Abstract

Pulmonary arterial hypertension (PAH) is a disorder with a large genetic component. Biallelic mutations of EIF2AK4, which encodes the kinase GCN2, are causal in two ultra-rare subtypes of PAH, pulmonary veno-occlusive disease and pulmonary capillary haemangiomatosis. EIF2AK4 variants of unknown significance have also been identified in patients with classical PAH, though their relationship to disease remains unclear. To provide patients with diagnostic information and enable family testing, the functional consequences of such rare variants must be determined, but existing computational methods are imperfect. We applied a suite of bioinformatic and experimental approaches to sixteen EIF2AK4 variants that had been identified in patients. By experimentally testing the functional integrity of the integrated stress response (ISR) downstream of GCN2, we determined that existing computational tools have insufficient sensitivity to reliably predict impaired kinase function. We determined experimentally that several EIF2AK4 variants identified in patients with classical PAH had preserved function and are therefore likely to be non-pathogenic. The dysfunctional variants of GCN2 that we identified could be subclassified into three groups: misfolded, kinase-dead, and hypomorphic. Intriguingly, members of the hypomorphic group were amenable to paradoxical activation by a type-1½ GCN2 kinase inhibitor. This experiment approach may aid in the clinical stratification of EIF2AK4 variants and potentially identify hypomorophic alleles receptive to pharmacological activation.

Introduction

Aberrant vascular remodelling in pulmonary arterial hypertension (PAH) raises pressures in the pulmonary vasculature to cause right heart failure [1]. Affected young adults often suffer progressive disease leading to premature death. Although classical PAH is most frequently caused by mutations in the TGFβ/BMP signalling axis [2–5], rare subtypes such as pulmonary veno-occlusive disease (PVOD) and pulmonary capillary haemangiomatosis (PCH) have distinct genetic associations and are refractory to current clinical management [6]. With no effective treatments apart from lung transplantation, death occurs within a year in 72% of patients diagnosed with these aggressive PAH subtypes [7].

Since the first report in 2014 linking biallelic mutations of EIF2AK4 to PVOD [8], approximately one hundred EIF2AK4 alleles have been reported to be associated with PAH and its subtypes [4, 8–14]. Although frameshift mutations constitute a large proportion, approximately a third [34] of these alleles are missense variants, the functional consequences of which are unknown (Fig. 1) [15]. Validating the pathogenicity of such variants of uncertain significance (VUSs) would aid in diagnosis, enable cascade genetic testing of relatives, and recruitment of patients to chemoprotective clinical trials [6].

Schematic of known patient-specific missense variants. GCN2 schematic and its domains. Variants over the domain schematic were from patients with classical PAH. Variants below were from patients with PCH and PVOD. Variants reported to be benign/likely benign in ClinVar database33 are in blue, those reported as pathogenic/likely pathogenic are in red, black represents variants of uncertain significance. Occurrences represent cumulative allele counts in the gnomAD database34 or published reports8–14. Variants highlighted in yellow are analysed experimentally in this study.
Figure 1

Schematic of known patient-specific missense variants. GCN2 schematic and its domains. Variants over the domain schematic were from patients with classical PAH. Variants below were from patients with PCH and PVOD. Variants reported to be benign/likely benign in ClinVar database33 are in blue, those reported as pathogenic/likely pathogenic are in red, black represents variants of uncertain significance. Occurrences represent cumulative allele counts in the gnomAD database34 or published reports8–14. Variants highlighted in yellow are analysed experimentally in this study.

EIF2AK4 encodes GCN2, a large serine/threonine kinase homodimer that responds to amino acid depletion by monitoring the efficiency of protein synthesis through its interaction with stalled ribosomes [16, 17]. GCN2 is comprised of an N-terminal RWD domain (20–137) involved in protein–protein interactions, pseudokinase (276–539) and kinase domains (585–1016) including GCN2’s dimerisation interface, an HisRS-like domain (1058–1490) and C-terminal domain (1533–1649) where binding to ribosomes and recognition of uncharged tRNAs occur [18]. When activated, GCN2 triggers a cellular signalling pathway termed the integrated stress response (ISR) by phosphorylating the α subunit of eukaryotic translation initiation factor 2 (eIF2α) [19, 20]. This attenuates most protein synthesis, while enhancing the translation of ISR-specific mRNAs owing to the presence of upstream open reading frames (uORFs) in their 5’UTRs [21]; an example is the transcription factor ATF4. PPP1R15A, a selective eIF2α phosphatase subunit, is similarly regulated and its expression eventually terminates the ISR [20–22]. Due to its fundamental biological roles, disruption of the ISR is implicated in many diseases [23–25].

Technological advances have made genomic sequencing readily available in the clinic, leading to a proliferation in the number of VUSs encountered. Currently, strategies to predict the impact of novel genetic variants are largely restricted to computational methods that rely on evolutionary conservation (e.g. SIFT, PolyPhen) [26] or integrate a range of scores from existing predictive tools (e.g. CADD, REVEL) [27, 28]. Other methodologies include the computation of folding free energy differences (FoldX) [29], which predicts protein stability. More recently, deep learning approaches have been developed (EVE, AlphaMissense) [30, 31] that account for both evolutionary constraints and structural information. Although these have improved predictive accuracy, computational approaches remain imperfect. We set out to examine patient-specific EIF2AK4 missense mutations both in silico and experimentally using existing bioinformatic tools and cell biological assays. In so doing, we subclassified PAH-associated GCN2 variants in functional (likely benign), destabilised/misfolded, or kinase impaired. Interestingly, a subset of the kinase impaired variants showed preserved target engagement. These hypomorphic variants were amenable to pharmacological rescue using an ATP-competitive GCN2 inhibitor [32]. Based on these results, we propose a simple methodology for the experimental validation of the functionality of EIF2AK4 VUSs, which outperforms existing computational approaches.

Results

Computational analysis of PAH-associated EIF2AK4 missense variants suggests heterogeneity

Thirty-four missense variants of EIF2AK4 (RefSeq: NC_000015.10) have been published [8–14] or reported in ClinVar [33] to be associated with PAH (Table 1). Genome sequencing of the general population [34] suggests that most of these variants are rare, with allele frequencies <  0.01, though three, I441L, E556G and G1306C, are common polymorphisms with frequencies > 0.1 (Fig. 1). Nine have already been classified as pathogenic/likely pathogenic (R585Q, G599R, V607G, L643R, S909R, G1109R, P1115L, H1202L and L1295R, depicted in red) and seven as benign/likely benign, including the three common variants (Y34C, I441L, E556G, P674S, D888H, G1306C and K1336R, depicted in blue), but most remain uncharacterised VUSs (Fig. 1, Table 1).

Table 1

Known pulmonary hypertension-associated missense variants of GCN2.

