Abstract
Background:
Pulmonary arterial hypertension (PAH) is a lethal vasculopathy. Hereditary cases are associated with germline mutations in BMPR2 and 16 other genes. However, these mutations occur in under 25% of idiopathic PAH patients (IPAH) and are rare in PAH associated with connective tissue diseases (APAH). Preclinical studies suggest epigenetic dysregulation, including altered DNA methylation, promotes PAH. Somatic mutations of Tet-methylcytosine-dioxygenase-2 (TET2), a key enzyme in DNA demethylation, occur in cardiovascular disease and are associated with clonal hematopoiesis, inflammation and adverse vascular remodeling. The role of TET2 in PAH is unknown.
Methods:
To test for a role of TET2, we utilized a cohort of 2572 cases from the PAH Biobank. Within this cohort, gene-specific rare variant association tests were performed using 1832 unrelated European PAH patients and 7509 non-Finnish European gnomAD subjects as controls. In an independent cohort of 140 patients, we quantified TET2 expression in peripheral blood mononuclear cells. To assess causality, we investigated hemodynamic and histologic evidence of PAH in hematopoietic Tet2-knockout mice.
Results:
We observed an increased burden of rare, predicted deleterious, germline variants in TET2 in PAH patients of European ancestry (9/1832) compared to controls (6/7509; relative risk=6, p=0.00067). Assessing the whole cohort, 0.39% (10/2572) of patients had 12 TET2 mutations (75% predicted germline and 25% somatic). These patients had no mutations in other PAH-related genes. Patients with TET2 mutations were older (71±7 years versus 48±19 years, p<0.0001) unresponsive to vasodilator challenge (0/7 vs 140/1055 (13.2%)), had lower PVR (5.2±3.1 versus 10.5±7.0 Woods units, p=0.02) and had increased inflammation (including elevation of IL-1β). Circulating TET2 expression did not correlate with age and was decreased in >86% of PAH patients. Tet2-knockout mice spontaneously developed PAH, adverse pulmonary vascular remodeling and inflammation, with elevated levels of cytokines, including IL-1β. Chronic therapy with an antibody targeting IL-1β blockade regressed PAH.
Conclusions:
PAH is the first human disease related to potential TET2 germline mutations. Inherited and acquired abnormalities of TET2 occur in 0.39% of PAH cases. Decreased TET2 expression is ubiquitous and has potential as a PAH biomarker.
Keywords: Associated pulmonary arterial hypertension (APAH), epigenetics, scleroderma, connective tissue disease, CREST syndrome, TET methyl-cytosine dioxygenase 2 (TET2), clonal hematopoiesis of indeterminate potential (CHIP), myelodysplastic syndrome (MDS), DNA methylation, canakinumab
Introduction
Pulmonary arterial hypertension (PAH) is a lethal vasculopathy hemodynamically characterized by increased mean pulmonary arterial pressure (mPAP >20 mmHg) and pulmonary vascular resistance (PVR >3 Wood units)1. Histologically PAH is characterized by obliterative pulmonary vascular remodeling. The current classification system divides Group 1 PAH into idiopathic (IPAH), hereditary (HPAH), and associated PAH (APAH; a category which includes patients with connective tissue diseases (CTD), such as scleroderma). 7-year survival rates of IPAH and CTD-PAH are 56%, and 35%, respectively2. High mortality rates in PAH are a consequence of late diagnosis, due to the non-specific clinical manifestations of the disease, the lack of biomarkers, and the absence of a curative treatment3. Moreover, the fundamental cause(s) of PAH remain elusive in many patients.
The etiology of PAH is heterogeneous and remains imperfectly understood, although it is characterized by increased inflammation and fibrosis and impaired angiogenesis, mitochondrial metabolism and mitochondrial dynamics4. At the genetic level, germline pathologic variants have been reported in 17 genes, by far the most prevalent of which is BMPR255–7. Epigenetic dysregulation of genes is also important in PAH, and pathological activation of DNA methyltransferases (DNMT) has been shown to increase DNA methylation of specific genes associated with disease progression8,9. DNA methylation is a dynamic process that reflects the balance between the activity of DNMT, which adds methyl groups, and ten-eleven translocation methylcytosine dioxygenase (TET), which removes methyl groups, from cytosine nucleotides in DNA. Excessive methylation generally inhibits gene transcription, although there are also interactions between methylation sites and methyl binding proteins, which can alter the expression of other genes10. Somatic inactivating TET2 mutations have recently been associated with development of inflammation11,12 and atherosclerosis13,14. In addition, acquired mutations in this gene underlie clonal hematopoiesis of indeterminate potential (CHIP), a precursor to myelodysplastic syndrome (MDS), myeloproliferative neoplasms (MPN) and even acute myeloid leukemia (AML)15,16. There is no known germline TET2 mutation syndrome.
In this study we evaluated gene specific TET2 exome sequencing data from the largest PAH cohort assembled to date, including 2572 patients in the PAH Biobank. Unlike prior genetic studies, the biobank includes subjects with APAH and non-European ancestry. We performed gene-specific rare variant association analyses using up to 1832 cases of European origin from the PAH Biobank and transcriptomic analysis in an independent cohort to assess TET2 expression. In the entire cohort, we identified 12 predicted deleterious variants in TET2 (75% predicted germline and 25% somatic), novel to PAH. None of the variant carriers were responsive to acute vasodilator challenge. This is the first time that putative germline TET2 mutation has been associated with a human disease. We also identified ubiquitous downregulation of the expression of TET2 in the peripheral blood mononuclear cells (PBMC) of IPAH and APAH patients. Finally, we evaluated Tet2 depleted mice and demonstrated that they spontaneously develop inflammation, pulmonary vascular obliteration, and pulmonary hypertension, providing biological plausibility that disorders in this pathway can cause PAH.
Methods
The data, analytic methods, and study materials for the purposes of reproducing the results or replicating procedures can be made available on request to the corresponding author who manages the information.
Subject study
All patient samples were obtained following written informed consent and approval from local Institutional Review Boards. The study was approved by an institutional review committee and the subjects gave informed consent. Clinical characteristics of the PAH Biobank and John Hopkins cohorts are described in Table-S1. Additional methodologies are available in online supplements.
Exome sequencing, bioinformatics and statistical analyses for genetic studies
DNA was extracted from whole blood from participants in the National Biological Sample and Data Repository for PAH (PAH Biobank), and exome sequencing was carried out in collaboration with the Regeneron Genetics Center or at the Cincinnati Children’s Hospital Medical Center DNA Sequencing and Genotyping Core, as described previously17,18. We used Burrows-Wheeler Aligner (BWA-MEM)19 to map and align paired-end reads to human reference genome (GRCh38), Picard MarkDuplicates to identify and flag PCR duplicates and GATK20 HaplotypeCaller to call genetic variants. We used ANNOVAR21 to aggregate variant annotations, including population allele frequency (gnomAD22 exome/genome, ExAC, and 1000 genomes), predicted functional effects based on RefSeq, and predicted deleterious scores by methods such as REVEL23.
For case-control comparisons, we performed principle components and relatedness analyses for the whole PAH cohort and identified 1832 unrelated European cases. We defined rare variants as variants with an allele frequency <0.01% across all gnomAD exome sequencing samples (European and non-European). We used heuristic filters to minimize technical artifacts between cases and controls, excluding variants that met any of the following criteria: missingness >10%, minimum alternate allele read depth ≤4 reads, alternative allele fraction ≤25%, or genotype quality <90. We used 7509 European gnomAD whole genome sequence (WGS) data as the control set. Only variants with FILTER “PASS” in gnomAD WGS (data release v 2.02) and located in the IDT xGen captured protein coding region were included in the analysis. Finally, we observed that the frequency of rare synonymous variants in cases and controls was virtually identical, indicating that the data sets are comparable (enrichment rate=1.0, p=0.11) (Table-S2). Assuming linkage disequilibrium is negligible among ultra-rare variants, we performed gene-wise burden tests of rare variants for TET2 using Fisher’s exact test to compare carrier frequency between cases and controls. We tested the association in three groups of variants between cases and controls: (a) rare deleterious missense variants (“D-MIS”, defined as REVEL score >0.5); (b) likely-gene-disrupting (LGD) variants; and (c) D-MIS+LGD. We tested 3 genes TET1, TET2, TET3. We also tested the association in IPAH alone and APAH alone. Thus, we performed 11 tests (3 genes × 3 sets of variants and 1 gene × 2 PAH subclasses) and set the Bonferroni-corrected threshold for significance at p<0.0045. We then assessed the entire PAH Biobank (n=2572 cases) for rare (AF<0.0001), deleterious (LGD or D-MIS) variants in TET2 using Integrative Genome Viewer (Illumina, San Diego, CA). Nearly 100% of both cases and controls had >10X sequencing coverage across the gene, and 99.6% of the targeted regions had >15X coverage in cases and 100% in controls (Figure-S1A,B), indicating that there was no systematic bias in the detection of TET2 variants in cases vs controls.
