scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Clonal hematopoiesis associated with Tet2 deficiency accelerates atherosclerosis development in mice

TL;DR: The effects of the expansion of Tet2-mutant cells in atherosclerosis-prone, low-density lipoprotein receptor–deficient mice are studied and it is found that partial bone marrow reconstitution with TET2-deficient cells was sufficient for their clonal expansion and led to a marked increase in Atherosclerotic plaque size.
Abstract: Human aging is associated with an increased frequency of somatic mutations in hematopoietic cells. Several of these recurrent mutations, including those in the gene encoding the epigenetic modifier enzyme TET2, promote expansion of the mutant blood cells. This clonal hematopoiesis correlates with an increased risk of atherosclerotic cardiovascular disease. We studied the effects of the expansion of Tet2-mutant cells in atherosclerosis-prone, low-density lipoprotein receptor–deficient (Ldlr–/–) mice. We found that partial bone marrow reconstitution with TET2-deficient cells was sufficient for their clonal expansion and led to a marked increase in atherosclerotic plaque size. TET2-deficient macrophages exhibited an increase in NLRP3 inflammasome–mediated interleukin-1β secretion. An NLRP3 inhibitor showed greater atheroprotective activity in chimeric mice reconstituted with TET2-deficient cells than in nonchimeric mice. These results support the hypothesis that somatic TET2 mutations in blood cells play a causal role in atherosclerosis.

Content maybe subject to copyright    Report

Clonal hematopoiesis associated with TET2 deficiency
accelerates atherosclerosis development in mice
José J. Fuster
1,*
, Susan MacLauchlan
1
, María A. Zuriaga
1
, Maya N. Polackal
1
, Allison C.
Ostriker
2
, Raja Chakraborty
2
, Chia-Ling Wu
1
, Soichi Sano
1
, Sujatha Muralidharan
1
, Cristina
Rius
3
, Jacqueline Vuong
1
, Sophia Jacob
1
, Varsha Muralidhar
1
, Avril A. B. Robertson
4
,
Matthew A. Cooper
4
, Vicente Andrés
3
, Karen K. Hirschi
5
, Kathleen A. Martin
2
, and Kenneth
Walsh
1,*
1
Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine,
Boston, MA 02118, USA.
2
Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and
Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT
06511, USA.
3
Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) and CIBER de
Enfermedades Cardiovasculares, Madrid, Spain.
4
Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland,
Australia.
5
Yale Cardiovascular Research Center and Yale Stem Cell Center, Yale University School of
Medicine, New Haven, CT 06511, USA.
Abstract
Human aging is associated with an increased frequency of somatic mutations in hematopoietic
cells. Several of these recurrent mutations, including those in the gene encoding the epigenetic
modifier enzyme TET2, promote expansion of the mutant blood cells. This clonal hematopoiesis
correlates with an increased risk of atherosclerotic cardiovascular disease. We studied the effects
of the expansion of
Tet2
-mutant cells in atherosclerosis-prone, low-density lipoprotein receptor–
deficient (
Ldlr
−/−
) mice. We found that partial bone marrow reconstitution with TET2-deficient
cells was sufficient for their clonal expansion and led to a marked increase in atherosclerotic
plaque size. TET2-deficient macrophages exhibited an increase in NLRP3 inflammasome–
mediated interleukin-1β secretion. An NLRP3 inhibitor showed greater atheroprotective activity in
chimeric mice reconstituted with TET2-deficient cells than in nonchimeric mice. These results
support the hypothesis that somatic
TET2
mutations in blood cells play a causal role in
atherosclerosis.
*
Corresponding author. jjfuster@bu.edu (J.J.F.); kxwalsh@bu.edu (K.W.).
SUPPLEMENTARY MATERIALS
www.sciencemag.org/content/355/6327/842/suppl/DC1
HHS Public Access
Author manuscript
Science
. Author manuscript; available in PMC 2017 August 03.
Published in final edited form as:
Science