DNA BaseAmino AcidPubicationClinVar classificationPatient DiagnosisSIFTPolyPhen2CADDv1.6REVELAlpha MissenseFoldX: ΔΔG (kcal/mol)Integrative in silico functional prediction
44C > TP15LHadinnapola 2017VUSPAH (Het)T022.60.1330.08340.23Benign
145-2A > GY34CUK cohort (unpublished)Benign/likely-benignPVOD (C Het)T0.999320.6340.45893.79Misfolded
220G > AD74NHadinnapola 2017VUSPAH (Het)NT0.954280.1660.17260.38Benign
361G > AV121MMontani 2017VUSPVOD (C Het)T1330.4670.7125−1.04Benign
1078C > TR360CClinVar RCV001287133VUSPCHNT0.99926.30.5660.0831.68Uncertain
1321A > CI441LClinVar RCV000999785Benign/likely-benignPCHT0.00116.90.0750.0839−0.70Benign
1466C > TL489PGómez 2015VUSPAHNT1290.8950.97377.89Misfolded
1667A > GE556GClinVar RCV000755517Benign/likely-benignPCHT022.50.0880.05230.63Benign
1694 T > CI565TClinVar RCV001285279VUSPCHT0.83522.20.3070.2246−0.36Benign
1754G > AR585QEyries 2014; Montani 2017Pathogenic/likely-pathogenicPVOD; PCHNT1320.8610.92722.12Kinase-deficient
1795G > CG599RHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (Hom)NT1320.9520.99910.83Kinase-deficient
1820 T > GV607GHadinnapola 2017Pathogenic/likely-pathogenicPAH (C Het); PCHNT0.998330.5570.80394.73Misfolded
1928 T > GL643REyries 2014; Levy 2016; Montani 2017Pathogenic/likely-pathogenicPVOD; PCHNT0.999320.8890.99119.65Misfolded
2020C > TP674SClinVar RCV001286399Benign/likely-benignPCHT0.02912.10.0770.0694−1.45Benign
2516 T > CI839THadinnapola 2017VUSPAH (Het)NT0.9929.60.7810.98542.90Benign
2609C > TA870VMontani 2017VUSPVOD (C Het)T128.10.6580.99430.95Uncertain
2662G > CD888HClinVar RCV001287878Benign/likely-benignPCHNT0.99824.80.2370.10050.59Benign
2727C > GS909RHadinnapola 2017Pathogenic/likely-pathogenicPAH (Het); PCHNT1280.4460.99855.67Misfolded
2827A > GT943AHadinnapola 2017VUSPAH (C Het)NT0.99725.40.4640.85971.54Benign
2965C > TR989WEichstaedt 2022VUSPVODNT128.90.8620.983838.59Misfolded
3218G > TR1073LHadinnapola 2017VUSPAHNT0.995320.4660.33740.15Benign
3325G > AG1109RHadinnapola 2017Pathogenic/likely-pathogenicPAH (C Het); PCHNT1320.7660.98569.20Uncertain
3344C > TP1115LGómez 2015; Tenorio 2015Pathogenic/likely-pathogenicPAH; PVOD; IPAH; PCHNT129.70.7020.84151.46Uncertain
3380C > TA1127VEichstaedt 2022VUSPVODT1320.2620.60496.08Misfolded
3443 T > CL1148SEichstaedt 2022VUSCTD-APAHT128.40.3410.26481.11Benign
3604C > TH1202YHadinnapola 2017VUSPAH (Het)T0.99626.50.7160.812−0.44Benign
3605A > TH1202LHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (Hom)NT0.99828.40.8020.95073.00Uncertain
3722A > GE1241GHadinnapola 2017VUSPAHNT0.971330.3420.70691.50Benign
3884 T > GL1295RHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (C Het)NT128.80.8140.94084.89Misfolded
3916G > TG1306CClinVar RCV000999798Benign/likely-benignPCHNT1310.3070.25962.26Uncertain
4007A > GK1336RClinVar RCV003103836Benign/likely-benignPCHT015.80.0630.05640.14Benign
4204G > TV1402LMontani 2017VUSIPAHT0.99724.70.2850.4847−0.19Benign
4646G > AR1549HHadinnapola 2017VUSPAH (Het)NT0.998320.3180.65170.30Benign
4910 T > CL1637PMontani 2017VUSPVOD (C Het)NT1310.6240.99825.98Misfolded
K619R-kinase-deadNT1320.7830.97690.20Kinase-deficient
DNA BaseAmino AcidPubicationClinVar classificationPatient DiagnosisSIFTPolyPhen2CADDv1.6REVELAlpha MissenseFoldX: ΔΔG (kcal/mol)Integrative in silico functional prediction
44C > TP15LHadinnapola 2017VUSPAH (Het)T022.60.1330.08340.23Benign
145-2A > GY34CUK cohort (unpublished)Benign/likely-benignPVOD (C Het)T0.999320.6340.45893.79Misfolded
220G > AD74NHadinnapola 2017VUSPAH (Het)NT0.954280.1660.17260.38Benign
361G > AV121MMontani 2017VUSPVOD (C Het)T1330.4670.7125−1.04Benign
1078C > TR360CClinVar RCV001287133VUSPCHNT0.99926.30.5660.0831.68Uncertain
1321A > CI441LClinVar RCV000999785Benign/likely-benignPCHT0.00116.90.0750.0839−0.70Benign
1466C > TL489PGómez 2015VUSPAHNT1290.8950.97377.89Misfolded
1667A > GE556GClinVar RCV000755517Benign/likely-benignPCHT022.50.0880.05230.63Benign
1694 T > CI565TClinVar RCV001285279VUSPCHT0.83522.20.3070.2246−0.36Benign
1754G > AR585QEyries 2014; Montani 2017Pathogenic/likely-pathogenicPVOD; PCHNT1320.8610.92722.12Kinase-deficient
1795G > CG599RHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (Hom)NT1320.9520.99910.83Kinase-deficient
1820 T > GV607GHadinnapola 2017Pathogenic/likely-pathogenicPAH (C Het); PCHNT0.998330.5570.80394.73Misfolded
1928 T > GL643REyries 2014; Levy 2016; Montani 2017Pathogenic/likely-pathogenicPVOD; PCHNT0.999320.8890.99119.65Misfolded
2020C > TP674SClinVar RCV001286399Benign/likely-benignPCHT0.02912.10.0770.0694−1.45Benign
2516 T > CI839THadinnapola 2017VUSPAH (Het)NT0.9929.60.7810.98542.90Benign
2609C > TA870VMontani 2017VUSPVOD (C Het)T128.10.6580.99430.95Uncertain
2662G > CD888HClinVar RCV001287878Benign/likely-benignPCHNT0.99824.80.2370.10050.59Benign
2727C > GS909RHadinnapola 2017Pathogenic/likely-pathogenicPAH (Het); PCHNT1280.4460.99855.67Misfolded
2827A > GT943AHadinnapola 2017VUSPAH (C Het)NT0.99725.40.4640.85971.54Benign
2965C > TR989WEichstaedt 2022VUSPVODNT128.90.8620.983838.59Misfolded
3218G > TR1073LHadinnapola 2017VUSPAHNT0.995320.4660.33740.15Benign
3325G > AG1109RHadinnapola 2017Pathogenic/likely-pathogenicPAH (C Het); PCHNT1320.7660.98569.20Uncertain
3344C > TP1115LGómez 2015; Tenorio 2015Pathogenic/likely-pathogenicPAH; PVOD; IPAH; PCHNT129.70.7020.84151.46Uncertain
3380C > TA1127VEichstaedt 2022VUSPVODT1320.2620.60496.08Misfolded
3443 T > CL1148SEichstaedt 2022VUSCTD-APAHT128.40.3410.26481.11Benign
3604C > TH1202YHadinnapola 2017VUSPAH (Het)T0.99626.50.7160.812−0.44Benign
3605A > TH1202LHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (Hom)NT0.99828.40.8020.95073.00Uncertain
3722A > GE1241GHadinnapola 2017VUSPAHNT0.971330.3420.70691.50Benign
3884 T > GL1295RHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (C Het)NT128.80.8140.94084.89Misfolded
3916G > TG1306CClinVar RCV000999798Benign/likely-benignPCHNT1310.3070.25962.26Uncertain
4007A > GK1336RClinVar RCV003103836Benign/likely-benignPCHT015.80.0630.05640.14Benign
4204G > TV1402LMontani 2017VUSIPAHT0.99724.70.2850.4847−0.19Benign
4646G > AR1549HHadinnapola 2017VUSPAH (Het)NT0.998320.3180.65170.30Benign
4910 T > CL1637PMontani 2017VUSPVOD (C Het)NT1310.6240.99825.98Misfolded
K619R-kinase-deadNT1320.7830.97690.20Kinase-deficient

ClinVar classification (VUS: variant of uncertain significance) and patient diagnosis where available: Hom = homozygous, Het = heterozygous, C Het = compound heterozygous. Scores SIFT (T: tolerated; NT: not tolerated), PolyPhen2, CADD v1.6, REVEL and AlphaMissense (high scores corresponding to more severe consequences). FoldX used to estimate difference in Gibbs free energy (ΔΔG, kcal/mol) between a variant and the non-mutated GCN2 (scores > 1 indicate predicted destabilisation, scores > 3 indicate predicted severe misfolding).

Table 1

Known pulmonary hypertension-associated missense variants of GCN2.

