You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/370059144

Epigenetic regulation by ASXL1 in myeloid malignancies

Article  in  International Journal of Hematology · April 2023


DOI: 10.1007/s12185-023-03586-y

CITATION READS

1 50

2 authors, including:

Feng-Chun Yang
University of Texas Health Science Center at San Antonio
231 PUBLICATIONS   7,931 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Feng-Chun Yang on 23 May 2023.

The user has requested enhancement of the downloaded file.


International Journal of Hematology
https://doi.org/10.1007/s12185-023-03586-y

PROGRESS IN HEMATOLOGY

Stem cell regulation and dynamics in myeloid malignancies

Epigenetic regulation by ASXL1 in myeloid malignancies


Feng‑Chun Yang1,2   · Joel Agosto‑Peña1,2

Received: 2 February 2023 / Revised: 2 March 2023 / Accepted: 22 March 2023


© Japanese Society of Hematology 2023

Abstract
Myeloid malignancies are clonal hematopoietic disorders that are comprised of a spectrum of genetically heterogeneous
disorders, including myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPN), chronic myelomonocytic
leukemia (CMML), and acute myeloid leukemia (AML). Myeloid malignancies are characterized by excessive prolifera-
tion, abnormal self-renewal, and/or differentiation defects of hematopoietic stem cells (HSCs) and myeloid progenitor cells
hematopoietic stem/progenitor cells (HSPCs). Myeloid malignancies can be caused by genetic and epigenetic alterations
that provoke key cellular functions, such as self-renewal, proliferation, biased lineage commitment, and differentiation.
Advances in next-generation sequencing led to the identification of multiple mutations in myeloid neoplasms, and many new
gene mutations were identified as key factors in driving the pathogenesis of myeloid malignancies. The polycomb protein
ASXL1 was identified to be frequently mutated in all forms of myeloid malignancies, with mutational frequencies of 20%,
43%, 10%, and 20% in MDS, CMML, MPN, and AML, respectively. Significantly, ASXL1 mutations are associated with a
poor prognosis in all forms of myeloid malignancies. The fact that ASXL1 mutations are associated with poor prognosis in
patients with CMML, MDS, and AML, points to the possibility that ASXL1 mutation is a key factor in the development of
myeloid malignancies. This review summarizes the recent advances in understanding myeloid malignancies with a specific
focus on ASXL1 mutations.

Keywords  ASXL1 · Myeloid malignancies · Epigenetic regulation · Mutation

Identification of ASXL1 mutation in human myeloproliferative neoplasms (MPN), chronic myelomono-


diseases cytic leukemia (CMML), and acute myeloid leukemia
(AML). MDS occurs as a result of disordered insufficient
ASXL1 mutations in myeloid malignancies development of blood cells, to a greater or lesser extent,
within the bone marrow, which can evolve to bone marrow
Myeloid malignancies are clonal hematopoietic disorders failure and variable rates of transformation to AML. Mye-
characterized by excessive proliferation, abnormal self- loid malignancies can be fatal in the majority of patients.
renewal, and/or differentiation defects of hematopoietic stem Advances in next-generation sequencing led to the identi-
cells (HSCs) and myeloid progenitor cells hematopoietic fication of multiple mutations in myeloid neoplasms, and
stem/progenitor cells (HSPCs) [1–3]. Myeloid malignancies many new gene mutations were identified as key factors in
are comprised of a spectrum of genetically heterogeneous driving the development of myeloid malignancies. Targeted
disorders, including myelodysplastic syndromes (MDS), therapies are desperately needed.
Additional sex comb-like 1 (ASXL1) is one of the most
frequently mutated genes in a variety of myeloid malignan-
* Feng‑Chun Yang
cies, including ~ 15–21% of MDS [4, 5], ~ 8–10% of MPN
yangf1@uthscsa.edu
[6, 7], ~ 43–49% of CMML [8, 9], ~ 7–8% of juvenile myelo-
1
Department of Cell Systems and Anatomy, University monocytic leukemia (JMML) [10, 11], and ~ 3–10% of AML
of Texas Health Science Center at San Antonio, [12, 13]. ASXL1 is a human homolog of the Drosophila asx
San Antonio, TX 78229, USA
gene, which maps to chromosome 20q11.21. ASXL1 encodes
2
Mays Cancer Center, University of Texas Health Science the additional sex combs–like protein 1 (ASXL1). It belongs
Center at San Antonio, San Antonio, TX 78229, USA

