in vitro 695

    treatment with the JAK1/2 blocker ruxolitinib. 696

    697

    698

    TABLE EI. Primer sequences used for site directed mutagenesis. 699

    700

    TABLE EII. TMEM173 primer sequences. 701

    702

    TABLE EIII. Summary data of substitutions of STING tested in vitro. WT: Wild 703

  •  

    type. 704

    705

    FIG E1. Multiple sequence alignment of STING proteins. Sequences are 706

    designated using UniProt identifiers. The regular secondary structures, as observed 707

    from experimental 3D structure (pdb 4ef4) are reported at the top. MAFFT and 708

    ESPript were used for building and rendering of the multiple alignment32,33. BOVIN: 709

    Bovine; XENTR: Xenopus tropicalis; DANRE: Danio rerio, NEMVE: Nematostella 710

    vectensis. 711

    712

    FIG E2. Double mutant p.D205H with p.R232A, p.R169A or p.S366A. A, B and 713

    C. IFNß-Luc reporter assay. HEK293T cells were transfected with IFNß-Firefly Luc, 714

    Renilla Luciferase, and indicated human STING plasmid (EV: Empty vector; WT: 715

    Wild type STING; p.D205H with or without a second mutation p.R232A, p.R169A or 716

    p.S366A respectively) and stimulated with increasing doses of cGAMP (none; 4 717

    µg/µl; 12 µg/µl). Luciferase activity was determined by Dual-luc assay 24 hours post-718

    stimulation. Mean and SDs shown, representative of 2 different experiments in 719

    triplicate. D and E. Immunoblot (IB) analysis of STING, IRF3 phosphorylated at 720

    p.S396, total IRF3 and cofilin in whole cell lysate of transfected HEK293T 721

    transfected with wild type (WT) human STING with or without cGAMP stimulation 722

    and p.D205H unstimulated, with or without double mutant p.R232A, p.S366A or 723

    p.R169A. 724

    * Uncharacterized band. Of note, in HEK293T cells transfected with WT plasmids, 725

    the amount of STING was greater at basal state than upon stimulation with cGAMP, 726

    which is consistent with a model of STING degradation, possibly through an 727

    autophagic-mediated mechanism24. 728