DNA BaseAmino AcidPubicationClinVar classificationPatient DiagnosisSIFTPolyPhen2CADDv1.6REVELAlpha MissenseFoldX: ΔΔG (kcal/mol)Integrative in silico functional prediction
44C > TP15LHadinnapola 2017VUSPAH (Het)T022.60.1330.08340.23Benign
145-2A > GY34CUK cohort (unpublished)Benign/likely-benignPVOD (C Het)T0.999320.6340.45893.79Misfolded
220G > AD74NHadinnapola 2017VUSPAH (Het)NT0.954280.1660.17260.38Benign
361G > AV121MMontani 2017VUSPVOD (C Het)T1330.4670.7125−1.04Benign
1078C > TR360CClinVar RCV001287133VUSPCHNT0.99926.30.5660.0831.68Uncertain
1321A > CI441LClinVar RCV000999785Benign/likely-benignPCHT0.00116.90.0750.0839−0.70Benign
1466C > TL489PGómez 2015VUSPAHNT1290.8950.97377.89Misfolded
1667A > GE556GClinVar RCV000755517Benign/likely-benignPCHT022.50.0880.05230.63Benign
1694 T > CI565TClinVar RCV001285279VUSPCHT0.83522.20.3070.2246−0.36Benign
1754G > AR585QEyries 2014; Montani 2017Pathogenic/likely-pathogenicPVOD; PCHNT1320.8610.92722.12Kinase-deficient
1795G > CG599RHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (Hom)NT1320.9520.99910.83Kinase-deficient
1820 T > GV607GHadinnapola 2017Pathogenic/likely-pathogenicPAH (C Het); PCHNT0.998330.5570.80394.73Misfolded
1928 T > GL643REyries 2014; Levy 2016; Montani 2017Pathogenic/likely-pathogenicPVOD; PCHNT0.999320.8890.99119.65Misfolded
2020C > TP674SClinVar RCV001286399Benign/likely-benignPCHT0.02912.10.0770.0694−1.45Benign
2516 T > CI839THadinnapola 2017VUSPAH (Het)NT0.9929.60.7810.98542.90Benign
2609C > TA870VMontani 2017VUSPVOD (C Het)T128.10.6580.99430.95Uncertain
2662G > CD888HClinVar RCV001287878Benign/likely-benignPCHNT0.99824.80.2370.10050.59Benign
2727C > GS909RHadinnapola 2017Pathogenic/likely-pathogenicPAH (Het); PCHNT1280.4460.99855.67Misfolded
2827A > GT943AHadinnapola 2017VUSPAH (C Het)NT0.99725.40.4640.85971.54Benign
2965C > TR989WEichstaedt 2022VUSPVODNT128.90.8620.983838.59Misfolded
3218G > TR1073LHadinnapola 2017VUSPAHNT0.995320.4660.33740.15Benign
3325G > AG1109RHadinnapola 2017Pathogenic/likely-pathogenicPAH (C Het); PCHNT1320.7660.98569.20Uncertain
3344C > TP1115LGómez 2015; Tenorio 2015Pathogenic/likely-pathogenicPAH; PVOD; IPAH; PCHNT129.70.7020.84151.46Uncertain
3380C > TA1127VEichstaedt 2022VUSPVODT1320.2620.60496.08Misfolded
3443 T > CL1148SEichstaedt 2022VUSCTD-APAHT128.40.3410.26481.11Benign
3604C > TH1202YHadinnapola 2017VUSPAH (Het)T0.99626.50.7160.812−0.44Benign
3605A > TH1202LHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (Hom)NT0.99828.40.8020.95073.00Uncertain
3722A > GE1241GHadinnapola 2017VUSPAHNT0.971330.3420.70691.50Benign
3884 T > GL1295RHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (C Het)NT128.80.8140.94084.89Misfolded
3916G > TG1306CClinVar RCV000999798Benign/likely-benignPCHNT1310.3070.25962.26Uncertain
4007A > GK1336RClinVar RCV003103836Benign/likely-benignPCHT015.80.0630.05640.14Benign
4204G > TV1402LMontani 2017VUSIPAHT0.99724.70.2850.4847−0.19Benign
4646G > AR1549HHadinnapola 2017VUSPAH (Het)NT0.998320.3180.65170.30Benign
4910 T > CL1637PMontani 2017VUSPVOD (C Het)NT1310.6240.99825.98Misfolded
K619R-kinase-deadNT1320.7830.97690.20Kinase-deficient
DNA BaseAmino AcidPubicationClinVar classificationPatient DiagnosisSIFTPolyPhen2CADDv1.6REVELAlpha MissenseFoldX: ΔΔG (kcal/mol)Integrative in silico functional prediction
44C > TP15LHadinnapola 2017VUSPAH (Het)T022.60.1330.08340.23Benign
145-2A > GY34CUK cohort (unpublished)Benign/likely-benignPVOD (C Het)T0.999320.6340.45893.79Misfolded
220G > AD74NHadinnapola 2017VUSPAH (Het)NT0.954280.1660.17260.38Benign
361G > AV121MMontani 2017VUSPVOD (C Het)T1330.4670.7125−1.04Benign
1078C > TR360CClinVar RCV001287133VUSPCHNT0.99926.30.5660.0831.68Uncertain
1321A > CI441LClinVar RCV000999785Benign/likely-benignPCHT0.00116.90.0750.0839−0.70Benign
1466C > TL489PGómez 2015VUSPAHNT1290.8950.97377.89Misfolded
1667A > GE556GClinVar RCV000755517Benign/likely-benignPCHT022.50.0880.05230.63Benign
1694 T > CI565TClinVar RCV001285279VUSPCHT0.83522.20.3070.2246−0.36Benign
1754G > AR585QEyries 2014; Montani 2017Pathogenic/likely-pathogenicPVOD; PCHNT1320.8610.92722.12Kinase-deficient
1795G > CG599RHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (Hom)NT1320.9520.99910.83Kinase-deficient
1820 T > GV607GHadinnapola 2017Pathogenic/likely-pathogenicPAH (C Het); PCHNT0.998330.5570.80394.73Misfolded
1928 T > GL643REyries 2014; Levy 2016; Montani 2017Pathogenic/likely-pathogenicPVOD; PCHNT0.999320.8890.99119.65Misfolded
2020C > TP674SClinVar RCV001286399Benign/likely-benignPCHT0.02912.10.0770.0694−1.45Benign
2516 T > CI839THadinnapola 2017VUSPAH (Het)NT0.9929.60.7810.98542.90Benign
2609C > TA870VMontani 2017VUSPVOD (C Het)T128.10.6580.99430.95Uncertain
2662G > CD888HClinVar RCV001287878Benign/likely-benignPCHNT0.99824.80.2370.10050.59Benign
2727C > GS909RHadinnapola 2017Pathogenic/likely-pathogenicPAH (Het); PCHNT1280.4460.99855.67Misfolded
2827A > GT943AHadinnapola 2017VUSPAH (C Het)NT0.99725.40.4640.85971.54Benign
2965C > TR989WEichstaedt 2022VUSPVODNT128.90.8620.983838.59Misfolded
3218G > TR1073LHadinnapola 2017VUSPAHNT0.995320.4660.33740.15Benign
3325G > AG1109RHadinnapola 2017Pathogenic/likely-pathogenicPAH (C Het); PCHNT1320.7660.98569.20Uncertain
3344C > TP1115LGómez 2015; Tenorio 2015Pathogenic/likely-pathogenicPAH; PVOD; IPAH; PCHNT129.70.7020.84151.46Uncertain
3380C > TA1127VEichstaedt 2022VUSPVODT1320.2620.60496.08Misfolded
3443 T > CL1148SEichstaedt 2022VUSCTD-APAHT128.40.3410.26481.11Benign
3604C > TH1202YHadinnapola 2017VUSPAH (Het)T0.99626.50.7160.812−0.44Benign
3605A > TH1202LHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (Hom)NT0.99828.40.8020.95073.00Uncertain
3722A > GE1241GHadinnapola 2017VUSPAHNT0.971330.3420.70691.50Benign
3884 T > GL1295RHadinnapola 2017Pathogenic/likely-pathogenicPAH; PCH (C Het)NT128.80.8140.94084.89Misfolded
3916G > TG1306CClinVar RCV000999798Benign/likely-benignPCHNT1310.3070.25962.26Uncertain
4007A > GK1336RClinVar RCV003103836Benign/likely-benignPCHT015.80.0630.05640.14Benign
4204G > TV1402LMontani 2017VUSIPAHT0.99724.70.2850.4847−0.19Benign
4646G > AR1549HHadinnapola 2017VUSPAH (Het)NT0.998320.3180.65170.30Benign
4910 T > CL1637PMontani 2017VUSPVOD (C Het)NT1310.6240.99825.98Misfolded
K619R-kinase-deadNT1320.7830.97690.20Kinase-deficient

ClinVar classification (VUS: variant of uncertain significance) and patient diagnosis where available: Hom = homozygous, Het = heterozygous, C Het = compound heterozygous. Scores SIFT (T: tolerated; NT: not tolerated), PolyPhen2, CADD v1.6, REVEL and AlphaMissense (high scores corresponding to more severe consequences). FoldX used to estimate difference in Gibbs free energy (ΔΔG, kcal/mol) between a variant and the non-mutated GCN2 (scores > 1 indicate predicted destabilisation, scores > 3 indicate predicted severe misfolding).

We applied existing computational methods including SIFT, PolyPhen2, CADDv1.6, REVEL and AlphaMissense in an effort to predict the functional significance of these GCN2 variants [26–28, 30] (Table 1). Higher scores represent higher predicted severity. We also used FoldX5.0 to estimate differences in Gibbs free energy (ΔΔG) between wildtype GCN2 and each variant (Table 1) [29]. Although broadly concordat, the algorithms yielded several discordant results. For example, although H1202Y was predicted to maintain stable folding by FoldX, and tolerated by SIFT and CADD, it was categorised as severe/likely pathogenic by the other methods. Conversely, Y34C was predicted to be destabilised by FoldX (ΔΔG > 1.5 kcal/mol) and likely-pathogenic by PolyPhen, CADD and REVEL, but tolerated by SIFT and categorised as ambiguous by AlphaMissense. Nevertheless, amid a few uncertain results, by integrating these computational methods we predicted missense variants of GCN2 to be either (i) benign, (ii) misfolded, or (iii) kinase deficient (Summarised in Table 1).

Expression of GCN2 variants and ISR reporter activation

To test the in silico predictions, we next performed experimental validation in cultured cells. A bioluminescent ISR reporter was generated by fusing the 5’UTR of ATF4 with NanoLuc® luciferase (ATF4::NanoLuc, Fig. 2A). We obtained optimal results when using human rather than murine ATF4, driven by a CMV promoter/SV40 enhancer (data not shown). uORFs in the 5’UTR of ATF4 mRNA impose translational regulation on the downstream coding sequence [21]. When endogenous GCN2 was deleted in reporter HeLa cells (GCN2 KO, Fig. S1), as expected activation of the ISR reporter by the histidyl-tRNA synthetase inhibitor histidinol was ablated (Fig. 2B). Re-expression of wild type human GCN2, but not a kinase-dead control mutant (K619R) rescued ATF4::NanoLuc responsiveness to histidinol, validating the system (Fig. 2B).