. 2017 February 24; 355(6327): 842–847. doi:10.1126/science.aag1381.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Cardiovascular disease (CVD) is the leading cause of death in the elderly, but almost 60% of
elderly patients with atherosclerotic CVD have either no conventional risk factors (e.g.,
hypertension, hypercholesterolemia, etc.) or just one risk factor (1). Furthermore, increasing
evidence suggests that most middle-aged individuals at low risk of CVD, based on
conventional risk factors, exhibit subclinical atherosclerosis (2, 3). These clinical data
suggest that unidentified age-dependent risk factors contribute to the development of CVD.
The accumulation of somatic DNA mutations is a hallmark of aging, particularly in
proliferating tissues, which over time may become a mosaic of cells with different genotypes
due to the clonal expansion of single de novo mutations (4). However, though human studies
suggest that somatic mutations may be associated with a broad spectrum of human disease
(5–7), there is little information on the potential causal role of somatic mutations in age-
associated disorders other than cancer. Recent human studies have shown that normal aging
is associated with an increased frequency of somatic mutations in the hematopoietic system,
which provide a competitive growth advantage to the mutant cell and allow its progressive
clonal expansion (clonal hematopoiesis) (7–11). This acquired clonal mosaicism in the
hematopoietic system of healthy individuals correlates with an increased risk of subsequent
hematologic cancer (7–9), but it has also been associated with higher prevalence of vascular
complications of diabetes, greater incidence of atherosclerotic conditions (i.e., coronary
heart disease, stroke), and increased frequency of CVD-related deaths (6, 7). Although these
human studies suggest an unexpected connection between somatic mutations in
hematopoietic cells, clonal hematopoiesis, and atherosclerosis, their descriptive nature does
not allow cause-effect relationships, or even directionality, to be established.
Most of the reported somatic mutations associated with age-related clonal hematopoiesis
occur in a small number of genes encoding epigenetic regulators (7–10). The present study
focuses on one of these genes,
TET2
(ten-eleven translocation 2), the first gene reported to
exhibit somatic mutations in blood cells in individuals with clonal hematopoiesis without
hematological malignancies (10). More than 70 different mutations have been reported in
this gene (7–10, 12). The protein encoded by
TET2
is an epigenetic regulatory enzyme that
catalyzes the oxidation of 5-methylcytosine (5mc) in DNA to 5-hydroxymethylcytosine
(5hmc) and also exerts noncatalytic actions. TET2 modulates hematopoietic stem and
progenitor cell (HSPC) self-renewal (13–16), but its role in CVD remains largely
unexplored.
To mimic the human scenario of clonal hematopoiesis and test whether clonal expansion of
TET2-deficient hematopoietic cells contributes to atherosclerosis, we used a competitive
bone marrow transplantation (BMT) strategy to generate atherosclerosis-prone, low-density
lipoprotein receptor–deficient (
Ldlr
−/−
) chimeric mice with a small proportion of TET2-
deficient HSPCs. Lethally irradiated
Ldlr
−/−
recipients were transplanted with suspensions
of bone marrow (BM) cells containing 10%
Tet2
−/−
cells and 90%
Tet2
+/+
cells [10%
knockout (KO)– BMT mice] and then fed a normal diet (ND) or a high-fat/high-cholesterol
(HFHC) diet for 9 weeks to induce atherosclerosis development (fig. S1). To distinguish
donor
Tet2
−/−
and
Tet2
+/+
cells in this experimental setting,
Tet2
+/+
cells were obtained from
mice carrying the CD45.1 variant of the CD45 hematopoietic antigen, whereas
Tet2
−/−
cells
were obtained from mice carrying the CD45.2 variant of this protein. Control mice [10%
Fuster et al. Page 2
Science
. Author manuscript; available in PMC 2017 August 03.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