13
Vol.:(0123456789)
F.-C. Yang, J. Agosto‑Peña

to the polycomb group (PcG) and trithorax complexes family to AML progression in univariate and multivariate analyses.
[14, 15]. Significantly, ASXL1 mutations are associated with [5] Despite this being widely acknowledged, it continues to
aggressiveness and poor prognosis in all myeloid malignan- remain controversial as to whether truncating mutations in
cies, including high-risk MDS, CMML, MPN, and second- ASXL1 result in gain- or loss-of-function, or if they confer
ary AML (sAML) [5, 7, 9, 12, 16]. ASXL1 mutations are dominant-negative activity in vivo. Hence, several groups
associated with a reduced time to progression in AML and including ours have investigated whether the presence of
constitute an independent prognostic marker in patients with the truncated forms of ASXL1 protein induces myeloid
MDS [5]. ASXL1 mutations also remained an independent malignancies.
adverse prognostic marker for time to AML progression in
CMML patients. [9] ASXL1 mutations in clonal hematopoiesis
ASXL1 mutations have also been described in some rare
myeloid malignancies. Mastocytosis is a heterogeneous Clonal hematopoiesis with somatic mutations is observed in
group of disorders which are characterized by abnormal at least 10% of people over 65 years of age, without apparent
growth and accumulation of mast cells in one or more organ hematologic disorders. Age-related clonal hematopoiesis is
systems. Traina et al. reported that ASXL1 mutations are a common premalignant condition and is associated with
found in two of eight systemic mastocytosis patients [17]. increased overall mortality and increased risk of hemato-
With a sixty-two SM-AHNMD patient cohort, Damaj et al. logic cancer, cardiovascular diseases, and atherosclerosis.
reported that 14% patients harbored ASXL1 mutations and Genes mutated in clonal hematopoiesis are similar to those
ASXL1 mutation remained an independent prognostic factor in myeloid malignancies. ASXL1 is one of the most fre-
that negatively affected overall survival. [18] quently involved somatic mutations detected in individuals
Interestingly, ASXL1 mutations have only been sporadi- with age-related clonal hematopoiesis [27–29]. A clinical
cally observed in lymphoid leukemia [20]. Compared with study by Yoshizato et al. showed that clonal hematopoie-
ASXL1 mutations found in all spectrum of myeloid malig- sis was prevalent in aplastic anemia, and ASXL1 mutations
nancies, ASXL2 mutations are restricted in a very specific are common in this population [19]. While these patients
subset of AML, ~ 23% of AML with t(8;21) translocation respond to immunosuppressive therapy, some still develop
[21, 22]. Interestingly, ASXL1 and ASXL2 mutations are MDS and AML.
mutually exclusive in t(8;21) AML, suggesting that the Several clinical studies demonstrate that ASXL1 muta-
mutations may have convergent downstream and/or have tions have been repeatedly identified in individuals with
synthetic lethal effects with each other. clonal hematopoiesis of indeterminate potential (CHIP),
Most ASXL1 mutations in myeloid malignancies are a condition whose frequency increases with advanced age
frameshift or nonsense mutations in exon 12 (last exon) [27–29], indicating that ASXL1 is one of the earliest genetic
before the PHD finger. Mutant ASXL1 transcripts are pre- events during the process of myeloid transformation. Close
dicted to produce C-terminally truncated ASXL1 protein by monitoring of clonal hematopoiesis by both deep sequencing
escaping from nonsense-mediated-decay [16, 23, 24]. These and SNP array will need to be combined with clinical evalu-
mutations are always heterozygous, suggesting that they ation for prognosis and to guide the management of patients
either have dominant-negative or gain-of-function effects. with aplastic anemia.
The most commonly detected ASXL1 mutation is ASXL1 In myeloid malignancies, clonal heterogeneity can evolve
NM_015338.5:c.1934dup;p.Gly646Trpfs*12 (ASXL1 upon disease progression and/or relapse AML. The shared
c.1934dupG), a frameshift mutation in ASXL1 exon 12 due somatic mutations in all leukemic cells reflect its clonal ori-
to a one nucleotide expansion of a contiguous repeat of gin, eg. the founding clone, and additional mutations pre-
eight guanine nucleotides (GGG​GGG​GG) that results in a sented in subpopulations of cells define subclones in leu-
truncated ASXL1 protein lacking the PHD finger [25, 26]. kemia. Sub-clonal mutations have been shown to be highly
ASXL2 mutations in myeloid malignancies, at least in AML enriched for epigenetic regulators. Therefore, understand-
with t(8;21)/RUNX1-RUNX1T1, are out-of-frame frameshift ing the mechanism by which ASXL1 mutations contribute to
mutations, exclusively heterozygous, and occur exclusively myeloid transformation is clinically important given the fact
in exons 11 and 12. [21] that ASXL1 is a key epigenetic regulator in hematopoiesis.
Inoue et al. reported that the truncating ASXL1 mutant Acquired aplastic anemia is a nonneoplastic bone marrow
can be detected in MDS cells, which may play a role in failure. Acquired aplastic anemia is an immune-mediated
MDS pathogenesis [23]. Thol et al. reported that, in an bone marrow aplasia, which has been known to associate
MDS cohort, ASXL1 frameshift mutations were inde- with clonal hematopoiesis upon marrow recovery. More
pendently associated with an unfavorable prognosis, and than 70% of patients with aplastic anemia develop somatic
patients harboring ASXL1 frameshift mutations not only mutations in their hematopoietic cells [30].Patient age at the
have a shorter overall survival, but also have reduced time diagnosis of aplastic anemia is known as one of the strongest