Variant in cell activity and expression. (A) ISR reporter construct comprising 5’ UTR of human ATF4 containing upstream open reading frames (uORF) cloned in frame with NanoLuc® luciferase. CMV promoter and SV40 enhancer. Under conditions of low eIF2α phosphorylation, uORF3 is translated resulting in low reporter signal. When eIF2α phosphorylation increases, ATF4 initiator AUG is translated leading to NanoLuc reporter synthesis. (B) Reporter activity (relative light units, RLU) in reporter HeLa cells without and with 3 mM histidinol 6 h. WT cells with endogenous GCN2. GCN2 KO = knockout cells lacking functional GCN2. Variants expressed in the GCN2 KO lines are indicated. K619R = kinase dead negative control. n = 3 biological replicates; 2-way ANOVA with group comparisons. (C) Representative immunoblots of GCN2 variant expression. Note—For clarity two blots are joined as indicated by vertical black lines between lanes 2&3, 3&4, and 11&12. (D) Quantification of C. n = 4, 1-way ANOVA with multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 2

Variant in cell activity and expression. (A) ISR reporter construct comprising 5’ UTR of human ATF4 containing upstream open reading frames (uORF) cloned in frame with NanoLuc® luciferase. CMV promoter and SV40 enhancer. Under conditions of low eIF2α phosphorylation, uORF3 is translated resulting in low reporter signal. When eIF2α phosphorylation increases, ATF4 initiator AUG is translated leading to NanoLuc reporter synthesis. (B) Reporter activity (relative light units, RLU) in reporter HeLa cells without and with 3 mM histidinol 6 h. WT cells with endogenous GCN2. GCN2 KO = knockout cells lacking functional GCN2. Variants expressed in the GCN2 KO lines are indicated. K619R = kinase dead negative control. n = 3 biological replicates; 2-way ANOVA with group comparisons. (C) Representative immunoblots of GCN2 variant expression. Note—For clarity two blots are joined as indicated by vertical black lines between lanes 2&3, 3&4, and 11&12. (D) Quantification of C. n = 4, 1-way ANOVA with multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Sixteen GCN2 exemplar variants were selected as representative of each class of functional prediction, across a range of disease severities and distributed throughout the GCN2 protein (highlighted in Fig. 1). Each variant was expressed in the GCN2-deleted reporter cells and bioluminescence was measured without or with histidinol treatment (Fig. 2B). We noted a striking correlation between ATF4::NanoLuc reporter activation and diagnosis (Table 2). In all but one case, when reporter activation was preserved (P15L, I839T, T943A, L1148S, H1202Y), the clinical diagnosis had been of classical PAH, rather than either PVOD or PCH. The exception was Y34C, which had preserved reporter activation despite having been identified in an individual with PVOD (that patient also had a high impact variant in their other EIF2AK4 allele: Lys190GlufsTer8, c.567dup). All other variants identified in PVOD or PCH showed impaired ATF4::NanoLuc responsiveness to histidinol (Fig. 2B).

Table 2

Variants analysis summary.

BaseAmino AcidDiagnosisATF4
Induction
Expression LevelGCN2-T899
autophosphorylation
DimerisationeIF2α-S51
phosphorylation
Classification
44C > TP15LPAH (Het)Yes0.83Yes--Benign
145-2A > GY34CPVOD (C het)Yes0.21Yes--Destabilised
1754G > AR585QPVOD; PCHNo1.09YesYesYes (32.6%)Hypomorphic
1820 T > GV607GPAH; PCHNo0.71YesYesYes (42.8%)Hypomorphic
1928 T > GL643RPVOD; PCHNo0.26NoYesNoKinase-dead
2516 T > CI839TPAH (Het)Yes0.85Yes--Benign
2609C > TA870VPVOD (C het)No1.00No--Kinase-dead
2727C > GS909RPAH; PCHNo0.39NoYes-Kinase-dead
2827A > GT943APAHYes1.06Yes--Benign
2965C > TR989WPVODNo0.09No--Misfolded
3325G > AG1109RPAH; PCHNo0.68Yes-Yes (27.2%)Hypomorphic
3344C > TP1115LPAH; PVOD; IPAH; PCHNo0.76Yes-Yes (12.9%)Hypomorphic
3443 T > CL1148SCTD-APAHYes0.99Yes--Benign
3604C > TH1202YPAHYes0.14Yes--Benign
3605A > TH1202LPAH; PCH (Hom)No0.12Yes-NoMisfolded
3884 T > GL1295RPAH; PCH (C Het)No0.10No-NoMisfolded
K619Rkinase-deadNo1.17No-NoKinase-dead
BaseAmino AcidDiagnosisATF4
Induction
Expression LevelGCN2-T899
autophosphorylation
DimerisationeIF2α-S51
phosphorylation
Classification
44C > TP15LPAH (Het)Yes0.83Yes--Benign
145-2A > GY34CPVOD (C het)Yes0.21Yes--Destabilised
1754G > AR585QPVOD; PCHNo1.09YesYesYes (32.6%)Hypomorphic
1820 T > GV607GPAH; PCHNo0.71YesYesYes (42.8%)Hypomorphic
1928 T > GL643RPVOD; PCHNo0.26NoYesNoKinase-dead
2516 T > CI839TPAH (Het)Yes0.85Yes--Benign
2609C > TA870VPVOD (C het)No1.00No--Kinase-dead
2727C > GS909RPAH; PCHNo0.39NoYes-Kinase-dead
2827A > GT943APAHYes1.06Yes--Benign
2965C > TR989WPVODNo0.09No--Misfolded
3325G > AG1109RPAH; PCHNo0.68Yes-Yes (27.2%)Hypomorphic
3344C > TP1115LPAH; PVOD; IPAH; PCHNo0.76Yes-Yes (12.9%)Hypomorphic
3443 T > CL1148SCTD-APAHYes0.99Yes--Benign
3604C > TH1202YPAHYes0.14Yes--Benign
3605A > TH1202LPAH; PCH (Hom)No0.12Yes-NoMisfolded
3884 T > GL1295RPAH; PCH (C Het)No0.10No-NoMisfolded
K619Rkinase-deadNo1.17No-NoKinase-dead

ATF4 induction and protein expression from Fig. 2. T899 autophosphorylation from Fig. 3. Dimerisation from Fig. 5. In vitro eIF2α phosphorylation from Fig. 6.

Table 2

Variants analysis summary.

BaseAmino AcidDiagnosisATF4
Induction
Expression LevelGCN2-T899
autophosphorylation
DimerisationeIF2α-S51
phosphorylation
Classification
44C > TP15LPAH (Het)Yes0.83Yes--Benign
145-2A > GY34CPVOD (C het)Yes0.21Yes--Destabilised
1754G > AR585QPVOD; PCHNo1.09YesYesYes (32.6%)Hypomorphic
1820 T > GV607GPAH; PCHNo0.71YesYesYes (42.8%)Hypomorphic
1928 T > GL643RPVOD; PCHNo0.26NoYesNoKinase-dead
2516 T > CI839TPAH (Het)Yes0.85Yes--Benign
2609C > TA870VPVOD (C het)No1.00No--Kinase-dead
2727C > GS909RPAH; PCHNo0.39NoYes-Kinase-dead
2827A > GT943APAHYes1.06Yes--Benign
2965C > TR989WPVODNo0.09No--Misfolded
3325G > AG1109RPAH; PCHNo0.68Yes-Yes (27.2%)Hypomorphic
3344C > TP1115LPAH; PVOD; IPAH; PCHNo0.76Yes-Yes (12.9%)Hypomorphic
3443 T > CL1148SCTD-APAHYes0.99Yes--Benign
3604C > TH1202YPAHYes0.14Yes--Benign
3605A > TH1202LPAH; PCH (Hom)No0.12Yes-NoMisfolded
3884 T > GL1295RPAH; PCH (C Het)No0.10No-NoMisfolded
K619Rkinase-deadNo1.17No-NoKinase-dead
BaseAmino AcidDiagnosisATF4
Induction
Expression LevelGCN2-T899
autophosphorylation
DimerisationeIF2α-S51
phosphorylation
Classification
44C > TP15LPAH (Het)Yes0.83Yes--Benign
145-2A > GY34CPVOD (C het)Yes0.21Yes--Destabilised
1754G > AR585QPVOD; PCHNo1.09YesYesYes (32.6%)Hypomorphic
1820 T > GV607GPAH; PCHNo0.71YesYesYes (42.8%)Hypomorphic
1928 T > GL643RPVOD; PCHNo0.26NoYesNoKinase-dead
2516 T > CI839TPAH (Het)Yes0.85Yes--Benign
2609C > TA870VPVOD (C het)No1.00No--Kinase-dead
2727C > GS909RPAH; PCHNo0.39NoYes-Kinase-dead
2827A > GT943APAHYes1.06Yes--Benign
2965C > TR989WPVODNo0.09No--Misfolded
3325G > AG1109RPAH; PCHNo0.68Yes-Yes (27.2%)Hypomorphic
3344C > TP1115LPAH; PVOD; IPAH; PCHNo0.76Yes-Yes (12.9%)Hypomorphic
3443 T > CL1148SCTD-APAHYes0.99Yes--Benign
3604C > TH1202YPAHYes0.14Yes--Benign
3605A > TH1202LPAH; PCH (Hom)No0.12Yes-NoMisfolded
3884 T > GL1295RPAH; PCH (C Het)No0.10No-NoMisfolded
K619Rkinase-deadNo1.17No-NoKinase-dead

ATF4 induction and protein expression from Fig. 2. T899 autophosphorylation from Fig. 3. Dimerisation from Fig. 5. In vitro eIF2α phosphorylation from Fig. 6.

Next, protein expression of the sixteen GCN2 variants was determined by immunoblotting (Fig. 2C-D). Significantly reduced expression relative to wildtype GCN2 was observed for eight variants (Y34C, V607G, L643R, S909R, R989W, H1202Y, H1202L, L1295R). Of these, despite their low expression, two retained significant ATF4::NanoLuc reporter activation (Y34C and H1202Y). Conversely, three of these naturally occurring variants lacked reporter activity despite preserved expression (R585Q, G1109R and P1115L), similar to that seen for the artificial kinase-dead control (K619R). Of note, V607G showed no reporter activation despite only modestly reduced expression. Expression levels of GCN2 variants therefore do not fully reflect activity.