wild-type (WT)–BMT] were transplanted with 10% CD45.2
+
Tet2
+/+
cells and 90%
CD45.1
+
Tet2
+/+
cells. Flow cytometry analysis of CD45.2
+
blood cells established that this
BMT strategy led to the clonal expansion of
Tet2
−/−
hematopoietic cells to an extent
consistent with variant allelic fractions for somatic
TET2
mutations observed in human
studies linking clonal hematopoiesis to accelerated CVD (7). At the start of ND or HFHC
diet feeding (4 weeks after BMT), CD45.2
+
cells represented ~28% of blood cells in 10%
KO-BMT mice, and they expanded further over time, reaching 42% of blood cells 6 weeks
after BMT and 56% 12 weeks after BMT (Fig. 1, A and B). This clonal expansion of TET2-
deficient hematopoietic cells is similar to that observed when human cells carrying somatic
TET2
mutations are transplanted into immune-deficient mice (17).
TET2 ablation in CD45.2
+
cells of HFHC-fed 10% KO-BMT mice was confirmed by
quantitative real-time polymerase chain reaction (qRT-PCR) analysis of CD45.2
+
white
blood cell (WBC) fractions (Fig. 1C and fig. S2A). No changes were observed in the
expression of TET1 or TET3, two related epigenetic modulators. Consistent with the
enzymatic activity of TET2, ablation of the gene was paralleled by a decrease in 5hmC
levels in WBCs and macrophages (fig. S2, B and C). Although the absolute number of
HSPCs [defined as lineage
, Sca1
+
, c-Kit
+
(LSK) cells] was comparable between genotypes
(fig. S3A), CD45.2
+
cells represented 69% of LSK cells in the BM (fig. S3B) and 61% in
the spleen (fig. S3C) of 10% KO-BMT mice at 13 weeks post-BMT, consistent with
previous studies reporting that TET2 inactivation enhances HSPC self-renewal (13–16).
Transplanted
Tet2
−/−
BM cells expanded into all blood cell lineages, regardless of type of
diet, although with a slight myeloid bias and a reduced expansion into the T-lymphoid
lineage in the BM, spleen, and blood (Fig. 1D and fig. S3, D to F), in agreement with
previous studies with TET2-deficient mice (13–16). The expansion of
Tet2
−/−
HSPCs did
not affect blood cell counts (fig. S3G), consistent with findings in cancer-free individuals
carrying
TET2
mutations in blood cells (7, 10).
Having demonstrated that the competitive BMT strategy leads to the clonal expansion of
TET2-deficient HSPCs and mimics the human scenario of clonal hematopoiesis associated
with
TET2
mutations, we next evaluated whether the clonal expansion of TET2-deficient
HSPCs affects atherogenesis and related metabolic abnormalities. We observed no effects on
body weight (fig. S4A), spleen weight (fig. S4B), blood glucose levels (fig. S4C), systemic
insulin sensitivity (fig. S4D), or plasma cholesterol levels (fig. S4E). ND-fed mice developed
no aortic atherosclerosis, regardless of BM genotype (fig. S4F). In contrast, clonal expansion
of TET2-deficient BM cells had a profound effect on HFHC-induced atherosclerosis, as 10%
KO-BMT mice exhibited 60% larger plaques in the aortic root than did WT controls (Fig.
1E). Competitive BMT experiments with
Tet2
+/−
cells revealed that TET2 heterozygosity is
sufficient to accelerate atherosclerosis, despite the slower kinetics of TET2-heterozygous
cell expansion (fig. S5). Increased atherogenesis in 10% KO-BMT mice was paralleled by
an increase in total macrophage content in the intima, although this parameter was not
statistically significant when normalized to plaque size (fig. S6). BM genotype did not affect
lesional content of collagen or vascular smooth muscle cells (fig. S6), apoptosis (fig. S7A),
necrotic core extension (fig. S7B), or proliferation rates of total plaque cells or lesional
macrophages (fig. S7C). Overall, these data demonstrate that clonal expansion of TET2-
deficient hematopoietic cells accelerates atherogenesis in a manner independent of
Fuster et al. Page 3
Science
. Author manuscript; available in PMC 2017 August 03.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