13
Epigenetic regulation by ASXL1 in myeloid malignancies

predictors of clonal hematopoiesis with MDS-associated homozygous or hematopoietic-specific loss of Asxl1


somatic mutations. Advances in cytogenetic and next-gen- results in the abnormal self-renewal capacity of HSCs and
eration sequencing demonstrated that clonal hematopoiesis the development of MDS or MDS/MPN-like disease, with
develops at a striking prevalence of 80–100% in adults with HSC/HPCs from the latter exhibiting decreased global lev-
aplastic anemia [19, 31–33]. Aplastic anemia patients have els of H3K27me3 and the increased expression of poste-
a disproportionate number of mutations in epigenetic regula- rior Hoxa genes. [44–46]
tors ASXL1, BCOR, and BCORL1. [19, 32, 34] In human AML, c.1934dupG; p.G646WfsX12 is the most
common ASXL1 mutation in myeloid malignancy patients
ASXL1 mutations in Bohring‑Opitz syndrome (BOS) [12]. Hsu and colleagues generated a mouse model (ASX-
L1tm/+) which bears human-like Asxl1 mutation [49]. They
In addition to somatic mutations in hematological malig- found that ASXL1tm/+ hematopoietic cells had higher short-
nancies, ASXL1 mutations have been discovered in in term in vitro proliferation capacities, and bone marrow cells
approximately 75% Bohring-Opitz syndrome (BOS) patients of Asxl1 G643WfsX12 heterozygotes showed compromised
[35–37]. BOS is a heterogeneous genetic condition charac- long-term in vivo repopulation and self-renewal capabilities.
terized by severe developmental delay, characteristic crani- ASXL1 G643WfsX12 mutation facilitated engraftment of
ofacial appearance, fixed contractures of the upper limbs, bone marrow cell overexpressing MN1, a proto-oncogene
abnormal posture, feeding difficulties, severe intellectual [49]. However, these mice do not show development of
disability, fetal microsomia, and failure to thrive [38–40]. blood malignancies, indicating that ASXL1 G643WfsX12
Most patients die in early childhood due to developmen- alone was not sufficient for development of blood malignan-
tal deficits, unexplained bradycardia, obstructive apnea, or cies in mice.
pulmonary infections [38]. In 2011, Hoischen et al. identi- Nagase et al. generated a conditional Asxl1-mutant knock-
fied de novo nonsense mutations of ASXL1 in patients with in mouse model (mimicking ASXL1 E635RfsX15) [50].
BOS [35]. Following this report, several groups reported that They found that expression of the C-terminal truncated
ASXL1 mutations account for around 50% of BOS patients. ASXL1 mutant in vivo led to myeloid skewing, thrombocy-
Bohring-Opitz syndrome occurs sporadically, suggesting tosis, age-dependent anemia, and dysplasia [50]. They also
that de novo mutations may be the underlying cause of this found that bone marrow cells from these mice had substan-
disorder. The majority of ASXL1 mutations in BOS are de tial reductions in the levels of H3K4me3 and H2AK119Ub,
novo frameshift and nonsense mutations. Regardless of the while the level of H3K27me3 was similar to that of control
clinical significance, it remains unknown whether ASXL1 cells. Mechanistically, they found that ASXL1 E635RfsX15-