GCN2 autophosphorylation is necessary but not sufficient for ISR induction

Autophosphorylation of GCN2 at T899 correlates with kinase activation in most circumstances [32, 35]. Treatment with histidinol or starvation of amino acids, a physiological stimulus of the kinase, increased T899 phosphorylation of wildtype but not kinase-dead K619R GCN2 expressed in knockout cells (Fig. 3A-B & S1B-C). When the naturally occurring variants were tested, eleven were capable of T899 autophosphorylation, while five were not (L643R, A870V, S909R, R989W, L1295R) (Fig. 3C-F, summarised in Table 2). These results show that stressful stimuli triggered T899 autophosphorylation in all GCN2 variants capable of activating the ATF4::NanoLuc reporter, but also in five ISR-deficient variants (R585Q, V607G, G1109R, P1115L, H1202L) albeit only weakly (Figs. 3C-F, summarised in Table 2).

Stress-dependent activation of GCN2. (A) Representative immunoblot of GCN2 KO HeLa cells transiently transfected with constructs encoding full-length human GCN2 (hGCN2) or kinase-dead negative control (K619R). Hygromycin-selected pools were treated with either 7 mM histidinol or starved of methionine and leucine (−met -Leu) for 7 h. (B). Quantifications of a. n = 4 biological replicates normalised to untreated, hGCN2-transfected (lane 4). 1-way ANOVA with multiple comparisons (meaningful comparisons are shown). *P < 0.05, **P < 0.01. (C-F) representative immunoblots in GCN2 KO HeLa cells transiently transfected with GCN2 variants treat as in a. n = 3 biological replicates.
Figure 3

Stress-dependent activation of GCN2. (A) Representative immunoblot of GCN2 KO HeLa cells transiently transfected with constructs encoding full-length human GCN2 (hGCN2) or kinase-dead negative control (K619R). Hygromycin-selected pools were treated with either 7 mM histidinol or starved of methionine and leucine (−met -Leu) for 7 h. (B). Quantifications of a. n = 4 biological replicates normalised to untreated, hGCN2-transfected (lane 4). 1-way ANOVA with multiple comparisons (meaningful comparisons are shown). *P < 0.05, **P < 0.01. (C-F) representative immunoblots in GCN2 KO HeLa cells transiently transfected with GCN2 variants treat as in a. n = 3 biological replicates.

These results suggested that GCN2 autophosphorylation at T899 is necessary but not sufficient for ISR induction. From a structural perspective, the A870 (Fig. 4, in purple) is located on the kinase activation loop, in close proximity to residue K619 (in orange) of the N-lobe, essential for kinase activity (K619R yields a dead kinase). Mutations in these key residues led to total loss of autophosphorylation and kinase activity. L643, S909 and R989 (Fig. 4, in magenta) are essential residues for the tight packing of the kinase domain C-lobe. L643R, S909R, R989W introduce larger and differently charged sidechains into the hinge region or the C-lobe of the kinase, disrupting the local protein structure required for kinase activity. On the other hand, I839 and T943 (Fig. 4, in blue) are located at the edge of the kinase helix. The I839T and T943A variants do not introduce larger sidechains nor disrupt local packing, hence retaining activity.

Structure of GCN2 kinase domain with mapped residues mutated in patients. Human GCN2 kinase domain structure (from PDB accession: 7QWK)41 highlighting the location of residues mutated in PAH, as well as K619 (in orange) mutated in the kinase-dead control. Mutations that led to the loss of kinase activity include A870V, located in the activation loop (in purple), L643R in the hinge region (in magenta), S909R and R989W which are in the core of the C-lobe (in magenta). Variants I839T and T943A (in cyan), localised at the edge of the kinase helix without introducing bigger sidechains, did not affect folding and retained kinase activity. Variants R585Q and V607G localised in the N-lobe (in green) were hypomorphic. Bound ATP is shown in yellow.
Figure 4

Structure of GCN2 kinase domain with mapped residues mutated in patients. Human GCN2 kinase domain structure (from PDB accession: 7QWK)41 highlighting the location of residues mutated in PAH, as well as K619 (in orange) mutated in the kinase-dead control. Mutations that led to the loss of kinase activity include A870V, located in the activation loop (in purple), L643R in the hinge region (in magenta), S909R and R989W which are in the core of the C-lobe (in magenta). Variants I839T and T943A (in cyan), localised at the edge of the kinase helix without introducing bigger sidechains, did not affect folding and retained kinase activity. Variants R585Q and V607G localised in the N-lobe (in green) were hypomorphic. Bound ATP is shown in yellow.

Inactive GCN2 exists as an antiparallel homodimer and transitions to a parallel conformation on activation [36, 37]. In yeast, stabilisation of the active state depends on the establishment of an intramolecular salt-bridge in the active conformation between residues R594 and D598, corresponding to R585 and E589 in the human protein [38] (Fig. S2). The patient-derived R585Q and V607G variants, localised in the kinase domain N-lobe (Fig. 4, in green) were unable to activate the ATF4::NanoLuc reporter despite preserved expression (Fig. 2) and autophosphorylation (Fig. 3). Since the R585Q substitution is predicted to reposition the dimerisation interface salt-bridge required for activation, we sought to test if dimerisation was impaired. GCN2 constructs were generated tagged at the C-terminus with either 3xFlag or V5. Tagging did not impair GCN2 activity (Fig. 5A). When co-expressed in GCN2 deleted cells, wildtype GCN2-3xFlag and GCN2-V5 formed mixed dimers detectable by anti-FLAG co-immunoprecipitation (Fig. 5B). The R585Q variant similarly formed mixed dimers that could be co-immunoprecipitated (Fig. 5C). The ISR-deficient kinase domain variants V607G, L643R, and S909R, predicted in silico to affect the folding of the kinase domain (Table 1), were similarly able to form mixed dimers, suggesting that, at least for these variants, loss of dimerisation does not contribute to their impaired function (Fig. 5C-F).

Dimerisation of variants of GCN2 variants. (A) Representative immunoblot of GCN2 KO HeLa cells transiently transfected with construct encoding 3xFlag tagged human GCN2 (hGCN2_3xFLAG) treated with either 7 mM histidinol or starved of methionine and leucine (−met -Leu) for 7 h. n = 3 biological replicates (B-F) anti-FLAG immunoprecipitation-immunoblots from GCN2 KO cells transiently transfected with constructs encoding GCN2 variants tagged at the C-terminus with either 3xFLAG (GCN2_3xFLAG) or V5 (GCN2_V5). Lysates (lanes 1–3) and immunoprecipitates (lanes 4–6). Note co-immunoprecipitation of each pair of constructs consistent with dimer formation.
Figure 5

Dimerisation of variants of GCN2 variants. (A) Representative immunoblot of GCN2 KO HeLa cells transiently transfected with construct encoding 3xFlag tagged human GCN2 (hGCN2_3xFLAG) treated with either 7 mM histidinol or starved of methionine and leucine (−met -Leu) for 7 h. n = 3 biological replicates (B-F) anti-FLAG immunoprecipitation-immunoblots from GCN2 KO cells transiently transfected with constructs encoding GCN2 variants tagged at the C-terminus with either 3xFLAG (GCN2_3xFLAG) or V5 (GCN2_V5). Lysates (lanes 1–3) and immunoprecipitates (lanes 4–6). Note co-immunoprecipitation of each pair of constructs consistent with dimer formation.

Hypomorphic GCN2 variants can be activated by an ATP-competitive inhibitor

We then sought to test whether autophosphorylation-competent, but ISR-deficient variants might have lost the ability to engage the substrate eIF2α. Tagged GCN2 was recovered by immunoprecipitation of 3xFlag from lysates of cells starved of amino acids (Fig. 6A). Immunopurified kinases were then tested for their ability to phosphorylate the N-terminal domain of eIF2α (eIF2α-NTD) in vitro (Fig. 6B-D). Wildtype but not kinase-dead K619R GCN2 showed enhanced T899 autophosphorylation when incubated with Mg-ATP, leading to increased phospho-GCN2 immunoreactivity and slower migration on SDS-PAGE (Fig. 6B-D). When incubated with Mg-ATP and eIF2α-NTD, wildtype but not kinase dead K619R GCN2 phosphorylated eIF2α-NTD on serine 51 (Fig. 6C-D). The autophosphorylation-competent variants R585Q, V607G, G1109R and P1115L, but not the kinase-dead S909R (Figs. 3C,E and 6B) also autophosphorylated their activation loop at T899 when incubated with Mg-ATP in vitro, and phosphorylated eIF2α-NTD albeit only weakly (Fig. 6C-E). The variants L643R, S909R, H1202L and L1295R, which showed either weak or no autophosphorylation combined with low expression (Fig. 6A,C) failed to phosphorylate eIF2α-NTD (Fig. 6C-D). These data suggest that while some PVOD/PCH-associated variants preserve target engagement in vitro, their weak kinase activity appears insufficient for downstream signalling in cells. We classified these variants as hypomorphic.