alterations in systemic metabolism, changes in blood cell counts, or macrophage
proliferation or apoptosis in the plaque.
Consistent with their above-mentioned preferential differentiation into myeloid cells, TET2-
deficient HSPCs expanded preferentially into the macrophage population in the
atherosclerotic vascular wall. CD45.2
+
cells represented 58% of total immune cells, 62% of
macrophages, and 35% of T cells present in the aortic wall of 10% KO-BMT mice (Fig. 1, F
and G, and fig. S8). On the basis of these findings, we hypothesized that TET2-deficient
hematopoietic cells accelerate atherosclerosis mainly by generating a pool of macrophages
with enhanced proatherogenic activities. To test this possibility, we used BMT and LysM-
Cre/LoxP strategies to generate atherosclerosis-prone mice exhibiting TET2 deficiency
restricted to myeloid cells (Mye-
Tet2
-KO mice). Although this strategy led to a partial
(~80%) inactivation of
Tet2
in BM-derived macrophages (Fig. 2A), it was sufficient to
increase plaque size in the aortic root of HFHC-fed mice (Fig. 2B), with no differences in
body or spleen weight, blood monocyte counts, or glucose and cholesterol levels (fig. S9).
These results demonstrate that TET2 deficiency in myeloid cells is sufficient to promote
atherogenesis and suggest that macrophages play a major role in the accelerated
atherosclerosis associated with expansion of TET2-deficient HSPCs. However, they do not
rule out a potential contribution from other BM-derived cells. Analysis of aorta and aorta-
draining mediastinal lymph nodes showed that the expansion of TET2-deficient HSPCs does
not affect T cell numbers or aortic expression of T cell activation markers (fig. S10, A to C),
although it leads to modest changes in the frequency of various T cell subsets (fig. S10, D to
G), consistent with recent studies (18, 19). Such changes were not observed in Mye-
Tet2
-
KO mice (fig. S10, H to J). Therefore, although a contribution of TET2-deficient T cells to
the atherogenic effects of the expansion of TET2-deficient HSPCs cannot be excluded, these
data demonstrate that changes in T cells are not essential for the accelerated atherosclerosis
associated with TET2 loss of function and suggest instead a predominant role of
macrophages in this context.
We next evaluated the effects of TET2 deficiency on the function of macrophages in culture.
Consistent with the in vivo observations, TET2 deficiency did not affect macrophage
proliferation (fig. S11A), apoptosis (fig. S11B), oxidized low-density lipoprotein (oxLDL)
uptake (fig. S11C), or the expression of cholesterol trafficking regulators (fig. S11, D and E).
To evaluate whether TET2 deficiency affects proinflammatory macrophage activation, we
performed Affymetrix microarray analysis on
Tet2
−/−
macrophages and WT controls in
resting conditions and after treatment with a combination of lipopolysaccharide (LPS) and
interferon-γ (IFN-γ). Whereas no genes were differentially expressed in unstimulated
macrophages (
q
value < 0.05; fig. S12), a widespread alteration in gene expression was
found in
Tet2
−/−
macrophages after a 10-hour treatment with LPS/IFN-γ. Expression of 475
genes was altered by more than a factor of 1.5 when compared with WT macrophages (
q
<
0.05; Fig. 2C). PANTHER functional annotation software revealed that transcripts encoding
cytokine, chemokine, and signaling molecules were the top overrepresented classes altered
in the transcriptome of LPS/IFN-γ–treated TET2-deficient macrophages (Fig. 2D). Genes in
these classes with known proinflammatory actions were mostly up-regulated in TET2-
deficient macrophages (Fig. 2E). Consistent with this observation, qRT-PCR analysis
revealed that TET2-deficient macrophages exhibit markedly increased expression of
Fuster et al. Page 4
Science
. Author manuscript; available in PMC 2017 August 03.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