To detect likely somatic variants (mosaicism) in TET2, we first used SAMtools (version 1.3.1–42)24 to improve calling of genetic variants with low allele fraction. We then processed SAMtools calls using a set of heuristic filters to remove variants located in repeat regions (mapability, segmental duplication) or showing evidence of strand bias, and screened all variants using Integrative Genome Viewer. We then took the union of variants called by GATK HaplotypeCaller and SAMtools and considered variants with alternative allele fraction less than 25% as likely somatic mutations. Mosaic mutations were confirmed by TA cloning (Thermo Fisher, Waltham, MA) of exonic PCR products followed by Sanger sequencing of individual clones (Figure-S1C). Details are available in the online supplements.
Microarray and gene expression
A microarray assay was performed on RNA extracted from peripheral blood mononuclear cells (PBMC) of 50 scleroderma-associated PAH (SScPAH), 30 IPAH, 19 scleroderma without PAH patients (SSc) and 41 healthy controls from Johns Hopkins PH Program and Johns Hopkins Scleroderma Center, as previously published25 and described in online supplementals. The expression data have been deposited in NCBI’s Gene Expression Omnibus and are accessible through GEO Series accession number GSE 33463. Analytical methods were performed as previously published25. Details are available in the online supplements.
Animal experiments
All experiments were performed in accordance with Queen’s University biosafety and ethical guidelines (ROMEO/TRAQ#6016826). The procedures followed were performed in accordance with institutional guidelines. Conditional, haematopoietic heterozygous (Tet2+/−) and homozygous (Tet2–/–) knockouts were generated by crossing parental floxed (Tet2f/f, B6;129S-Tet2tm1.1Iaai/J) and Vav1-Cre (B6.Cg-Tg(Vav1-Cre)A2Kio/J) mice (Jackson Laboratory, Bar Harbor, ME) and validated, as previously described12,26. IL-1β PAH regression experiments were performed on 5 Tet2f/f and 10 Tet2–/–. 10 Tet2–/– mice (7 months old) were randomly distributed in 2 groups of 5 age/sex-matched mice and treated with anti-IL-1β antibody (a generous gift from Novartis Pharma AG) at a dose of 10 mg/kg/week IP for 6 weeks) or IgG2a (a generous gift from Novartis Germany) as a placebo. Treatments and data collection were performed by scientists blinded to treatment groups. PAH development was assessed by echocardiography, right heart catheterization, 2-photon, confocal microscopy lung perfusion and histology, as described in online supplements. Inflammation was assessed by fluorescence-activated cell sorting (FACS) and NanoString nCounter PanCancer Immune Panel Profiling (Seattle; WA), as previously published12 (see online supplements).
Results
Exome sequencing identifies rare deleterious variants in TET2.
Detailed characterization of the PAH Biobank and gnomAD cohort are described in a separate report18 and in Table-S1. The cohort includes 2572 total cases: 43% IPAH, 48% APAH 4% FPAH and 5% other (Table S1). Among APAH patients, 58% (722/1239) had some form of connective tissue disease. The majority of cases are adult-onset, with a 3.7:1 ratio of females to males, typical of adult PAH. The genetically determined ancestries were 72% European, 12% Hispanic, 11% African, and smaller percentages of other ancestries.
We first limited the association analysis to 1832 unrelated European cases and 7509 non-Finnish, European controls. Using a REVEL score >0.5 to define D-MIS variants, we tested for association in three categories of variants: D-MIS, LGD or D-MIS+LGD. Among all PAH cases, we observed significant enrichment of LGD (8/1832 cases vs 4/7509 controls; RR=8.18, p=0.0005) as well as D-MIS+LGD variants for TET2 (9/1832 cases vs 6/7509 controls; RR=6.15, p=0.00068) (Table-1). The association was largely due to patients with IPAH (RR=10.79, p=8.483e-05) (Table-2).
Table 1.
Enrichment of rare predicted deleterious variants* in candidate PAH risk gene TET2† amongst European PAH cases.
TET2 | ||||
---|---|---|---|---|
Mutation type‡ | PAH cases (n=1832) | gnomAD WGS controls (n=7509) | Relative risk | P-value |
D-MIS | 1 | 2 | 2.05 | 0.48 |
LGD | 8 | 4 | 8.18 | 0.0005 |
D-MIS or LGD | 9 | 6 | 6.15 | 0.00068 |
Table 2.
Enrichment of rare predicted deleterious variants* in candidate PAH risk gene, TET2†, among European APAH and IPAH cases.
TET2 | |||
---|---|---|---|
PAH subclass | D-MIS or LGD‡ | Relative risk | P-value |
APAH (n=843) | 2 | 3.00 | 0.19 |
IPAH (n=812) | 7 | 10.79 | 8.483e-05 |
Variant filters: MAF <0.0001 and likely gene damaging or missense with REVEL score >0.5.
TET2 transcript: NM_001127208.2.
D-MIS, missense with REVEL score >0.5; LGD, likely gene damaging
Abbreviations: APAH associated pulmonary arterial hypertension; MAF, minor allele frequency; IPAH, idiopathic pulmonary arterial hypertension; WGS, whole genome sequencing.
We then identified 1 additional mutation in the 740 non-European patients of the PAH Biobank (Table-3 and S3A). In the total cohort of 2572 cases, we identified 9 unique likely germline variants in 8 patients (Table-3 and S3A) and 3 somatic variants in 3 patients (Table-3 and S3B and Figure 1A). All mutations were unique, meaning the affected patients were not shown to carry variants in known PAH risk genes7,27,28. In addition, none of the TET2 mutant patient carriers had hematologic malignancies at the time of enrollment in the PAH Biobank. Two-dimensional structure of the encoded protein showed that 6 deleterious variants (both germline and somatic) localized to the conserved catalytic (TET/J-binding protein methylcytosine dioxygenase activity) domain (Figure 1B).
Table 3.
Rare, predicted deleterious germline and somatic variants in candidate PAH risk gene TET2 amongst 2572 PAH Biobank cases.
