Fidler
TP
,
Xue
C
,
Yalcinkaya
M
, et al
.
The AIM2 inflammasome exacerbates atherosclerosis in clonal haematopoiesis
.
Nature
.
2021
;
592
(
7853
):
296
301
. .

Clonal hematopoiesis (CH) is defined as the presence of an expanded somatic blood-cell clone in persons without other hematologic abnormalities. The presence of CH increases with age and is usually present in 10 to 20 percent of people older than 70 years.1  The most common myeloid mutations associated with CH are TET2, ASXL1, DNMT3A, and JAK2. Carriers of CH have an approximately twofold increased risk of atherosclerotic cardiovascular disease (CVD).1,2  Mouse models of TET2 CH have shown increased macrophage inflammation and atherosclerosis, heightened IL-1β/IL-6 signaling, and also that inhibition of the NLRP3 inflammasome (that activates IL-1β, a key inducer of IL-6) ameliorates atherosclerosis.1,3  These observations suggest that inhibiting the IL-6/IL-1β pathway may be effective in reducing CVD risk in humans with CH.1,3  This is of particular interest as a trial of anti-IL-1ß (canakinumab) found up to 15 percent reduction in cardiovascular events; however, whether response was dependent on the presence of an underlying CH mutation is unknown. Furthermore, whether different myeloid mutations increase the risk of thrombosis through different mechanisms is also unknown.

JAK2 V617F mutation (JAK2VF) –positive CH is associated with a particularly high risk of premature coronary artery disease. This is of particular relevance as JAK2VF-driven myeloproliferative neoplasms are also associated with a significantly increased risk of thrombosis and vascular disease, which remains the major cause of morbidity and early mortality in these conditions (thrombosis risk, 2.5-10.9% per annum).4,5  Although thrombosis risk is highest around the time of initial diagnosis, over time the risk of arterial thrombosis remains about twofold higher, and this risk is relatively higher for younger people with myeloproliferative neoplasms.6  The JAK2VF driver mutation results in constitutive activation of JAK2 and downstream activation of the signal transducer and activator of transcription (STAT) 1, 3, and 5; mitogen-activated protein kinase; and phosphoinositide 3-kinase pathways. Exactly how this may result in increased atherosclerosis and thrombosis is not yet well understood. However, it is noteworthy that although individual randomized controlled trials of ruxolitinib, a tyrosine kinase inhibitor that inhibits JAK1 and JAK2, have not been powered to study thromboembolic events, preliminary evidence suggests that ruxolitinib treatment may reduce thromboembolic events.7,8 

To understand the mechanism by which the JAK2VF mutation might lead to an increased risk of vascular disease, Dr. Trevor Fidler and colleagues first studied gene expression changes associated with JAK2VF expression in splenic myeloid cells from wildtype or hyperlipidemic (Ldlr-/-) mice. This analysis revealed enrichment of genes associated with cellular proliferation, DNA damage repair, and metabolic pathways associated with presence of JAK2VF mutation in myeloid cells. To specifically model the impact of myeloid cell–specific CH, chimeric mice were generated where JAK2VF was expressed in only a proportion of blood cells, including a series of elegant experiments using an array of different mouse models, which allow induction of the JAK2VF mutation in a myeloid cell–specific manner (i.e., only in neutrophils or monocytes). Mice in which JAK2VF was expressed specifically in neutrophils showed no changes in atherosclerotic plaque area or morphology in hyperlipidemic mice. In contrast, both the chimeric mice modelling CH and mice that expressed JAK2VF selectively in macrophages showed increased atherosclerotic plaque area with proliferation of macrophages and increased necrotic cores. These atherosclerotic lesions showed increased expression of the inflammasome gene Aim2 as well as oxidative DNA damage and DNA replication stress. Single-cell RNA sequencing analysis of JAK2VF lesions demonstrated anenriched inflammatory myeloid cell landscape. Even though both Nlrp3 and AIM2 inflammasomes could be activated in cultured JAK2VF macrophages, Nlrp3 deficiency had no significant effect on the lesions or necrotic cores, whereas Aim2 deficiency markedly reduced both (in contrast to the observations made in Tet2 deficient-mice mentioned earlier). Thus, the AIM2 inflammasome was the key pathway promoting atherosclerotic lesion formation in these JAK2VF mice, possibly reflecting increased expression of AIM2 downstream of JAK/STAT signaling. Deletion of the downstream inflammasome components caspase 1 and 11 or pyroptosis executioner gasdermin D also partially reversed the changes and increased the stability of the plaques. These findings suggest that JAK2VF mutations drive increased atherosclerosis primarily through the impact on macrophages, by increased proliferation, glycolytic metabolism, DNA replication stress, and activation of the AIM2 inflammasome, and increased IL-1ß. The group went on to study potential therapeutic interventions. Ruxolitinib treatment was associated with slightly smaller atherosclerotic lesions, but concerningly, they had increased necrotic cores and thinner caps, suggesting less-stable plaques and indicating that ruxolitinib promoted lesional necrosis. In contrast, inhibition of IL-1 or the inflammasome product IL-1ß reduced macrophage proliferation and necrotic core formation while increasing the thickness of fibrous caps, indicating plaque stabilization.