proinflammatory cytokines (Fig. 2F and fig. S13, A and B), chemokines (fig. S13C), and
enzymes (fig. S13D). This pattern of gene expression was also evident in
Tet2
+/−
macrophages and macrophages isolated from Mye-
Tet2
-KO mice (fig. S13, E and F). TET2
deficiency also resulted in increased interleukin-6 (IL-6) protein levels in macrophage
culture supernatants (fig. S13G). These data suggest that TET2 acts as a negative
transcriptional regulator of proinflammatory responses and are consistent with a previous
study reporting that TET2 represses LPS-induced IL-6 expression (20).
However, the situation in vivo in the atherosclerotic plaque is particularly complex, as
lesional macrophages are exposed to multiple signals simultaneously. Therefore, the anti-
inflammatory actions of TET2 in cultured macrophages were further evaluated by testing
their effect on macrophage response to a cocktail of low doses of oxLDL, tumor necrosis
factor (TNF), and IFN-γ, three stimuli present in atherosclerotic plaques. These conditions
minimized the impact of TET2 deficiency on cytokine and chemokine expression (fig. S14),
with the exception of IL-1β, which was markedly up-regulated in TET2-deficient
macrophages at all time points of oxLDL/TNF/IFN-γ stimulation (Fig. 3A). Supporting a
predominant role for IL-1β in the exacerbated atherosclerosis associated with TET2 loss of
function, gene expression analysis of the aortic arch of HFHC-fed mice revealed a
significant >twofold increase in transcript levels of IL-1β in 10% KO-BMT versus 10% WT-
BMT mice (Fig. 3B), whereas few significant differences were observed in the aortic
expression of other cytokines and chemokines or other macrophage-enriched genes (fig.
S15). 10% KO-BMT also exhibited increased IL-1β protein levels in atherosclerotic plaques
and plaque macrophages, as revealed by immunofluorescence staining coupled to confocal
microscopy (Fig. 3, C and D).
To examine the molecular mechanisms underlying the effects of TET2 deficiency on IL-1β
expression, we performed cell culture studies with LPS/IFN-γ–treated macrophages
transiently overexpressing either WT TET2 or a TET2 mutant unable to catalyze the
oxidation of 5mC to 5hmC. Both WT-TET2 and mutant-TET2 overexpression led to a ~90%
reduction in IL-1β expression and blunted differences between
Tet2
+/+
and
Tet2
−/−
macrophages (Fig. 3E), suggesting that TET2 modulates IL-1β expression independent of its
catalytic activity. Therefore, we investigated noncatalytic mechanisms of TET2-mediated
transcriptional repression. Consistent with previous studies showing that TET2 inhibits gene
transcription via histone deacetylase (HDAC)–mediated histone deacetylation (20),
treatment with the HDAC inhibitor trichostatin A (TSA) increased IL-1β expression in LPS/
IFN-γ–treated macrophages and abolished expression differences between TET2-deficient
and WT genotypes (Fig. 3F). Further supporting a role for histone deacetylation in TET2-
mediated repression of IL-1β, chromatin immunoprecipitation (ChIP)–qPCR analysis
revealed greater histone H3 acetylation at the
Il1b
gene promoter in TET2-deficient
macrophages (Fig. 3G). In contrast, treatment with TSA reduced the expression of
transcripts corresponding to other proinflammatory cytokines (such as IL-6), in agreement
with previous reports (21), and did not affect differences between genotypes (fig. S16A).
Furthermore, H3 acetylation at the
Il6
gene promoter was not affected by TET2 ablation
(fig. S16B). Overall, these data suggest that a reduction in HDAC-mediated histone
deacetylation accounts for the effects of TET2 loss of function on IL-1β expression in
macrophages, whereas alternative mechanisms contribute to its effects on other genes.
Fuster et al. Page 5
Science
. Author manuscript; available in PMC 2017 August 03.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Citations
More filters
Journal ArticleDOI
TL;DR: Antiinflammatory therapy targeting the interleukin‐1β innate immunity pathway with canakinumab at a dose of 150 mg every 3 months led to a significantly lower rate of recurrent cardiovascular events than placebo, independent of lipid‐level lowering.
Abstract: BackgroundExperimental and clinical data suggest that reducing inflammation without affecting lipid levels may reduce the risk of cardiovascular disease. Yet, the inflammatory hypothesis of atherothrombosis has remained unproved. MethodsWe conducted a randomized, double-blind trial of canakinumab, a therapeutic monoclonal antibody targeting interleukin-1β, involving 10,061 patients with previous myocardial infarction and a high-sensitivity C-reactive protein level of 2 mg or more per liter. The trial compared three doses of canakinumab (50 mg, 150 mg, and 300 mg, administered subcutaneously every 3 months) with placebo. The primary efficacy end point was nonfatal myocardial infarction, nonfatal stroke, or cardiovascular death. ResultsAt 48 months, the median reduction from baseline in the high-sensitivity C-reactive protein level was 26 percentage points greater in the group that received the 50-mg dose of canakinumab, 37 percentage points greater in the 150-mg group, and 41 percentage points greater in t...