TET2* | ||||||||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Patient ID | Mutation type | Nucleotide change | Amino acid change |
MAF, gnomAD WES | VAF | CADD phred | REVEL score |
PAH class | Gender | Genetic ancestry | Age (y) enrollment | Age (y) onset | Mean PAP, (mmHg) | Mean PCWP (mmHg) | CO (L/min) |
PVR (WU) |
Mean SAP (mmHg) | Mean SAP:PAP |
03–057 | Germ | c.1530_1531insCACCT | p.Lys513Thrfs22 | . | 31% | . | . | IPAH | F | EUR | 74 | 65 | 38 | 14 | 4.2 | 5.71 | NA | NA |
03–100 | Germ | c.2233C>T | p.Gln745✶ | . | 38% | 37 | . | IPAH | F | EUR | 71 | 70 | 47 | 10 | 5.55 | 6.67 | NA | NA |
12–206 | Germ | c.2872C>T | p.Gln958✶ | 1.22E-05 | 39% | 41 | . | IPAH | F | EUR | 84 | 84 | 55 | 8 | 3.67 | 12.81 | 87 | 1.58 |
29–016† | Germ Soma |
c.4081G>A c.5618T>C |
p.Gly1361Ser p.Ile1873Thr |
. 1.33E-05 |
49% 46% |
34 26 |
0.57 0.61 |
IPAH | F | EUR | 72 | 69 | 47 | 17 | 9.23 | 3.25 | 80 | 1.7 |
19–036 | Germ Germ |
c.4546C>T c.817C>T |
p.Arg1516✶ p.Gln273✶ |
1.59E-05 | 54% 51% |
40 36 |
. | IPAH | F | EUR | 74 | 67 | 33 | 9 | 9.44 | 2.54 | 117 | 3.5 |
12–156 | Germ | c.4893T>G | p.Tyr1631✶ | . | 36% | 36 | . | IPAH | M | EUR | 84 | 83 | 40 | 12 | 3.3 | 8.48 | 88 | 2.2 |
13–025 | Germ | c.3336delA | p.Asp1113Ilefs✶4 | . | 26% | . | . | APAH-Porto | M | EUR | 67 | 66 | 36 | 13 | 4.8 | 4.79 | NA | NA |
08–055 | Germ | c.4396C>T | p.Gln1466✶ | . | 47% | 45 | . | APAH-CTD | F | EUR | 73 | 69 | 45 | 10 | 3.7 | 9.46 | NA | NA |
30–037 | Soma | c.2862G>A | p.Trp954* | . | 18% | 39 | . | APAH-CTD | M | Hispanic | 48 | 47 | 46 | 5 | 6.53 | 6.28 | 116 | 2.52 |
23–014 | Soma | c.5639T>C | p.Leu1880Pro | . | 26% | 26 | 0.62 | APAH-CHD | F | EUR | 86 | 79 | 46 | 4 | 5 | 8.4 | NA | NA |
Mean ± SD, TET2 carriers | 3.5:1 | 73.3± 10.9 | 66.9 ± 10.7 | 43.3± 6.5 | 10.4 ± 3.6 | 5.5 ± 2.2 | 6.8 ± 3.1 | 97.6 ± 17.5 | 2.3 ± 0.8 | |||||||||
n, TET2 carriers | 10 | 10 | 10 | 10 | 10 | 10 | 5 | 5 | ||||||||||
Mean ± SD, cohort APAH+IPAH excluding TET2 carriers | 52 ± 18 | 48 ± 19 | 50 ± 14 | 10 ± 4 | 4.5 ± 1.8 | 10.5 ± 7.0 | 90 ± 19 | 2.0 ± 0.7 | ||||||||||
n, cohort APAH+IPAH excluding TET2 carriers | 2340 | 2340 | 2292 | 2232 | 1641 | 1595 | 1399 | 1397 | ||||||||||
p-Value | 1.00E-05 | 1.00E-05 | 0.002 | NS | NS | 0.02 | NS | NS |
Variant filter: allele frequency <0.0001 and likely gene disrupting (stop/gain, frameshift or canonical splicing) or missense with REVEL score >0.5.
TET2 transcript: NM_001127208.2
Exceptions to the mosaic pipeline variant filter (MAF and alternate allele fraction); detected as known mutation hotspot in cancer.
Abbreviations: APAH-CHD, pulmonary arterial hypertension associated with congenital heart disease; APAH-CTD, pulmonary arterial hypertension associated with connective tissue diseases; APAH-Porto, pulmonary arterial hypertension associated with porto-pulmonary hypertension; FPAH, familial pulmonary arterial hypertension; Germ, germline; MAF, minor allele frequency; IPAH, idiopathic pulmonary arterial hypertension; PAP, pulmonary artery pressure; PCWP, pulmonary capillary wedge pressure; PVR, pulmonary vascular resistance; SAP, systemic arterial pressure; Soma, somatic; WES, whole exome sequencing; VAF, variant allele frequency; WU, Wood unit. Unpaired t-test
Figure 1. Increased mutation and decreased TET2 gene expression in blood from PAH patients.
Topologic analysis and gene expression of the Human DNA tet methylcytosine dioxygenase 2 (TET2) in pulmonary arterial hypertension (PAH). A) Distribution of TET2 somatic and germline deleterious variants. B) Locations of rare deleterious PAH patient-derived TET2 variants within the two-dimensional protein structures. The numbers of variants at each amino acid position are indicated on the y-axis. Germline variants are shown above the protein schematic; mosaic variants are below. D-MIS, predicted damaging missense; LGD, likely-gene-disrupting (stopgain, frameshift). TET/JBP, TET/J-binding protein catalytic domain. Protein domain coordinates were modified according to UniProtKB). C) TET2 gene expression in peripheral blood mononuclear cells (PBMCs) from 41 healthy control, 30 idiopathic PAH (IPAH), 50 scleroderma associated PAH (SSc-PAH) and 19 scleroderma without PAH (SSc). Violin plot was used to visualise data distribution; black line is the median; red lines are the upper and lower quartile. one-way ANOVA. **P<0.01; ****P<0.0001; n.s. non-significant.
TET2 variant carriers exhibited increased overall inflammation compared to age/sex-matched PAH non-mutated patients, and age/sex-match controls, as assessed by measurement of 42 circulating inflammatory markers (area under the curve AUC 266.9 ±81.74 for TET2 carriers; 117.1 ±58.9 TET2 non-carriers; 41±5 for healthy control p<0.05) (Figure S2A–B). We focused on the expression of pro-inflammatory cytokines/chemokines and reported that TET2 variant carriers had increased expression of 30 pro-inflammatory cytokines (AUC 204.3 ±72.14 versus 75.26 ±17.92 p<0.001) (Figure 2A–B and S2A–B and Table-S4). IL-1β expression was increased in 70% of TET2-mutated patients compared to their matched non-mutated PAH patients (Figure S2C).
Figure 2. TET2 mutation is associated with a pro-inflammatory phenotype in blood of PAH patients.
Expression of 30 pro-inflammatory cytokines were assessed in blood of 9 healthy patients, 10 age/sex-matched PAH TET2 variant carriers and 10 matched PAH TET2 non-carriers patients. A) TET2 variant carriers show increased levels of 28 cytokines (IFNa2; IP-10; IL-12p40; IL12p70; IL-6; IL-1β; IL-2; Fractalkine; IFNy; IL-15; IL-1a; IL-18; IL-3; G-CSF; IL-7; TNFa; IL-17A; MIP-1a; MIP-1B; MDC; TNFB; IL-5; Flt-3L; MCP-3; IL-8; RANTES; GRO alpha) and decreased levels of 2 cytokines (MCP-1; Eotaxin) compared to matched non-carriers patients. Values are expressed as fold change compared to healthy patients. B) TET2 carriers display overall increased levels of pro-inflammatory markers measured by an increase of the area under the curve. One-way ANOVA. Values are expressed as mean±SEM. ***P<0.001
Clinical phenotypes, including demographics, PAH medication and right heart catheterization (RHC) data, are provided in Tables 3 and 4. None of the TET2 variant carriers (0/7) vs 140/1055 (13.2%) in the remainder of the cohort, were vasodilator responders, as assessed by standard criteria (Table 4)29. Consistent with the severity associated with lack of vasodilator responsiveness, the use of endothelin receptor inhibitors and soluble guanylate cyclase stimulators was increased in TET2 variant carriers (Table 4). Two-thirds (6/10) of the patients with rare deleterious germline or somatic TET2 variants had IPAH (Table 4 and S3) Female:male ratio and genetic ancestry of TET2 germline and somatic carriers were similar to the overall cohort whilst the mean age-of-onset (66.9 ± 10.7years) was significantly older than that of all APAH+IPAH (48±19 years, p<0.0001) (Table 3). In addition the age of TET2 subjects in our study was older than in previous reports for patients with PAH related to mutations in BMPR25,6,17 and TBX417. TET2 variant carriers had lower mPAP (43.3±6.5 mmHg) and PVR (6.8±3.1 Woods units) compared to all the remaining APAH+IPAH subjects in our cohort (50±14 mmHg, p<0.0001 and 10.5±7.0 Woods units, p=0.02) (Table 3).
Table 4.