By generating a JAK2VF mouse model that allows controlled expression of JAK2 in neutrophils and monocytes as well as a chimeric model of CH, Dr. Fidler and colleagues demonstrated that the expression of JAK2VF specifically in monocytes is critical for driving accelerated atherosclerosis. Atherosclerotic lesions are enriched with JAK2VF macrophages that exhibit increased proliferation markers, increased necrotic cores, and reduced thickness of fibrous caps, indicating less-stable plaques. Their results demonstrate that the AIM2 inflammasome and IL-1β signaling are crucial for this process. In contrast, it has previously been reported that in mice with Tet2 deficiency, increased lesion formation was not associated within macrophage proliferation within plaques and was reversed using an inhibitor of the NLRP3 inflammasome.3  Taken together, these results suggest that distinct mechanisms promote atherosclerosis in CH associated with different myeloid mutations. In developing new therapies for atherosclerotic CVD, broad inhibition of IL-1, or its downstream mediator IL-6, may help slow the accelerated atherosclerosis associated with CH, although inhibition of upstream targets such as NLRP3 and AIM2 inflammasomes may need to be targeted depending on the precise underlying myeloid mutation.

Dr. Shapiro and Dr. Mead indicated no relevant conflicts of interest. Dr. Shapiro and Dr. Mead are supported by the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, and Dr. Shapiro is funded by the Medical Research Council (MR/ T024054/1).

1.
Jaiswal
S
,
Natarajan
P
,
Silver
AJ
, et al
.
Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease
.
N Engl J Med
.
2017
;
377
(
2
):
111
121
.
2.
Bick
AG
,
Weinstock
JS
,
Nandakumar
SK
, et al
.
Inherited causes of clonal haematopoiesis in 97,691 whole genomes
.
Nature
.
2020
;
586
(
7831
):
763
768
.
3.
Fuster
JJ
,
MacLauchlan
S
,
Zuriaga
MA
, et al
.
Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice
.
Science
.
2017
;
355
(
6327
):
842
847
.
4.
Landolfi
R
,
Marchioli
R
,
Kutti
J
, et al
.
Efficacy and safety of low-dose aspiring in polycythemia vera
.
N Engl J Med
.
2004
;
350
(
2
):
114
124
.
5.
Tefferi
A
,
Rumi
E
,
Finazzi
G
, et al
.
Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study
.
Leukemia
.
2013
;
27
(
9
):
1874
1881
.
6.
How
J
,
Story
C
,
Connors
JM
, et al
.
Prevention of recurrent thromboembolism in myeloproliferative neoplasms: review of literature and focus on direct oral anticoagulants
.
Postgrad Med
.
2021
;
1
9
.
7.
Verstovsek
S
,
Passamonti
F
,
Rambaldi
A
, et al
.
A phase 2 study of ruxolitinib, an oral JAK1 and JAK2 inhibitor, in patients with advanced polycythemia vera who are refractory or intolerant to hydroxyurea
.
Cancer
.
2014
;
120
(
4
):
513
520
.
8.
Masciulli
A
,
Ferrari
A
,
Carobbio
A
, et al
.
Ruxolitinib for the prevention of thrombosis in polycythemia vera: a systematic review and meta-analysis
.
Blood Adv
.
2020
;
4
(
2
):
380
386
.