5,660 citations

Journal ArticleDOI
TL;DR: The presence of CHIP in peripheral‐blood cells was associated with nearly a doubling in the risk of coronary heart disease in humans and with accelerated atherosclerosis in mice.
Abstract: BackgroundClonal hematopoiesis of indeterminate potential (CHIP), which is defined as the presence of an expanded somatic blood-cell clone in persons without other hematologic abnormalities, is common among older persons and is associated with an increased risk of hematologic cancer. We previously found preliminary evidence for an association between CHIP and atherosclerotic cardiovascular disease, but the nature of this association was unclear. MethodsWe used whole-exome sequencing to detect the presence of CHIP in peripheral-blood cells and associated such presence with coronary heart disease using samples from four case–control studies that together enrolled 4726 participants with coronary heart disease and 3529 controls. To assess causality, we perturbed the function of Tet2, the second most commonly mutated gene linked to clonal hematopoiesis, in the hematopoietic cells of atherosclerosis-prone mice. ResultsIn nested case–control analyses from two prospective cohorts, carriers of CHIP had a risk of c...

1,536 citations

Journal ArticleDOI
Sagi Abelson1, Grace Collord2, Grace Collord3, Stanley W.K. Ng4, Omer Weissbrod5, Netta Mendelson Cohen5, Elisabeth Niemeyer5, Noam Barda, Philip C. Zuzarte6, Lawrence E. Heisler6, Yogi Sundaravadanam6, Robert Luben2, Shabina Hayat2, Ting Ting Wang1, Ting Ting Wang4, Zhen Zhao1, Iulia Cirlan1, Trevor J. Pugh4, Trevor J. Pugh1, Trevor J. Pugh6, David Soave6, Karen Ng6, Calli Latimer3, Claire Hardy3, Keiran Raine3, David T. Jones3, Diana Hoult2, Abigail Britten2, John Douglas Mcpherson6, Mattias Johansson7, Faridah Mbabaali6, Jenna Eagles6, Jessica Miller6, Danielle Pasternack6, Lee Timms6, Paul M. Krzyzanowski6, Philip Awadalla6, Rui Costa8, Eran Segal5, Scott V. Bratman6, Scott V. Bratman4, Scott V. Bratman1, Philip A. Beer3, Sam Behjati2, Sam Behjati3, Inigo Martincorena3, Jean C.Y. Wang1, Jean C.Y. Wang4, Jean C.Y. Wang9, Kristian M. Bowles10, Kristian M. Bowles11, J. Ramón Quirós, Anna Karakatsani12, Carlo La Vecchia13, Antonia Trichopoulou, Elena Salamanca-Fernández14, José María Huerta, Aurelio Barricarte, Ruth C. Travis15, Rosario Tumino, Giovanna Masala16, Heiner Boeing, Salvatore Panico17, Rudolf Kaaks18, Alwin Krämer18, Sabina Sieri, Elio Riboli19, Paolo Vineis19, Matthieu Foll7, James McKay7, Silvia Polidoro, Núria Sala, Kay-Tee Khaw2, Roel Vermeulen20, Peter J. Campbell2, Peter J. Campbell3, Elli Papaemmanuil21, Elli Papaemmanuil3, Mark D. Minden, Amos Tanay5, Ran D. Balicer, Nicholas J. Wareham2, Moritz Gerstung3, Moritz Gerstung8, John E. Dick4, John E. Dick1, Paul Brennan7, George S. Vassiliou3, George S. Vassiliou2, Liran I. Shlush5, Liran I. Shlush1 
09 Jul 2018-Nature
TL;DR: Deep sequencing is used to analyse genes that are recurrently mutated in AML to distinguish between individuals who have a high risk of developing AML and those with benign ARCH, providing proof-of-concept that it is possible to discriminate ARCH from pre-AML many years before malignant transformation.
Abstract: The incidence of acute myeloid leukaemia (AML) increases with age and mortality exceeds 90% when diagnosed after age 65. Most cases arise without any detectable early symptoms and patients usually present with the acute complications of bone marrow failure1. The onset of such de novo AML cases is typically preceded by the accumulation of somatic mutations in preleukaemic haematopoietic stem and progenitor cells (HSPCs) that undergo clonal expansion2,3. However, recurrent AML mutations also accumulate in HSPCs during ageing of healthy individuals who do not develop AML, a phenomenon referred to as age-related clonal haematopoiesis (ARCH)4–8. Here we use deep sequencing to analyse genes that are recurrently mutated in AML to distinguish between individuals who have a high risk of developing AML and those with benign ARCH. We analysed peripheral blood cells from 95 individuals that were obtained on average 6.3 years before AML diagnosis (pre-AML group), together with 414 unselected age- and gender-matched individuals (control group). Pre-AML cases were distinct from controls and had more mutations per sample, higher variant allele frequencies, indicating greater clonal expansion, and showed enrichment of mutations in specific genes. Genetic parameters were used to derive a model that accurately predicted AML-free survival; this model was validated in an independent cohort of 29 pre-AML cases and 262 controls. Because AML is rare, we also developed an AML predictive model using a large electronic health record database that identified individuals at greater risk. Collectively our findings provide proof-of-concept that it is possible to discriminate ARCH from pre-AML many years before malignant transformation. This could in future enable earlier detection and monitoring, and may help to inform intervention.

567 citations

Journal ArticleDOI
01 Apr 2021-Nature
TL;DR: In this article, the authors review the evidence and discuss its implications for understanding of atherosclerosis, and examine its implications in the treatment of cardiovascular disease. But they do not discuss the role of the bone marrow in the pathogenesis of the disease.
Abstract: Emerging evidence has spurred a considerable evolution of concepts relating to atherosclerosis, and has called into question many previous notions. Here I review this evidence, and discuss its implications for understanding of atherosclerosis. The risk of developing atherosclerosis is no longer concentrated in Western countries, and it is instead involved in the majority of deaths worldwide. Atherosclerosis now affects younger people, and more women and individuals from a diverse range of ethnic backgrounds, than was formerly the case. The risk factor profile has shifted as levels of low-density lipoprotein (LDL) cholesterol, blood pressure and smoking have decreased. Recent research has challenged the protective effects of high-density lipoprotein, and now focuses on triglyceride-rich lipoproteins in addition to low-density lipoprotein as causal in atherosclerosis. Non-traditional drivers of atherosclerosis—such as disturbed sleep, physical inactivity, the microbiome, air pollution and environmental stress—have also gained attention. Inflammatory pathways and leukocytes link traditional and emerging risk factors alike to the altered behaviour of arterial wall cells. Probing the pathogenesis of atherosclerosis has highlighted the role of the bone marrow: somatic mutations in stem cells can cause clonal haematopoiesis, which represents a previously unrecognized but common and potent age-related contributor to the risk of developing cardiovascular disease. Characterizations of the mechanisms that underpin thrombotic complications of atherosclerosis have evolved beyond the ‘vulnerable plaque’ concept. These advances in our understanding of the biology of atherosclerosis have opened avenues to therapeutic interventions that promise to improve the prevention and treatment of now-ubiquitous atherosclerotic diseases. This Review discusses recent research that has transformed our understanding of the biology of atherosclerosis, and examines its implications for the treatment of atherosclerotic cardiovascular disease.