Vasodilator response* and PAH medication among TET2 deleterious variant carrier
Vasodilator used | Cohort APAH+IPAH excluding TET2 carriers | APAH+IPAH germline TET2 carriers | APAH+IPAH somatic TET2 carriers | APAH+IPAH TET2 carriers† |
---|---|---|---|---|
Oxygen + Nitric Oxide | 50/286 (17.5%) | 0/3 (0%) | 0/1 (0%)‡ | 0/3 (0%) |
IV Epoprostenol | 13/110 (11.8%) | - | - | - |
Inhaled Nitric Oxide | 60/564 (10.6%) | 0/2 (0%) | 0/1 (0%) | 0/3 (0%) |
Oxygen | 5/16 (31.2%) | - | - | - |
IV Nitroprusside | 1/7 (14.3%) | - | - | - |
IV Adenosine | 5/37 (13.5%) | - | 0/1 (0%) | 0/1 (0%) |
Inhaled Iloprost | 1/2 (50%) | - | - | - |
Inhaled Epoprostenol | 1/6 (16.7%) | - | - | - |
Alprostadil | 2/8 (25%) | - | - | - |
Calcium Channel Blocker | 0/3 (0%) | - | - | - |
Unknown | 2/16 (12.5%) | - | - | - |
Total | 140/1055 (13.2%) | 0/5 (0%) | 0/3 (0%) | 0/7 (0%) |
PAH medication | Cohort APAH+IPAH excluding TET2 carriers | APAH+IPAH TET2 carriers† | ||
PDE5 inhibitor | 2014/2674 (75.3%) | 14/18 (77.8%) | ||
Prostacyclin analog | 1356/2674 (50.7%) | 10/18 (55.6%) | ||
Endothelin receptor inhibitor | 1036/2674 (38.7%) | 11/18 (61.1%)‡ | ||
Stimulator of soluble guanylate cyclase | 44/2674 (1.6%) | 1/18 (5.6%) | ||
Calcium channel blocker | 239/2674 (8.9 %) | 0/18 (0%) | ||
RTK inhibitor | 5/2674 (0.2%) | 0/18 (0%) | ||
Unknown | 29/2674 (1.1%) | 1/18 (5.6%) |
Positive vasodilator responders were defined according to standard criteria as a drop of mean pulmonary arterial pressure (mPAP) >10mmHg to mPAP at rest <40mmHg with preserved or improved cardiac output.
Somatic + germline carrier (patient 29–016)
Difference statistically significant compared to cohort APAH+IPAH excluding DNMT3A and TET2 carriers; p< 0.05, z-test.
APAH, associated PAH; IPAH, idiopathic PAH; IV, intra-venous; PDE5, phosphodiesterase 5; RTK, receptor tyrosine kinase
Decreased TET2 gene expression in PAH
Having established the occurrence of TET2 rare deleterious variants in PAH patients, we next investigated TET2 expression in PBMCs. Gene expression omnibus (GEO) analysis was acquired from 50 SSc-PAH patients, 30 IPAH patients, 19 scleroderma without PAH patients (SSc) and 41 healthy controls. All the cases had adult-onset PAH, with genetically determined ancestries as follows: 82.1% Caucasian, 14.3% African, with smaller percentages of other ancestries. The sex ratios (female:male) were control 4.9:1, IPAH 5:1, and SSc-PAH 3.7:1 whilst all SSc patients were female (Table-S1). TET2 gene expression, relative to healthy subjects, was decreased in 86% of SSc-PAH patients (relative expression; 0.75; p<0.0001); 86.7% of IPAH patients (relative expression; 0.74; p<0.0001) and 68% of scleroderma patients without PAH (SSc; relative expression; 0.79; p<0.01) (expression normalized to 1) (Figure-1C). Receiver operating characteristic curve (ROC) analysis was performed for 41 healthy control and 80 PAH patients (IPAH/SSc-PAH) and revealed a potential biomarker value for TET2 expression (AUC:0.78; p<0.0001) (Figure S3A–G). When assessed using multiple logistic regression analysis, including both age and TET2 expression, ROC curve analysis reveals an AUC of 0.86, p<0.0001 [specificity 81%, sensitivity 80% (Figure S3–H)]. The AUC is significantly higher in the model including age and TET2 compared to a model including age alone (Z-score =2.47), as analysed following the method described by Hanley and McNeil30. This observation confirms that TET2 expression has a potential value as biomarker for PAH. Gene expression of other TET paralogs, TET1 and TET3, is not affected in PAH. Moerover,TET2 expression does not correlate with the age (Table-S5A–B, Figure S3I,J)
Experimental Tet2 depletion is associated with spontaneous development of PH in mice
We next assessed the biologic plausibility that hematopoietic TET2 depletion could result in PAH. We performed hemodynamic measurement in 15 conditional Tet2 KO mice (Tet2−/−) and 15 sex and age-matched control mice (Tet2f/f) (9 males; 6 females; age 7 to 10 months). Mice with Vav-Cre-mediated Tet2 depletion spontaneously developed pulmonary hypertension, evident as a significant decrease in PAAT (p<0.01), an increase in RVSP (p<0.001), TPR (p<0.01), arterial elastance (p<0.01), adverse pulmonary vascular remodeling (p<0.05) and decreased perfusion of distal pulmonary arteries (p<0.01), compatible with obliteration of the microvasculature (Figure-3A–H and Figure-S4A–F; S5A–D; S6A). We observed no change in heart rate, stroke volume, cardiac output, RV-dP/dTmax, RV-dP/dTmin or LV function (LVS weight, E/E’, MAPSE, arterial systolic and diastolic pressure, LV-EF, LVSP, LVEDP, Tau Mirsky, dP/dtmax, dP/dtmin) (Figure-S6B–F and S7A–K). Heterozygous animals (Tet2+/−) also had increased RVSP (p<0.05) and TPR (p<0.05), with a trend toward increased arterial elastance but no changes in CO (Figure-S8A–D). We assessed PH development in 2-month-old Tet2−/− mice (5 males) and showed that, compared to age-matched Tet2f/f mice, Tet2 depletion in young animals was not associated with PH phenotype (no significant differences in RVSP, TPR, CO) (Figure-S9A–C).
Figure 3. TET2 depletion in hematopoietic cells and pulmonary hypertension in a murine model.
Haemodynamic assessment of pulmonary hypertension using right heart catheterization and echocardiography of 6–15 Tet2−/− and age-sex matched Tet2f/f mice show A) increased right ventricular systolic pressure (RVSP), B) decreased pulmonary artery acceleration time (PAAT) and, C) increased total pulmonary resistance (TPR) in Tet2−/− mice compared to Tet2f/f animals. D) Pulmonary arterial vascular remodelling was blindly quantified by the percent of wall thickness: (total diameter−internal diameter)/total diameter by immunofluorescence (smooth muscle actin); 10 arteries per mice on 5 mice per group. Pulmonary vascular wall thickness is increase in Tet2−/− mice compared to Tet2f/f. Perfusion of pulmonary vessels has been assessed in Tet2−/− and Tet2f/f mice (FITC albumin perfusion, 2 photon microscopy). Perfused vessels have been clustered according to their volumes in 3 categories: small (15–225μm3), intermediate (225–3347μm3) and big (3347–50000μm3). Tet2−/− mice display decreased numbers of E) small and F) intermediate (P=0.08) perfused vessels whilst G) number of large vessels perfused remains the same compared to Tet2f/f (number of vessels/1e6 μm3). H) Representative pictures of vessels perfused by FITC-albumin in the lung of Tet2−/− and Tet2f/f (500X550μm). I) Compared to Tet2f/f animals, lungs from mutated mice (Tet2−/−) show an elevated macrophages population measured by fluorescence activated cell sorting (FACS; F4/80; CD11b; n=6 Tet2−/− and age-sex matched Tet2f/f). J) Quantification of inflammatory cytokines and chemokines in total lung of 3 Tet2−/− and age-sex matched Tet2f/f mice displays up-regulation of Il1b, Cxcr2, Csf3r, C5ar1, Fpr2, Amica1, Ccr1, Mmp9, Cd33, Itgam, H2-Q10, Arg2, Clec4n, Il1rn, Rsad2, Il1r2 and downregulation of Ccr6 gene expression in mutated animals. Results show change >1.8 fold Log2-transformed normalized NanoString mRNA counts. Unpaired t- test. Values are expressed as mean±SEM. n.s.: non-significant; *P<0.05; **P<0.01; ***P<0.001.
PH development was associated with increased inflammation in the lungs of Tet2−/− mice, evident as macrophage accumulation (p<0.01; Figure-I) and dysregulation of 61 inflammatory markers (59 upregulated and 2 downregulated, Table-S6). Seventeen inflammatory markers had a change >1.8-fold (up-regulation of Il1b, Cxcr2, Csf3r, C5ar1, Fpr2, Amica1, Ccr1, Mmp9, Cd33, Itgam, H2-Q10, Arg2, Clec4n, Il1rn, Rsad2, Il1r2 and downregulation of Ccr6) (Figure-3J). Tet2 is a critical regulator of DNA methylation. It is not surprising that we observed an increased DNA methylation (5mC) level in bone marrow and lung tissue of Tet2−/− mice (Figure-S10A–C). Tet2 depletion was confirmed by PCR and by decreased Tet2 protein in the lung tissue of Tet2−/− mice (Figure-S11A–B). Thus, hemopoietic Tet2 depletion is sufficient to increase DNA methylation and exacerbate inflammation and induce PH in mice, providing biological plausibility for the importance of the mutations and pathway perturbations we found in patients.