In vitro GCN2 kinase assay. (A) Representative immunoprecipitation-immunoblots from cells expressing 3xFlag tagged GCN2. Cells were starved of arginine and lysine for 6 h before harvesting. Intact GCN2 was eluted by competition via addition of excess FLAG peptide. Phosphorylation on threonine 899 reports autophosphorylation, note its absence for the K619R kinase-dead control. Note—For clarity two blots are joined as indicated by vertical black lines between 6&7. (B) Schematic of in vitro kinase assay drawn using Biorender.com. Tagged protein immunopurified using anti-FLAG beads then eluted with FLAG peptide. Bacterially expressed recombinant eIF2α-N-terminal domain (NTD) served as a specific substrate. (C). Representative immunoblots of reaction products. Note—For clarity two blots are joined as indicated by vertical black lines between lanes 12&13. (D) Quantification of C; data presented with median value. n = 4; 1-way ANOVA, compared to hGCN2-transfected control. ****P < 0.0001.
Figure 6

In vitro GCN2 kinase assay. (A) Representative immunoprecipitation-immunoblots from cells expressing 3xFlag tagged GCN2. Cells were starved of arginine and lysine for 6 h before harvesting. Intact GCN2 was eluted by competition via addition of excess FLAG peptide. Phosphorylation on threonine 899 reports autophosphorylation, note its absence for the K619R kinase-dead control. Note—For clarity two blots are joined as indicated by vertical black lines between 6&7. (B) Schematic of in vitro kinase assay drawn using Biorender.com. Tagged protein immunopurified using anti-FLAG beads then eluted with FLAG peptide. Bacterially expressed recombinant eIF2α-N-terminal domain (NTD) served as a specific substrate. (C). Representative immunoblots of reaction products. Note—For clarity two blots are joined as indicated by vertical black lines between lanes 12&13. (D) Quantification of C; data presented with median value. n = 4; 1-way ANOVA, compared to hGCN2-transfected control. ****P < 0.0001.

It was recently shown that GCN2 can be activated paradoxically by sub-inhibitory concentrations of ATP-competitive kinase inhibitors [39, 40]. Carlson et al. showed activity of GCN2-R585Q, one of the hypomorphic variants identified here, though not the L643R variant, by treatment with the type-1½ kinase inhibitor Gcn2iB [32]. We therefore tested the ability of such a small molecule, to activate the ATF4::NanoLuc reporter in cells expressing PAH-associated GCN2 variants. We found that all the identified hypomorphs (R585Q, V607G, G1109R and P1115L) were rescued in their reporter activation by Gcn2iB, while misfolded or kinase-dead variants were not (K619R, L643R and S909R; Fig. 7).

Activity of hypomorphic variants of GCN2 treated with Gcn2iB. ATF4::nanoLuc ISR reporter activity in GCN2 KO cells transiently transfected with constructions wild-type (hGCN2), kinase-dead (K619R), or selected patient variants. Cells were treated with 25 nM Gcn2iB for 6 h before luciferase assay. n = 3 biological replicates; 2-way ANOVA with group comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 7

Activity of hypomorphic variants of GCN2 treated with Gcn2iB. ATF4::nanoLuc ISR reporter activity in GCN2 KO cells transiently transfected with constructions wild-type (hGCN2), kinase-dead (K619R), or selected patient variants. Cells were treated with 25 nM Gcn2iB for 6 h before luciferase assay. n = 3 biological replicates; 2-way ANOVA with group comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Discussion

Detection of biallelic pathogenic EIF2AK4 mutations establishes the diagnosis of PVOD or PCH without the need for histological confirmation [6]. Validating the pathogenicity of EIF2AK4 variants is therefore of significant diagnostic value, which is important since therapies developed for classical PAH can be detrimental in these rare subtypes [7]. Segregation studies, although the gold standard, are not always feasible. By contrast, genetic testing is routinely used in clinical practice. We found that integrative computational analysis failed to identify some ISR-defective variants of GCN2. However, our in cellulo assay using a sensitive ATF4::nanoLuc reporter cell line could reliably identify pathogenic variants of GCN2. This approach is simple and inexpensive, making it a feasible addition to characterisation workflows in specialist clinical practice.

We showcased this approach by evaluating sixteen of the thirty-four known missense variants of GCN2 associated with PAH. Our observation that variants identified by genomic sequencing of individuals with classical PAH had preserved ISR reporter activity is consistent with GCN2 playing only a minor, or even no, role in that condition. Conversely, variants identified in individuals with either PVOD or PCH showed loss of ISR functionality, underlining the key role played by GCN2 in these disorders. The Y34C variant was a notable exception, maintaining some ISR reporter activity despite having been identified in an individual with PVOD. Of note, that patient also harboured a high-impact mutation of their second EIF2AK4 allele. In our study, GCN2 Y34C was expressed at a significantly reduced level compared to the wild-type protein, raising the possibility that when combined with a null allele, the level of GCN2 generated might be insufficient to prevent development of the disease.

Importantly, we identified a subset of GCN2 variants with preserved target engagement but reduced kinase activity. R585Q and V607G locate to the kinase domain N-lobe which is dominated by an anti-parallel beta-sheet and contains most of the residues involved in ATP binding. This portion of the kinase also participates in dimerisation and stabilisation of the active parallel conformation [41]. Though our data exclude lack of dimerisation, it remains possible that mutants in the N-lobe could affect dimer activation. Conversely, G1109 and P1115, corresponding to G1085 and Q1091 in yeast, have been shown to be involved in tRNA binding [37]. G1109R and P1115L mutations change mainchain flexibility and introduce bulky sidechains, likely to alter such interaction. Strikingly, these hypomorphs could be activated by the ATP-competitive inhibitor Gcn2iB, a type 1½ inhibitor that stabilises the enzyme in a non-productive, yet active-like conformation (DFG-in, αC helix-out). It is believed that such binding to one protomer of a GCN2 dimer causes activation of the second drug-free promoter [39]. This suggests a potential therapeutic strategy for individuals with such hypomorphic variants. Hypomorphs can be identified by our ATF4::nanoLuc reporter assay by treatment with Gcn2iB.

The experimental validation of GCN2 variants enabled us to evaluate computational predictive methods. It is recognised that evolutionary-based methods relying on homologous sequence alignment (e.g. SIFT, MAPP, PANTHER) are outperformed as stand-alone tools by approaches that integrate additional information such as structural features [42]. We compared the integrative tools PolyPhen2 and CADD and found that out of sixteen variants they incorrectly predicted enzyme activity in 5 and 6 instances respectively, giving a positive predictive value of approximately only 60%. Indeed, functional predictions using these methods are not recommended for diagnostic purposes for this reason [43]. Accuracy was improved with machine learning-based approaches, but even these rarely exceed 80% accuracy [44]. It has been estimated that 75% of disease-causing variants are linked to protein destabilisation [45]. The computation of Gibbs free energy variations with FoldX was recently reported as the best performing method for the identification of disease-causing mutations [46]. FoldX predicted protein expression levels of GCN2 missense variants in our cell system, but the existence of stable but kinase-dead or hypomorphic variants limits its value. Nevertheless, integrating conservation-based approaches with structural features improves predictive performance. Meta-predictors, such as the ensemble method REVEL [28], are better correlated with benchmark clinical datasets like ClinVar (reviewed in reference (47)). Although better than other tools, REVEL only returned 75% accuracy in our study. Recently AlphaMissense was developed, a deep-learning AlphaFold-derived system that combines residue structural context with unsupervised modelling of evolutionary constraints by comparing related sequences, claiming 90% accuracy in predicting the pathogenicity of missense variants when tested against the ClinVar dataset [31]. When using ISR-reporter activation as our gold standard, AlphaMissense incorrectly assigned 3 of the 16 GCN2 variants examined. The complexity of modelling protein–protein and protein-ribosome interactions adds to the challenge of using computational approaches to classify GCN2 variants [16, 35].

In summary, in cellulo evaluation of ISR signalling using an ATF4::Nanoluc reporter outperformed existing computational approaches. This approach can not only identify pathogenic variants but can also recognise hypomorphs that can be revitalised by an ATP-pocket-binding small molecule drug.

Materials and methods

Cloning and plasmids

All cloning and mutagenesis were performed via Gibson Assembly. A human codon-optimised GCN2 ORF was cloned into pcDNA3.1-Hygro(+) vectors and tagged with either 3xFLAG or V5 at the C-terminus. Site-directed mutagenesis was performed with specific primers for 16 naturally occurring variants and a kinase-dead control. A human ATF4 5’UTR ORF was inserted into pGL4.2 vectors and cloned in frame with Nluc-PEST® luciferase. A stop codon was inserted before the C-terminal degron to allow accumulation of the reporter.

CRISPR/Cas9 knockout of EIF2AK4 in HeLa cells

Human EIF2AK4 specific guide RNAs were selected from the Brunello sgRNA library [48]. After primer duplex formation, guides were inserted in pSpCas9(BB)-2A-mCherry plasmids. Parental HeLa cells, cultured in DMEM supplemented with 10% FBS were transfected in 6-well dishes using 1 μg DNA and Lipofectamine 2000 (1:3 ratio) in OptiMEM for 24 h. At day 3 post-transfection mCherry-positive cells were sorted on a DB Melody cell sorter, as single cells into 96 well plates. Clones were screened by GCN2-targeting western blotting. Genetic mutations in KO clones were confirmed by genomic DNA extraction (100 mM Tris, 5 mM EDTA, 200 mM NaCl, 0.25% SDS, 0.2 mg/ml Proteinase K: incubation at 50°C overnight, then for 20 minutes at 98°C and clarification in a benchtop centrifuge at 10 000 g for 5 minutes), PCR to amplify the locus targeted by the guide and subsequent NGS. Data were analysed using MacVector.