540 citations

Journal ArticleDOI
16 Apr 2019-Immunity
TL;DR: The varied roles of IL-1 family members in immune homeostasis and their contribution to pathologies, including autoimmunity and auto-inflammation, dysmetabolism, cardiovascular disorders, and cancer are discussed.

515 citations

References
More filters
Journal ArticleDOI
TL;DR: Age-related clonal hematopoiesis is a common condition that is associated with increases in the risk of hematologic cancer and in all-cause mortality, with the latter possibly due to an increased risk of cardiovascular disease.
Abstract: Background The incidence of hematologic cancers increases with age. These cancers are associated with recurrent somatic mutations in specific genes. We hypothesized that such mutations would be detectable in the blood of some persons who are not known to have hematologic disorders. Methods We analyzed whole-exome sequencing data from DNA in the peripheral-blood cells of 17,182 persons who were unselected for hematologic phenotypes. We looked for somatic mutations by identifying previously characterized single-nucleotide variants and small insertions or deletions in 160 genes that are recurrently mutated in hematologic cancers. The presence of mutations was analyzed for an association with hematologic phenotypes, survival, and cardiovascular events. Results Detectable somatic mutations were rare in persons younger than 40 years of age but rose appreciably in frequency with age. Among persons 70 to 79 years of age, 80 to 89 years of age, and 90 to 108 years of age, these clonal mutations were observed in 9.5% (219 of 2300 persons), 11.7% (37 of 317), and 18.4% (19 of 103), respectively. The majority of the variants occurred in three genes: DNMT3A, TET2, and ASXL1. The presence of a somatic mutation was associated with an increase in the risk of hematologic cancer (hazard ratio, 11.1; 95% confidence interval [CI], 3.9 to 32.6), an increase in all-cause mortality (hazard ratio, 1.4; 95% CI, 1.1 to 1.8), and increases in the risks of incident coronary heart disease (hazard ratio, 2.0; 95% CI, 1.2 to 3.4) and ischemic stroke (hazard ratio, 2.6; 95% CI, 1.4 to 4.8). Conclusions Age-related clonal hematopoiesis is a common condition that is associated with increases in the risk of hematologic cancer and in all-cause mortality, with the latter possibly due to an increased risk of cardiovascular disease. (Funded by the National Institutes of Health and others.)

3,183 citations

Journal ArticleDOI
29 Apr 2010-Nature
TL;DR: It is shown that cholesterol crystals activate the NLRP3 inflammasome in phagocytes in vitro in a process that involves phagolysosomal damage and that crystalline cholesterol acts as an endogenous danger signal and its deposition in arteries or elsewhere is an early cause rather than a late consequence of inflammation.
Abstract: The inflammatory nature of atherosclerosis is well established but the agent(s) that incite inflammation in the artery wall remain largely unknown. Germ-free animals are susceptible to atherosclerosis, suggesting that endogenous substances initiate the inflammation. Mature atherosclerotic lesions contain macroscopic deposits of cholesterol crystals in the necrotic core, but their appearance late in atherogenesis had been thought to disqualify them as primary inflammatory stimuli. However, using a new microscopic technique, we revealed that minute cholesterol crystals are present in early diet-induced atherosclerotic lesions and that their appearance in mice coincides with the first appearance of inflammatory cells. Other crystalline substances can induce inflammation by stimulating the caspase-1-activating NLRP3 (NALP3 or cryopyrin) inflammasome, which results in cleavage and secretion of interleukin (IL)-1 family cytokines. Here we show that cholesterol crystals activate the NLRP3 inflammasome in phagocytes in vitro in a process that involves phagolysosomal damage. Similarly, when injected intraperitoneally, cholesterol crystals induce acute inflammation, which is impaired in mice deficient in components of the NLRP3 inflammasome, cathepsin B, cathepsin L or IL-1 molecules. Moreover, when mice deficient in low-density lipoprotein receptor (LDLR) were bone-marrow transplanted with NLRP3-deficient, ASC (also known as PYCARD)-deficient or IL-1alpha/beta-deficient bone marrow and fed on a high-cholesterol diet, they had markedly decreased early atherosclerosis and inflammasome-dependent IL-18 levels. Minimally modified LDL can lead to cholesterol crystallization concomitant with NLRP3 inflammasome priming and activation in macrophages. Although there is the possibility that oxidized LDL activates the NLRP3 inflammasome in vivo, our results demonstrate that crystalline cholesterol acts as an endogenous danger signal and its deposition in arteries or elsewhere is an early cause rather than a late consequence of inflammation. These findings provide new insights into the pathogenesis of atherosclerosis and indicate new potential molecular targets for the therapy of this disease.