IL1-β blockade reverses the PH phenotype in Tet2 depleted mice.
Having established that PH development in Tet2 mutated mice is associated with increased inflammation, we assessed the potential therapeutic effect of anti-inflammatory therapy. We treated 5 Tet2−/− mice (3 males and 2 females) with anti-IL-1β antibody (IP; 10 mg/kg/week; 6weeks) and 5 age/sex-matched Tet2−/− mice with IgG2a isotype control (IP; 10 mg/kg/week; 6weeks) at the age of PH onset (7 months) assessed by decreased PAAT (Figure-S12A). After 6 weeks of treatments, we first confirmed that animals treated with placebo developed PH evident as increased Fulton index, decreased PAAT, increased RVSP, mPAP, and TPR index (Figure-4A–E and Figure-S12A). Anti-IL-1β antibody treatment prevented weight loss observed in Tet2−/− mice and improved PH hemodynamics and phenotype parameters to healthy Tet2f/f, levels (PAAT, RVSP, mPAP, TPRI) (Figure-4A–E). Note that IL1-β antibody treatment induced no kidney or liver toxicity (Table-S7). Our results suggest that targeting inflammation through IL1-β blockade improves PH in Tet2−/− mice and confirms the contribution of inflammation (especially IL1-β) in the etiology of PH related to Tet2 depletion.
Figure 4. IL-1β blockade reverses PH in Tet2 mutated mice.
10, 7-monthTet2−/− mice were treated with an antibody against IL-1β (Tet2−/− + IL-1β ab; IP; 10mg/kg/week; 6weeks) or IgG2a (Tet2−/− + IL-1β ab IgG2a; IP; 10mg/kg/week; 6 weeks). Tet2−/− + IL-1β ab mice showed A) significant increased pulmonary artery acceleration time (PAAT); B) decreased right ventricular systolic pressure (RVSP); C) decreased mean pulmonary arterial hypertension (mPAP); D) reduced Total pulmonary resistance (TPR) normalized by body weight associated. E) IL-1β antibody treatment significantly prevents weight loss observed in Tet2−/− mice treated with IgG2a. n=5 per group. One-way ANOVA; mixed effect ANOVA. Values are expressed as mean±SEM. *P<0.05, **P<0.01, ***P<0.001.
Discussion
We used the largest cohort of PAH patients available to identify TET2 as a novel gene that is mutated in PAH. TET2 mutation, observed in 0.39% of PAH cases (40% APAH, 60% IPAH), is associated with a 6.15-fold increased risk of PAH, relative to the gnomAD control database. TET2 mutations occurred in patients who were confirmed to be free of mutations in established PAH genes. Based on their allele fraction in peripheral blood, 75% of TET2 mutations are predicted to be germline versus 25% somatic. In an independent cohort, a decrease of TET2 expression was found in >86% of APAH and IPAH patients. Finally, we reported that conditional hematopoietic Tet2 knockout is sufficient to induce PH in mice.
Epigenetic mechanisms link genes and environment. They include changes in DNA methylation, histone acetylation and production of micro-RNAs. TET enzymes are key regulators of DNA demethylation, catalyzing the conversion of the methylated nucleotide 5-methylcytosine into 5-hydroxymethylcytosine. By subtraction of methyl groups on DNA and association with histone deacetylases31, TET enzymes contribute to the epigenetic regulation of gene expression. Our group and others showed that hyper-methylation of specific target genes contributes to the development of PAH8,9,32. Supporting our observation, TET2 loss of function leads to increased DNA methylation whilst TET2 hematopoietic depletion results in inflammation and cardiac dysfunction33,34. We observed increased adverse pulmonary vascular remodeling in the lungs of Tet2−/− mice (Figure 3D), consistent with previous observations implicating Tet2 depletion in development of atherosclerotic lesions35. We also confirmed that hematopoietic Tet2 depletion increases DNA methylation (Figure-S10) and inflammation12 evident as macrophage accumulation, and increased expression of inflammatory mediators (Il1b, Cxcr2, Csf3r, Ccr1, Mmp9, Cd33, Itgam, Il1rn, Il1r2) in Tet2−/− lungs (Figure-3I, J). These factors are known to be associated with the development of PAH36. Validating our observations, we reported a global increase of pro-inflammatory markers in the blood of human PAH patients with TET2 deleterious variants compared to age/sex-matched non-carriers (Figure 2). The fact that this mouse spontaneously develops PAH experimentally links hematopoietic Tet2 inactivation to vascular remodeling and inflammation, two common features of the PAH phenotype36.
We generated conditional hematopoietic Tet2 mutated mice to mimic TET2 deleterious variants in human patients and the ubiquitous decrease in TET2 expression observed in the peripheral blood cells of PAH patients. We showed that homozygous Tet2−/− mice spontaneously developed a PH phenotype and notably manifested a profound loss of the pulmonary microvasculature. Heterozygous Tet2+/− mice have a genotype closer to the human condition described in this article and have a less pronounced phenotype (p<0.05), suggesting gene dose-effect response. This observation experimentally shows that, in an animal model, a total or partial loss of Tet2 function can induce vascular remodeling secondary to increased inflammation.
Age of onset of PAH is significantly higher in patients harboring somatic and germline TET2 variants compared to the remainder of the cohort (66.9 ±10.7 VS 48 ±19 years) (Table 3). Consistent with this observation, older (7 month) Tet2−/−mice spontaneously developed PH, whilst we did not detect significant PH in younger (2 month) Tet2−/−mice (Figure 3 and Figure S9). This observation suggests that aging contributes to PH related to TET2 mutations in both humans and mice. We believe a second hit may be required to elicit clinically evident PAH, such as occurs in CHIP and myeloid cancer risk. Indeed, Tet2 normally restrains inflammation, but an initial trigger may be required to induce inflammation. Thus, TET2 depletion exacerbates inflammation when it occurs in a pro-inflammatory environment. Chronic inflammation associated with aging might potentially be such a second hit37 and thereby elicit clinically evident PAH in TET2 deleterious variant carriers.
Mutations in 12 established risk genes and 5 recently-validated risk genes predispose to PAH, with BMPR2 mutations as the most common genetic cause of HPAH and IPAH7,18. Here we identify an additional novel PAH gene and report that predicted deleterious germline and/or somatic variants of TET2 underlie 0.39% of PAH cases. Our limited read depth, using whole exome data, likely underestimates the prevalence of somatic mutations (i.e. clonal hematopoiesis of indeterminate potential, CHIP). These TET2 mutations are observed in patients who were shown to be free of the other known PAH risk genes variants. Interestingly, TET2 mutations also occur in APAH patients, who are known to have more severe inflammatory states and greater mortality rates than other PAH groups38. Similarly, we reported that TET2 variant carriers exhibit a pro-inflammatory phenotype compared to age/sex-matched, non-carrier, PAH patients. It is possible that patients with APAH might also be inflamed as a result of their connective disease. However, the restricted sample size of this sub-study (n=10 patients with or without TET2 mutations) and the variability in blood cytokine levels amongst patients, required us to perform our analysis on all mutation carriers, not just those with IPAH. Thus, we cannot exclude some confounding effects related to the inclusion of 4 patients with APAH and TET2 mutations in this cohort.
Unlike our study, the NIHR BioResource-Rare Diseases PAH study, which investigated deleterious variation in 1048 IPAH/FPAH patients, did not enroll non-European subjects or APAH patients and did not report deleterious variants in TET239. Our study has a ~2.5-fold larger sample size, used exome rather than genome sequencing, specifically assessed somatic mutations, and included APAH patients (49% of the 2572 patients). This is the first gene associated with APAH aside from those associated with congenital heart disease.
Over 86% of PAH patients have decreased TET2 expression, suggesting an acquired mechanism of TET2 gene dysregulation in PAH. Consistent with this speculation, hypoxia, as well as metabolic disorders observed in PAH, are also associated with pathologic regulation of TET2 expression40. The difference between the rarity of TET2 mutations versus the ubiquitous dysregulation of TET2 expression mirrors what is seen with BMPR2 in PAH. BMPR2 mutations in non-hereditary PAH occur in the minority of subjects whereas BMPR2 mRNA and protein expression is downregulated in most forms of human and experimental PAH5,6.