Transfection and cell treatments

2×105 HeLa cells were plated in 6-well dishes and let attach overnight before transfection. 1 μg of plasmid DNA was mixed with Lipofectamine 2000 (1:3 ratio) in OptiMEM and incubated for 20 minutes at room temperature. 500 μL of transfection medium were added onto the washed cells and topped up with additional 500 μL of 10% FBS-DMEM. Transfection medium was removed after 24 h. On day 2 cells were split and transfected cells were selected by 300 μg/ml hygromycin treatment for 3 days. Cells were then maintained in 150 μg/ml hygromycin 10% FBS-DMEM. Cell treatments were carried out as follows: 7 mM histidinol for 7 h (for western blotting); amino acid starvation in SILAC medium supplemented with 10% dialysed FBS and 25 mM D-glucose (after 1× wash in PBS) without leucine only, leucine and methionine or lysine and arginine, for 7 h.

Immunoblotting

Cells were washed with PBS on ice and lysed in low-salt buffer (Buffer H: 10 mM HEPES pH 7.9, 50 mM NaCl, 0.5 M sucrose, 0.1 mM EDTA, 0.5% TX-100, 1 mM DTT, 10ul/ml cOmplete™ Protease Inhibitor Cocktail (Roche, SKU 11836170001), phosphatase inhibitor cocktail mix (10 mM tetrasodium pyrophosphate, 17.5 mM beta glycerophosphate, 100 mM sodium fluoride), 1 mM PMSF) and 10 mM DTT. Samples were clarified at 4°C in a benchtop centrifuge at 10′000 g for 10 minutes. Protein concentrations were estimated using Pierce BCA assay following the manufacturer’s instructions and equalised in lysis buffer and 6X loading buffer (1X: 60 mM Tris–HCl pH 6.8, 10% glycerol, 2% SDS, 0.02% bromophenol blue, 1 mM DTT). 8–14% polyacrylamide, 0.4% SDS gels were casted for SDS-PAGE. 60-80 μg of protein were loaded per lane. Proteins were transferred to 25 μm nitrocellulose membranes using 5% methanol transfer buffer. Blocking was done in 5% BSA in TBS as well as incubations with antibodies. Antibodies: GCN2 (in house, Ron lab: NY168); GCN2-phosphorT899 (Abcam, ab75836); β-actin (Abcam, ab8226); FLAG (Invitrogen, 740 001); V5 (Abcam, ab27671).

NanoGlo® luciferase assay

GCN2 KO, ATF4::NanoLuc reporter cells were transfected with GCN2 variant constructs as described above. Cells were seeded in 384-well plates at a density of 2.5×103 cells per well in 20 μL. Left-over cells were pelleted and lysed to confirm transfection efficiency via western blot. Wells were previously filled with 5 μL of 15 mM histidinol (5X) in 10% FBS-DMEM (final concentration 3 mM), or 125 nM (5X) Gcn2iB (final concentration 25 nM). After 6 h of incubation at 37°C and 5% CO2, 25 μL of NanoGlo® luciferase assay reagent (buffer + substrate, 50:1) were added to the wells. Plates were mixed with orbital shaking for 1 minute and after 10 minutes of incubation at room temperature, bioluminescence signal (360-545 nm) was acquired on a Tecan Spark plate reader.

Dimerisation assay

GCN2 KO HeLa cells were transfected with 3xFLAG and/or V5 tagged GCN2 variant constructs as described above. After selection, cells were grown to confluence in 6 cm dishes. Dishes were transferred on ice, washed once with PBS and lysed in 50 mM Tris, pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% Triton-X, plus protease and phosphatase inhibitors. Samples were clarified by centrifugation (> 10 000 g at 4°C for 10 minutes) and transferred in clean tubes on ice. Protein concentrations were measured using Bradford assay and equalised with lysis buffer. 80 μg of protein samples were set aside as input controls, 600-800 μg of protein were diluted in 200 μL and added to 50 μL of anti-FLAG® M2 affinity gel slurry (Millipore, A2220). Samples were incubated for 1 h rotating at 4°C. Three washes were performed via centrifugation (1000 rpm) using 1 mL of lysis buffer without Triton-X. Elution was performed in 30 μL of wash buffer plus 5 μL of 1 mg/ml 3xFLAG peptide solution (Pierce™ 3x DYKDDDDK, Thermo Scientific, A36805). Samples were incubated on ice for 15 minutes. Beads were pelleted by centrifugation (1000 rpm). 15 μL of eluate were used for western blotting.

In vitro kinase assay

GCN2 KO HeLa cells were transfected with 3xFLAG GCN2 variant constructs as described above. After selection, cells were grown to confluence in 6 cm dishes. Cells were washed once with PBS and incubated with amino acid depleted (lysine and arginine) SILAC medium supplemented with 10% dialysed FBS and 25 mM D-glucose, for 6 h. After immunoprecipitation (as described above) 10 μL of eluate per reaction were added to PCR strip tubes or plates that were previously coated with 100 mg/ml BSA for 3 h, then washed 3X with PBS and dried thoroughly. Kinase assay was performed by adding recombinant eIF2α-NTD, amino acids 2–187 (kindly donated by the Ron lab) to a final concentration on 1 μM and 500 μM ATP in reaction buffer (1X: 50 mM HEPES, pH 7.4, 100 mM potassium acetate, 5 mM magnesium acetate, 250 μg/ml BSA, 10 mM magnesium chloride, 5 mM DTT, 5 mM β-glycerophosphate). Reactions were incubated at 32°C for 10 minutes, then immediately quenched with 5 μL of 6X SDS sample buffer for western blotting, pre-warmed at 95°C.

Funding

SJM was supported by the MRC (MCMB MR/V028669/1 and MR/R009120/1), EPSRC (EP/R03558X/1), Cambridge Biomedical Research Centre (BRC-1215-20 014); British Lung Foundation (BLF), Asthma+Lung UK (ALUK), Royal Papworth Hospital, and the Victor Philip Dahdaleh Foundation. GE were supported by philanthropic funding from Rick Medlock and by awards from EPSRC (EP/R03558X/1), BHF (RE/18/1/34212), Evelyn Trust (Grant 22/03), and the Cambridge-Tsinghua collaborative programme on sustainability and emerging technologies. JZ Doctoral Training Programme in Medical Research (DTP-MR; Cambridge Trust). This research was supported by the CIMR Flow Cytometry Core Facility. We wish to thank Reiner Schulte and Gabriela Grondys-Kotarba for their advice and support in flow cytometry cell sorting. We thank David Ron and Heather Harding for valuable discussions, advice, and reagents. We thank Glenn Masson for sharing reagents. W.L. is funded by a British Heart Foundation Senior Basic Science Research Fellowship (FS/SBSRF/20/31005).

References

1.

Machado
 
RD
,
Pauciulo
 
MW
,
Thomson
 
JR
. et al.  
BMPR2 haploinsufficiency as the inherited molecular mechanism for primary pulmonary hypertension
.
Am J Hum Genet
 
2001
;
68
:
92
102
.

2.

Deng
 
Z
,
Morse
 
JH
,
Slager
 
SL
. et al.  
Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene
.
Am J Hum Genet
 
2000
;
67
:
737
744
.

3.

International PPHC
,
Lane
 
KB
,
Machado
 
RD
. et al.  
Heterozygous germline mutations in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension
.
Nat Genet
 
2000
;
26
:
81
84
.

4.

Eichstaedt
 
CA
,
Song
 
J
,
Benjamin
 
N
. et al.  
EIF2AK4 mutation as "second hit" in hereditary pulmonary arterial hypertension
.
Respir Res
 
2016
;
17
:
141
.

5.

Morrell
 
NW
,
Aldred
 
MA
,
Chung
 
WK
. et al.  
Genetics and genomics of pulmonary arterial hypertension
.
Eur Respir J
 
2019
;
53
:
1801899
.

6.

Humbert
 
M
,
Kovacs
 
G
,
Hoeper
 
MM
. et al.  
2022 ESC/ERS guidelines for the diagnosis and treatment of pulmonary hypertension
.
G Ital Cardiol (Rome)
 
2023
;
24
:
1e
116e
.

7.

Holcomb
 
BW
 Jr,
Loyd
 
JE
,
Ely
 
EW
. et al.  
Pulmonary veno-occlusive disease: a case series and new observations
.
Chest
 
2000
;
118
:
1671
1679
.

8.

Eyries
 
M
,
Montani
 
D
,
Girerd
 
B
. et al.  
EIF2AK4 mutations cause pulmonary veno-occlusive disease, a recessive form of pulmonary hypertension
.
Nat Genet
 
2014
;
46
:
65
69
.

9.

Tenorio
 
J
,
Navas
 
P
,
Barrios
 
E
. et al.  
A founder EIF2AK4 mutation causes an aggressive form of pulmonary arterial hypertension in Iberian gypsies
.
Clin Genet
 
2015
;
88
:
579
583
.

10.

Gomez
 
J
,
Reguero
 
JR
,
Alvarez
 
C
. et al.  
A semiconductor Chip-based next generation sequencing procedure for the main pulmonary hypertension genes
.
Lung
 
2015
;
193
:
571
574
.