2,904 citations

Journal ArticleDOI
TL;DR: Clonal hematopoiesis with somatic mutations is readily detected by means of DNA sequencing, is increasingly common as people age, and is associated with increased risks of hematologic cancer and death.
Abstract: Cancers arise from multiple acquired mutations, which presumably occur over many years. Early stages in cancer development might be present years before cancers become clinically apparent. Methods We analyzed data from whole-exome sequencing of DNA in peripheral-blood cells from 12,380 persons, unselected for cancer or hematologic phenotypes. We identified somatic mutations on the basis of unusual allelic fractions. We used data from Swedish national patient registers to follow health outcomes for 2 to 7 years after DNA sampling. Results Clonal hematopoiesis with somatic mutations was observed in 10% of persons older than 65 years of age but in only 1% of those younger than 50 years of age. Detectable clonal expansions most frequently involved somatic mutations in three genes (DNMT3A, ASXL1, and TET2) that have previously been implicated in hematologic cancers. Clonal hematopoiesis was a strong risk factor for subsequent hematologic cancer (hazard ratio, 12.9; 95% confidence interval, 5.8 to 28.7). Approximately 42% of hematologic cancers in this cohort arose in persons who had clonality at the time of DNA sampling, more than 6 months before a first diagnosis of cancer. Analysis of bone marrow–biopsy specimens obtained from two patients at the time of diagnosis of acute myeloid leukemia revealed that their cancers arose from the earlier clones. Conclusions Clonal hematopoiesis with somatic mutations is readily detected by means of DNA sequencing, is increasingly common as people age, and is associated with increased risks of hematologic cancer and death. A subset of the genes that are mutated in patients with myeloid cancers is frequently mutated in apparently healthy persons; these mutations may represent characteristic early events in the development of hematologic cancers. (Funded by the National Human Genome Research Institute and others.)

2,497 citations

Journal ArticleDOI
TL;DR: This work reorganized probes on more than a dozen popular GeneChips into gene-, transcript- and exon-specific probe sets in light of up-to-date genome, cDNA/EST clustering and single nucleotide polymorphism information, and demonstrates that the original Affymetrix probe set definitions are inaccurate.
Abstract: Genome-wide expression profiling is a powerful tool for implicating novel gene ensembles in cellular mechanisms of health and disease The most popular platform for genome-wide expression profiling is the Affymetrix GeneChip However, its selection of probes relied on earlier genome and transcriptome annotation which is significantly different from current knowledge The resultant informatics problems have a profound impact on analysis and interpretation the data Here, we address these critical issues and offer a solution We identified several classes of problems at the individual probe level in the existing annotation, under the assumption that current genome and transcriptome databases are more accurate than those used for GeneChip design We then reorganized probes on more than a dozen popular GeneChips into gene-, transcript- and exon-specific probe sets in light of up-to-date genome, cDNA/EST clustering and single nucleotide polymorphism information Comparing analysis results between the original and the redefined probe sets reveals ∼30–50% discrepancy in the genes previously identified as differentially expressed, regardless of analysis method Our results demonstrate that the original Affymetrix probe set definitions are inaccurate, and many conclusions derived from past GeneChip analyses may be significantly flawed It will be beneficial to re-analyze existing GeneChip data with updated probe set definitions

1,849 citations

Journal ArticleDOI
TL;DR: MCC950 treatment rescued neonatal lethality in a mouse model of CAPS and was active in ex vivo samples from individuals with Muckle–Wells syndrome, and is a potential therapeutic for NLRP3-associated syndromes, and a tool for further study of theNLRP3 inflammasome in human health and disease.
Abstract: The NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3) inflammasome is a component of the inflammatory process, and its aberrant activation is pathogenic in inherited disorders such as cryopyrin-associated periodic syndrome (CAPS) and complex diseases such as multiple sclerosis, type 2 diabetes, Alzheimer's disease and atherosclerosis. We describe the development of MCC950, a potent, selective, small-molecule inhibitor of NLRP3. MCC950 blocked canonical and noncanonical NLRP3 activation at nanomolar concentrations. MCC950 specifically inhibited activation of NLRP3 but not the AIM2, NLRC4 or NLRP1 inflammasomes. MCC950 reduced interleukin-1β (IL-1β) production in vivo and attenuated the severity of experimental autoimmune encephalomyelitis (EAE), a disease model of multiple sclerosis. Furthermore, MCC950 treatment rescued neonatal lethality in a mouse model of CAPS and was active in ex vivo samples from individuals with Muckle-Wells syndrome. MCC950 is thus a potential therapeutic for NLRP3-associated syndromes, including autoinflammatory and autoimmune diseases, and a tool for further study of the NLRP3 inflammasome in human health and disease.

1,798 citations

Related Papers (5)