TET2 is one of the two most commonly mutated genes in CHIP, which is associated with an increased risk of atherosclerosis and elevated inflammation41. CHIP mutations lead to elevation of interleukin 1β42. The Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS)43, which investigated the effect of a monoclonal antibody targeting interleukin-1β (Canakinumab) in cardiovascular diseases, identified the presence of CHIP (and TET2 mutation) in 8.8% of the cohort. Patients with a somatic mutation in TET2 showed a greater magnitude of risk for major adverse cardiovascular events (MACE); however, they also displayed an improved therapeutic response to canakinumab. We showed that conditional hematopoietic Tet2 depletion is associated with accumulation of IL-1β in the lung of mice (Figure 3J) and reported that IL-1β blockade reverses PH in Tet2−/− mice (Figure 4). In PAH patients, elevated serum levels of IL-1β correlate with a worse outcome and targeting this cytokine, in a preclinical model of PAH, improved the disease44. An IL-1β receptor antagonist is currently in phase 1 clinical trial in PAH (NCT03057028). As observed for CANTOS and in Tet2−/− mice, our results suggest that genetic investigation of TET2 variants and expression might be relevant to predict the likelihood of benefit from IL-1β based therapy. Thus, discovery of this new gene mutation in PAH may have therapeutic consequences.
CHIP is also a precursor to myeloid neoplasms such as MDS and AML13,15. Intriguingly, previous studies have shown that PAH is observed in 15–48% of patients with MDS, where TET2 is one of the most common mutated genes45. We demonstrated that hematopoietic Tet2 depletion is associated with PAH and pulmonary vascular remodeling in mice. The same mutant mice ultimately develop an MDS-like disease26, which experimentally confirms the link between Tet2 depletion, PAH and MDS; also suggesting a relationship between CHIP and PAH. The moderate severity of pulmonary hypertension reported in our animals (RVSP 30.2 ± 1.3mmHg; Figure-3A) and the milder severity of the hemodynamic derangement in TET2 mutant patients (Table 3) suggests this gene mutation might result in a less symptomatic state in patients, which would be hard to detect without active surveillance. Borderline elevation of systolic PAP (29mmHg at rest) was recently reported in a cohort of 34 MDS patients, lending support to our experimental observations46.
Our observations suggest that CHIP and PAH are two manifestations of similar mutations. Whether the manifestation of the mutations will be vascular or hematopoietic (or both), likely reflects a complex interplay of factors including: mutation burden, mutation-related inflammatory consequences, and cellular/tissue distribution of the mutations. The Tet2-knockout mouse is an accepted model of human myeloid neoplasia and our discovery that PAH develops spontaneously as these mice age, indicates that both age and TET2 deficiency may also interact to yield different disease manifestations.
Limitations
Using the largest PAH cohort to date, we reported enrichment of rare deleterious variants for TET2 among 2572 PAH patients. However, we did not investigate the occurrence of TET2 mutations in other forms of pulmonary hypertension. This limitation precludes any conclusion regarding the specificity of the mutation to group 1 PH versus patients with group 2–5 PH. Similarly, as observed for BMPR2 mutation, which is mainly reported in PAH but also observed in cancer47 (e.g. gastric, colorectal, breast cancer), we acknowledge that TET2 mutation might not be specific for PAH. Indeed, it is known that somatic mutations in TET2 are common in MDS and AML and can portend poor prognosis48. However, none of our patients had known active myeloid neoplasms, and we have provided strong evidence of the contributions of deleterious mutations in TET2 to PAH’s etiology.
Kaasinen and colleagues have recently reported germline TET2 frameshift mutation in a lymphoma family49. They did not mark observation of unusual predisposition to atherosclerosis nor abnormal pro-inflammatory cytokine or chemokine expression in this family. However, their study was conducted in a Finnish population which was not investigated in our work. The mutation (c.4500delA) reported by Kaasinen and al, was not observed in the PAH cohort. In their report, TET2 mutation was not associated with significantly decreased TET2 expression in peripheral blood cells. Based on this observation and our results, we can speculate that decrease TET2 expression in peripheral blood cells is required to induce a pro-inflammatory phenotype and cardiovascular disorders.
The PAH Biobank used the REVEAL relaxed criteria for enrollment of patients deemed to have Group 1 disease by their PH specialist. This allowed enrollment of patients with a PCWP ≤18 mmHg, which is the case for patient 12–20650. While patient 29–016 had a PCWP of 17mmHg, which is above the conventional cut-off of 15mmHg, as defined by the World Symposium on Pulmonary Hypertension1, this likely reflects the technical heterogeneity of PCWP measurements, rather than clinical misclassification. This patient’s PAH physician integrated all data to make the best clinical diagnosis.
We provided the hemodynamic data obtained in closest temporal proximity to enrollment in the biobank. In most cases these were the diagnostic hemodynamics, obtained prior to treatment; however in one patient (19–036), the low PVR we reported reflected the fact they were already being treated with two PH-targeted medications. At the time of their diagnosis, prior to PH-targeted treatment, their hemodynamics were typical of PAH (mPAP of 67mmHg and a PVR of 9.96WU).
The use of gnomAD as a control cohort can be consider as a limitation of the study. GnomAD spans 15708 genomes from unrelated individuals sequenced as part of various disease-specific and population genetic studies. GnomAD excludes individuals known to be affected by any severe pediatric disease, as well as their first-degree relatives; however, some individuals with severe disease may still be included in the data set, albeit likely at a frequency equivalent to or lower than that seen in the general population. Thus, the 7509 non-Finnish European gnomAD cohort used in our study likely includes individuals with severe diseases, potentially including hematologic disorders or atherosclerosis. This limitation could explain the 6 patients with TET2 variants observed in the gnomAD cohort. Regardless of this limitation we observed a 5.5-fold enrichment of TET2 deleterious variants in the PAH cohort.
Conclusion.
We identify TET2 as a new PAH-associated gene and highlight its importance as a potential mechanism for the component of PAH pathogenesis resulting from inflammation. The TET2 pathway offers potential new biomarkers and therapeutic targets for PAH therapy, including canakinumab.
Supplementary Material
Clinical Perspective.
What is new:
TET2, encoding an epigenetic regulator that demethylates cytosine, is found to be mutated in both idiopathic pulmonary hypertension (IPAH) and associated PAH (APAH)
TET2 expression is ubiquitously decreased in peripheral blood cells of both IPAH and APAH patients
TET2 depletion creates a pro-inflammatory phenotype
We present a new preclinical model of PAH, the Tet2 −/− mouse
Il-1β blockade improves PAH in mice with Tet2 depletion
What are the clinical implications:
TET2 expression might represent a potential PAH biomarker
TET2 mutation(s) are a risk factor for PAH.
The role of TET2 mutations in clonal hematopoiesis of indeterminant potential (CHIP) and PAH suggest a relationship between PAH and hematopoietic disorders.
TET2 mutation(s) are a potential target for personalized medicine and potential indicator of benefit from anti-inflammatory therapies in PAH.
Acknowledgement.