11.

Levy
 
M
,
Eyries
 
M
,
Szezepanski
 
I
. et al.  
Genetic analyses in a cohort of children with pulmonary hypertension
.
Eur Respir J
 
2016
;
48
:
1118
1126
.

12.

Hadinnapola
 
C
,
Bleda
 
M
,
Haimel
 
M
. et al.  
Phenotypic characterization of EIF2AK4 mutation carriers in a large cohort of patients diagnosed clinically with pulmonary arterial hypertension
.
Circulation
 
2017
;
136
:
2022
2033
.

13.

Montani
 
D
,
Girerd
 
B
,
Jais
 
X
. et al.  
Clinical phenotypes and outcomes of heritable and sporadic pulmonary veno-occlusive disease: a population-based study
.
Lancet Respir Med
 
2017
;
5
:
125
134
.

14.

Eichstaedt
 
CA
,
Sassmannshausen
 
Z
,
Shaukat
 
M
. et al.  
Gene panel diagnostics reveals new pathogenic variants in pulmonary arterial hypertension
.
Respir Res
 
2022
;
23
:
74
.

15.

Emanuelli
 
G
,
Nassehzadeh-Tabriz
 
N
,
Morrell
 
NW
. et al.  
The integrated stress response in pulmonary disease
.
Eur Respir Rev
 
2020
;
29
:
200184
.

16.

Pochopien
 
AA
,
Beckert
 
B
,
Kasvandik
 
S
. et al.  
Structure of Gcn1 bound to stalled and colliding 80S ribosomes
.
Proc Natl Acad Sci USA
 
2021
;
118
(14):e2022756118. https://doi.org/10.1073/pnas.2022756118.

17.

Wu
 
CC
,
Peterson
 
A
,
Zinshteyn
 
B
. et al.  
Ribosome collisions trigger general stress responses to regulate cell fate
.
Cell
 
2020
;
182
:
404
416 e414
.

18.

Masson
 
GR
.
Towards a model of GCN2 activation
.
Biochem Soc Trans
 
2019
;
47
:
1481
1488
.

19.

Cordova
 
RA
,
Misra
 
J
,
Amin
 
PH
. et al.  
GCN2 eIF2 kinase promotes prostate cancer by maintaining amino acid homeostasis
.
elife
 
2022
;
11
:e81083. https://doi.org/10.7554/eLife.81083.

20.

Harding
 
HP
,
Zhang
 
Y
,
Zeng
 
H
. et al.  
An integrated stress response regulates amino acid metabolism and resistance to oxidative stress
.
Mol Cell
 
2003
;
11
:
619
633
.

21.

Young
 
SK
,
Wek
 
RC
.
Upstream open reading frames differentially regulate gene-specific translation in the integrated stress response
.
J Biol Chem
 
2016
;
291
:
16927
16935
.

22.

Yan
 
Y
,
Harding
 
HP
,
Ron
 
D
.
Higher-order phosphatase-substrate contacts terminate the integrated stress response
.
Nat Struct Mol Biol
 
2021
;
28
:
835
846
.

23.

van der
 
Knaap
 
MS
,
Leegwater
 
PA
,
Konst
 
AA
. et al.  
Mutations in each of the five subunits of translation initiation factor eIF2B can cause leukoencephalopathy with vanishing white matter
.
Ann Neurol
 
2002
;
51
:
264
270
.

24.

Abdulkarim
 
B
,
Nicolino
 
M
,
Igoillo-Esteve
 
M
. et al.  
A missense mutation in PPP1R15B causes a syndrome including diabetes, short stature, and microcephaly
.
Diabetes
 
2015
;
64
:
3951
3962
.

25.

Delepine
 
M
,
Nicolino
 
M
,
Barrett
 
T
. et al.  
EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome
.
Nat Genet
 
2000
;
25
:
406
409
.

26.

Flanagan
 
SE
,
Patch
 
AM
,
Ellard
 
S
.
Using SIFT and poly Phen to predict loss-of-function and gain-of-function mutations
.
Genet Test Mol Biomarkers
 
2010
;
14
:
533
537
.

27.

Rentzsch
 
P
,
Witten
 
D
,
Cooper
 
GM
. et al.  
CADD: predicting the deleteriousness of variants throughout the human genome
.
Nucleic Acids Res
 
2019
;
47
:
D886
D894
.

28.

Ioannidis
 
NM
,
Rothstein
 
JH
,
Pejaver
 
V
. et al.  
REVEL: an ensemble method for predicting the pathogenicity of rare missense variants
.
Am J Hum Genet
 
2016
;
99
:
877
885
.

29.

Schymkowitz
 
J
,
Borg
 
J
,
Stricher
 
F
. et al.  
The fold X web server: an online force field
.
Nucleic Acids Res
 
2005
;
33
:
W382
W388
.

30.

Frazer
 
J
,
Notin
 
P
,
Dias
 
M
. et al.  
Disease variant prediction with deep generative models of evolutionary data
.
Nature
 
2021
;
599
:
91
95
.

31.

Cheng
 
J
,
Novati
 
G
,
Pan
 
J
. et al.  
Accurate proteome-wide missense variant effect prediction with alpha missense
.
Science
 
2023
;
381
:
eadg7492
.

32.

Carlson
 
KR
,
Georgiadis
 
MM
,
Tameire
 
F
. et al.  
Activation of Gcn2 by small molecules designed to be inhibitors
.
J Biol Chem
 
2023
;
299
:
104595
.

33.

Landrum
 
MJ
,
Lee
 
JM
,
Riley
 
GR
. et al.  
Clin Var: public archive of relationships among sequence variation and human phenotype
.
Nucleic Acids Res
 
2014
;
42
:
D980
D985
.

34.

Karczewski
 
KJ
,
Francioli
 
LC
,
Tiao
 
G
. et al.  
The mutational constraint spectrum quantified from variation in 141,456 humans
.
Nature
 
2020
;
581
:
434
443
.

35.

Harding
 
HP
,
Ordonez
 
A
,
Allen
 
F
. et al.  
The ribosomal P-stalk couples amino acid starvation to GCN2 activation in mammalian cells
.
Elife
 
2019
;
8
:e50149. https://doi.org/10.7554/eLife.50149.

36.

Padyana
 
AK
,
Qiu
 
H
,
Roll-Mecak
 
A
. et al.  
Structural basis for autoinhibition and mutational activation of eukaryotic initiation factor 2alpha protein kinase GCN2
.
J Biol Chem
 
2005
;
280
:
29289
29299
.

37.

Lageix
 
S
,
Zhang
 
J
,
Rothenburg
 
S
. et al.  
Interaction between the tRNA-binding and C-terminal domains of yeast Gcn2 regulates kinase activity in vivo
.
PLoS Genet
 
2015
;
11
:
e1004991
.

38.

Dey
 
M
,
Cao
 
C
,
Sicheri
 
F
. et al.  
Conserved intermolecular salt bridge required for activation of protein kinases PKR, GCN2, and PERK
.
J Biol Chem
 
2007
;
282
:
6653
6660
.

39.

Tang
 
CP
,
Clark
 
O
,
Ferrarone
 
JR
. et al.  
GCN2 kinase activation by ATP-competitive kinase inhibitors
.
Nat Chem Biol
 
2022
;
18
:
207
215
.

40.

Szaruga
 
M
,
Janssen
 
DA
,
de
 
Miguel
 
C
. et al.  
Activation of the integrated stress response by inhibitors of its kinases
.
Nat Commun
 
2023
;
14
:
5535
.

41.

Maia de Oliveira
 
T
,
Korboukh
 
V
,
Caswell
 
S
. et al.  
The structure of human GCN2 reveals a parallel, back-to-back kinase dimer with a plastic DFG activation loop motif
.
Biochem J
 
2020
;
477
:
275
284
.

42.

Miosge
 
LA
,
Field
 
MA
,
Sontani
 
Y
. et al.  
Comparison of predicted and actual consequences of missense mutations
.
Proc Natl Acad Sci USA
 
2015
;
112
:
E5189
E5198
.

43.

Dorfman
 
R
,
Nalpathamkalam
 
T
,
Taylor
 
C
. et al.  
Do common in silico tools predict the clinical consequences of amino-acid substitutions in the CFTR gene?
 
Clin Genet
 
2010
;
77
:
464
473
.

44.

Gnad
 
F
,
Baucom
 
A
,
Mukhyala
 
K
. et al.  
Assessment of computational methods for predicting the effects of missense mutations in human cancers
.
BMC Genomics
 
2013
;
14
:
S7
.

45.

Yue
 
P
,
Li
 
Z
,
Moult
 
J
.
Loss of protein structure stability as a major causative factor in monogenic disease
.
J Mol Biol
 
2005
;
353
:
459
473
.

46.

Gerasimavicius
 
L
,
Liu
 
X
,
Marsh
 
JA
.
Identification of pathogenic missense mutations using protein stability predictors
.
Sci Rep
 
2020
;
10
:
15387
.

47.

Liu
 
Y
,
Yeung
 
WSB
,
Chiu
 
PCN
. et al.  
Computational approaches for predicting variant impact: an overview from resources, principles to applications
.
Front Genet
 
2022
;
13
:
981005
.

48.

Doench
 
JG
,
Fusi
 
N
,
Sullender
 
M
. et al.  
Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9
.
Nat Biotechnol
 
2016
;
34
:
184
191
.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.

Supplementary data