The authors acknowledge the generous gift of mouse IL-1β surrogate antibody and the control antibody from Novartis, Switzerland, Basel. The authors would like to thank Christina Ferrone for her contribution to the mouse care. The authors would like to thank staff of the animal facility for their contribution to the mouse care. The authors acknowledge the technical and scientific support of the Queen’s Cardiopulmonary Unit (QCPU), which was instrumental in performing these studies. Samples and/or Data from the National Biological Sample and Data Repository for PAH, which receives government support under an investigator-initiated grant (R24 HL105333) awarded by the National Heart Lung and Blood Institute (NHLBI), were used in this study. We thank contributors, including the Pulmonary Hypertension Centers who collected samples used in this study, as well as patients and their families, whose help and participation made this work possible. We thank and acknowledge the contribution of Russel Hirsch MD, Michelle Cash, S. Melissa Magness, Mukta Barve, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States; R. James White MD, PhD, Alison Light, Alison Theuer, University of Rochester Medical Center, Rochester NY, United States; Marc Simon MD, Traci McGaha, University of Pittsburgh, Pittsburgh PA, United States; David Badesch MD, Holly del Junco, Lisa Nicotera, Kelly Hannon, University of Colorado Denver, Aurora CO, United States; Erika Rosenzweig MD, Daniela Brady, Columbia University, New York NY, United States; Charles Burger MD, Inna Abrea, Andrea Tavlarides, Mayo Clinic Florida, Jacksonville FL, United States; Murali Chakinala MD, Sharon Heuerman, Washington University, St. Louis MO, United States; Thenappan Thenappan MD, Gretchen Peichel, Gina Paciotti, Brenda Vang, University of Minnesota, Minneapolis MN, United States; Greg Elliott MD, David Tomer, Quinn Montgomery, Department of Medicine at Intermountain Medical Center and the University of Utah, Murray UT, United States; Hap Farber, MD, Robert Simms MD, Eric Stratton, Boston University School of Medicine, Boston MA, United States; Robert Frantz MD, Louise Durst, Kristal Rohwer, Mayo Clinic, Rochester MN, United States; Jean Elwing MD, Tammy Roads, Autumn Studer, University of Cincinnati, Cincinnati OH, United States; Nicholas Hill MD, Karen Visnaw, Tufts Medical Center, Boston MA, United States; Dunbar Ivy MD, Kathleen Miller-Reed, Karlise Lewis, Children’s Hospital of Colorado, University of Colorado Denver, Aurora CO, United States; James Klinger MD, Amy Palmisciano, Meghan Ahearn, Rhode Island Hospital, Providence RI, United States; Steven Nathan MD, Merte Lemma, Inova Heart and Vascular Institute, Falls Church VA, United States; Ronald Oudiz MD, Joy Beckmann, Bindu John, LA Biomedical Research Institute at Harbor-UCLA, Torrance CA, United States; Ivan Robbins MD, Shannon Cordell, Vanderbilt University Medical Center, Nashville TN, United States; Robert Schilz DO, PhD, Mary Andrews, University Hospital of Cleveland, Cleveland OH, United States; Terry Fortin MD, Karla Kennedy, Susana Almeida-Peters, Duke University Medical Center, Durham NC, United States; Jeffrey Wilt MD, Kimberly McClain, Spectrum Health Hospitals, Grand Rapids MI, United States; Delphine Yung MD, Anne Davis, Linnea Brody, Seattle Children’s Hospital, Seattle WA, United States; Eric Austin MD, Karen Chaffin, Vanderbilt University-Peds, Nashville TN, United States; Ferhaan Ahmad MD, PhD, Page Scovel, Division of Cardiovascular Medicine, University of Iowa, Iowa City IA, United States; Nitin Bhatt MD, Joseph Santiago, Ohio State University, Columbus OH, United States; Tim Lahm MD, Jennifer Marks, Debra Broach, Indiana University, Indianapolis IN, United States; Adaani Frost MD, Zeenat Safdar MD, Jennifer Lee, Royanne Holy, Weill Cornell Medical College and The Houston Methodist Hospital, Houston TX, United States; Zia Rehman MD, Anagha Malur, East Carolina University, Greenville NC, United States; Robert Walter MD, Tracy Norwood, Donna Singleton, LSU Health, Shreveport LA, United States; Fernando Torres MD, Oluwatosin Igenoza, UT Southwestern, Dallas TX, United States; Sahil Bakshi DO, Audrey Anderson, Renesa Whitman, Natalia Feliz, Baylor Research Institute, Plano TX, United States; Stephen Archer MD, Lindsey Hawke, Mark Ormiston, Queen’s University, Kingston ON, Canada; Rahul Argula MD, Courtney Rowley, Daniel Larimore, Jill Spears, Medical University of South Carolina, Charleston SC, United States; Christopher Barnett MD, Sara Ahmed, Hellina Birru, Medstar Health, Washington D.C., United States; Raymond Benza MD, Priscilla Correa, Allegheny-Singer Research Institute, Pittsburgh PA, United States. Exome sequencing and genotyping data were generated by the Regeneron Genetics Center for the CUMC-CCHMC-RGC collaboration. We thank Mohamad Reza Afghah of Queen’s University for his assistance in statistical analysis. Finally, this work was supported by funding from the Vanier Scholarship Program to EKC, Ontario Institute for Cancer Research and Southeastern Academic Medical Organization to MJR, SLA and his research are supported by a CIHR Foundation Grant (NIH-RO1-HL071115, 1RC1HL099462), a Tier 1 Canada Research Chair in Mitochondrial Dynamics, the William J Henderson Foundation, the CIHR Vascular Network, the Canadian Vascular Network, and the Queen’s Cardiopulmonary Unit (QCPU)
Source of funding.
This study was supported in part by U.S. National Institutes of Health (NIH) grants NIH 1R01HL113003–01A1 (S.L.A.), NIH 2R01HL071115–06A1 (S.L.A), NIH R24HL105333 (W.C.N., M.W.P., K.A.L., A.W.C), Canada Foundation for Innovation and the Queen’s Cardiopulmonary Unit (QCPU) 229252 and 33012 (S.L.A.), Tier 1 Canada Research Chair in Mitochondrial Dynamics and Translational Medicine 950–229252 (S.L.A.), Canadian Institutes of Health Research (CIHR) Foundation Grant CIHR FDN 143261, the William J. Henderson Foundation (S.L.A.), and Canadian Vascular Network Scholar Award (L.T.; F.P.), and the JPB Foundation (W.K.C.), and Paroian Family PH Research Scholarship from the pulmonary hypertension association of Canada (F.P.). Further support was provided by a Vanier Canada Graduate Scholarship and CIHR MD/PhD Studentship (E.C.), an Ontario Molecular Pathology Research Network (OMPRN)/ Ontario Institute for Cancer Research (OICR) Cancer Pathology Translational Research Grant, and a Canada Foundation for Innovation Grant (M.J.R.).
Footnotes
Disclosures. None
References.
- 1.Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, Williams PG, Souza R. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J. 2019;53:1801913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Benza RL, Miller DP, Barst RJ, Badesch DB, Frost AE, McGoon MD. An Evaluation of Long-term Survival From Time of Diagnosis in Pulmonary Arterial Hypertension From the REVEAL Registry. Chest. 2012;142:448–456. [DOI] [PubMed] [Google Scholar]
- 3.Wijeratne DT, Lajkosz K, Brogly SB, Lougheed MD, Jiang L, Housin A, Barber D, Johnson A, Doliszny KM, Archer SL. Increasing Incidence and Prevalence of World Health Organization Groups 1 to 4 Pulmonary Hypertension. Circ Cardiovasc Qual Outcomes. 2018;11:e003973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Thenappan T, Ormiston ML, Ryan JJ, Archer SL. Pulmonary arterial hypertension: pathogenesis and clinical management. BMJ. 2018;360:j5492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Lane KB, Machado RD, Pauciulo MW, Thomson JR, Phillips JA, Loyd JE, Nichols WC, Trembath RC, Trembath RC. Heterozygous germline mutations in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat Genet. 2000;26:81–4. [DOI] [PubMed] [Google Scholar]
- 6.Rudarakanchana N, Flanagan JA, Chen H, Upton PD, Machado R, Patel D, Trembath RC, Morrell NW. Functional analysis of bone morphogenetic protein type II receptor mutations underlying primary pulmonary hypertension. Hum Mol Genet. 2002;11:1517–25. [DOI] [PubMed] [Google Scholar]
- 7.Morrell NW, Aldred MA, Chung WK, Elliott CG, Nichols WC, Soubrier F, Trembath RC, Loyd JE. Genetics and genomics of pulmonary arterial hypertension. Eur Respir J. 2018;1801899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Archer SL, Marsboom G, Kim GH, Zhang HJ, Toth PT, Svensson EC, Dyck JRB, Gomberg-Maitland M, Thébaud B, Husain AN et al. Epigenetic attenuation of mitochondrial superoxide dismutase 2 in pulmonary arterial hypertension: a basis for excessive cell proliferation and a new therapeutic target. Circulation. 2010;121:2661–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Potus F, Ruffenach G, Dahou A, Thebault C, Breuils-Bonnet S, Tremblay È, Nadeau V, Paradis R, Graydon C, Wong R et al. Downregulation of miR-126 Contributes to the Failing Right Ventricle in Pulmonary Arterial Hypertension. Circulation. 2015;132:932–43. [DOI] [PubMed] [Google Scholar]
- 10.Smith ZD, Meissner A. DNA methylation: roles in mammalian development. Nat Rev Genet. 2013;14:204–220. [DOI] [PubMed] [Google Scholar]
- 11.Leoni C, Montagner S, Rinaldi A, Bertoni F, Polletti S, Balestrieri C, Monticelli S. Dnmt3a restrains mast cell inflammatory responses. Proc Natl Acad Sci U S A. 2017;114:E1490–E1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Cull AH, Snetsinger B, Buckstein R, Wells RA, Rauh MJ. Tet2 restrains inflammatory gene expression in macrophages. Exp Hematol. 2017;55:56–70.e13. [DOI] [PubMed] [Google Scholar]
- 13.Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, McConkey M, Gupta N, Gabriel S, Ardissino D et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med. 2017;377:111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Libby P Mechanisms of Acute Coronary Syndromes and Their Implications for Therapy. N Engl J Med. 2013;368:2004–2013. [DOI] [PubMed] [Google Scholar]
- 15.Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, Chambert K, Mick E, Neale BM, Fromer M et al. Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence. N Engl J Med. 2014;371:2477–2487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, Lindsley RC, Mermel CH, Burtt N, Chavez A et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371:2488–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhu N, Gonzaga-Jauregui C, Welch CL, Ma L, Qi H, King AK, Krishnan U, Rosenzweig EB, Ivy DD, Austin ED et al. Exome Sequencing in Children With Pulmonary Arterial Hypertension Demonstrates Differences Compared With Adults. Circ Genomic Precis Med. 2018;11:e001887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zhu N, Pauciulo MW, Welch CL, Lutz KA, Coleman AW, Gonzaga-Jauregui C, Wang J, Grimes JM, Martin LJ, He H et al. Novel risk genes and mechanisms implicated by exome sequencing of 2,572 individuals with pulmonary arterial hypertension. bioRxiv. 2019;550327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Srivastava AK, Wang Y, Huang R, Skinner C, Thompson T, Pollard L, Wood T, Luo F, Stevenson R, Polimanti R et al. Human genome meeting 2016 : Houston, TX, USA. 28 February - 2 March 2016. Hum Genomics. 2016;10 Suppl 1:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164–e164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lek M, Karczewski KJ, Minikel EV., Samocha KE, Banks E, Fennell T, O’Donnell-Luria AH, Ware JS, Hill AJ, Cummings BB et al. Exome Aggregation Consortium. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536:285–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ioannidis NM, Rothstein JH, Pejaver V, Middha S, McDonnell SK, Baheti S, Musolf A, Li Q, Holzinger E, Karyadi D et al. REVEL: An Ensemble Method for Predicting the Pathogenicity of Rare Missense Variants. Am J Hum Genet. 2016;99:877–885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R, 1000 Genome Project Data Processing Subgroup. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cheadle C, Berger AE, Mathai SC, Grigoryev DN, Watkins TN, Sugawara Y, Barkataki S, Fan J, Boorgula M, Hummers L et al. Erythroid-specific transcriptional changes in PBMCs from pulmonary hypertension patients. PLoS One. 2012;7:e34951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Moran-Crusio K, Reavie L, Shih A, Abdel-Wahab O, Ndiaye-Lobry D, Lobry C, Figueroa ME, Vasanthakumar A, Patel J, Zhao X et al. Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell. 2011;20:11–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Garcia-Rivas G, Jerjes-Sánchez C, Rodriguez D, Garcia-Pelaez J, Trevino V. A systematic review of genetic mutations in pulmonary arterial hypertension. BMC Med Genet. 2017;18:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Rhodes CJ, Batai K, Bleda M, Haimel M, Southgate L, Germain M, Pauciulo MW, Hadinnapola C, Aman J, Girerd B et al. Genetic determinants of risk in pulmonary arterial hypertension: international genome-wide association studies and meta-analysis. Lancet Respir Med. 2018;7:227–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Montani D, Savale L, Natali D, Jais X, Herve P, Garcia G, Humbert M, Simonneau G, Sitbon O. Long-term response to calcium-channel blockers in non-idiopathic pulmonary arterial hypertension. Eur Heart J. 2010;31:1898–1907. [DOI] [PubMed] [Google Scholar]
- 30.Hanley JA, McNeil BJ. A Method of Comparing the Areas under Receiver Operating Characteristic Curves Derived from the Same Cases’. 1983. [DOI] [PubMed]
- 31.Zhang Q, Zhao K, Shen Q, Han Y, Gu Y, Li X, Zhao D, Liu Y, Wang C, Zhang X et al. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature. 2015;525:389–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hautefort A, Chesné J, Preussner J, Pullamsetti SS, Tost J, Looso M, Antigny F, Girerd B, Riou M, Eddahibi S et al. Pulmonary endothelial cell DNA methylation signature in pulmonary arterial hypertension. Oncotarget. 2017;8:52995–53016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhang X, Su J, Jeong M, Ko M, Huang Y-H, Park HJ, Guzman A, Lei Y, Huang Y-H, Rao A, Li W, Goodell MA. DNMT3A and TET2 compete and cooperate to repress lineage-specific transcription factors in hematopoietic stem cells. Nat Genet. 2016;48:1014–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sano S, Oshima K, Wang Y, Katanasaka Y, Sano M, Walsh K. CRISPR-Mediated Gene Editing to Assess the Roles of Tet2 and Dnmt3a in Clonal Hematopoiesis and Cardiovascular Disease. Circ Res. 2018;123:335–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fuster JJ, MacLauchlan S, Zuriaga MA, Polackal MN, Ostriker AC, Chakraborty R, Wu C-L, Sano S, Muralidharan S, Rius C et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science (80-). 2017;355:842–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rabinovitch M, Guignabert C, Humbert M, Nicolls MR. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res. 2014;115:165–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cook EK, Izukawa T, Young S, Rosen G, Jamali M, Zhang L, Johnson D, Bain E, Hilland J, Ferrone CK et al. Comorbid and inflammatory characteristics of genetic subtypes of clonal hematopoiesis. Blood Adv. 2019;3:2482–2486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Chung L, Farber HW, Benza R, Miller DP, Parsons L, Hassoun PM, McGoon M, Nicolls MR, Zamanian RT. Unique predictors of mortality in patients with pulmonary arterial hypertension associated with systemic sclerosis in the REVEAL registry. Chest. 2014;146:1494–1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gräf S, Haimel M, Bleda M, Hadinnapola C, Southgate L, Li W, Hodgson J, Liu B, Salmon RM, Southwood M, Machado RD et al. Identification of rare sequence variation underlying heritable pulmonary arterial hypertension. Nat Commun. 2018;9:1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sales VM, Ferguson-Smith AC, Patti M-E. Cell Metabolism Perspective Epigenetic Mechanisms of Transmission of Metabolic Disease across Generations. Cell Metab. 2017;25:559–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Khetarpal SA, Qamar A, Bick AG, Fuster JJ, Kathiresan S, Jaiswal S, Natarajan P. Clonal Hematopoiesis of Indeterminate Potential Reshapes Age-Related CVD: JACC Review Topic of the Week. J Am Coll Cardiol. 2019;74:578–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Steensma DP. Clinical consequences of clonal hematopoiesis of indeterminate potential. Blood Adv. 2018;2:3404–3410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD et al. CANTOS Trial Group. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med. 2017;377:1119–1131. [DOI] [PubMed] [Google Scholar]
- 44.Groth A, Vrugt B, Brock M, Speich R, Ulrich S, Huber LC. Inflammatory cytokines in pulmonary hypertension. Respir Res. 2014;15:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kqiku X, Kovacs G, Reitter S, Sill H, Olschewski H. Pulmonary Hypertension May Be a Relevant Comorbidity in Patients with Myelodysplastic Syndromes. Blood. 2011;118. [Google Scholar]
- 46.Sill H, Kqiku-Kryeziu X, Avian A, Kovacs G, Gaal S, Zebisch A, Olschewski H. Pulmonary arterial pressure in patients with myelodysplastic syndromes. Leuk Lymphoma. 2016;57:2723–2726. [DOI] [PubMed] [Google Scholar]
- 47.Slattery ML, John EM, Torres-Mejia G, Herrick JS, Giuliano AR, Baumgartner KB, Hines LM, Wolff RK. Genetic variation in bone morphogenetic proteins and breast cancer risk in hispanic and non-hispanic white women: The breast cancer health disparities study. Int J Cancer. 2013;132:2928–2939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ley TJ, Ding L, Walter MJ, McLellan MD, Lamprecht T, Larson DE, Kandoth C, Payton JE, Baty J, Welch J et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363:2424–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kaasinen E, Kuismin O, Rajamäki K, Ristolainen H, Aavikko M, Kondelin J, Saarinen S, Berta DG, Katainen R, Hirvonen EAM et al. Impact of constitutional TET2 haploinsufficiency on molecular and clinical phenotype in humans. Nat Commun. 2019;10:1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.McGoon MD, Miller DP. REVEAL: a contemporary US pulmonary arterial hypertension registry. Eur Respir Rev. 2012;21:8–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.