mutations in BOS results in a loss-of-function, gain-of-func- KI HSCs displayed substantial reductions in H3K4me3 and
tion, or exert a dominant role. Deletion of Asxl1 (Asxl1−/−) in H2AK119Ub without significant reductions in H3K27me3,
mice leads to abnormalities of axial skeletal patterning (pos- distinct from the effects of Asxl1 loss [50]. Chromatin
terior and anterior), which suggests disruption of both PcG immunoprecipitation followed by next-generation sequenc-
and TrxG activities. [41–43]. Thus, as regulators of PcG and ing analysis demonstrated opposing effects of wild-type and
TrxG activities, the ASXL proteins are crucial in maintain- mutant Asxl1 on H3K4me3.
ing the lineage-specific gene expression patterns during cell We established a Vav1 promoter driven Asxl1Y588X trans-
division and replication. genic mouse model (Asxl1Y588XTg) expressing the analogous
protein product of the mutant ASXL1Y591X, frequently seen in
human patients. Asxl1Y588XTg mice had shortened survival
Mouse models of ASXL1 rates, and predisposition to a spectrum of myeloid malig-
nancies, closely recapitulating the characteristics of myeloid
Mouse models have been pivotal in understanding the malignancy patients with ASXL1-truncation mutations [47].
mechanisms by which they contribute to biological func- We found that ASXL1 truncation exerts an oncogenic role
tions and pathogenesis of diseases. Several mouse mod- in HSCs, at least in part through the gained interaction with
els have been developed by different research groups BET bromodomain-containing protein 4 along with altered
(Table  1), which allow for investigating the impact of gene expression. More importantly, Asxl1Y588XTg BM cells
ASXL1 alteration in hematopoiesis and myeloid malig- are sensitive to BET bromodomain inhibitors, suggesting
nancies. [15, 24, 44–47] Fisher et al. reported that Asxl1 potential novel therapies targeting the cells bearing the trun-
is required for normal hematopoiesis in an Asxl1-mutant cation mutation of ASXL1 through the use of BET inhibitors.
mouse model. [43, 48] However, these Asxl1-mutant mice Despite the significant impact of ASXL1 mutations in leuke-
did not reveal a profound HSC/HPC cellular phenotype mia, the underlying mechanisms by which they contribute to
or MDS/leukemia, likely due to an incomplete loss-of- myeloid malignancies remain unknown, which hinders the
function of Asxl1. [48] We and others have reported that development of targeted therapeutics.

13

13
Table 1  Summary of mouse models used to study the functions of Asxl1 in hematopoietic systems
Mouse model HSC phenotype Disease phenotype Disease type Histone modification References

Asxl1fl/fl;Mx1Cre or VavCre Increased LT-HSC and LSK Leukopenia, anemia, erythroid MDS-like disease Decreased H3K27me3 [45]
dysplasia, multiple cytopenia
Asxl1-MT (derived from the Not evaluated Multilineage myelodysplasia, MDS-like disease, occasional Decreased H3K27me3 [24]
most common ASXL1 mutation, pancytopenia progression to AML
1934dupG;G646WfsX12)
Asxl1−/− and Asxl1± Decreased LSK fractions in Multiple cytopenias; dysplastic MDS-like and MDS/MPN-like Reduced H3K4me3 and [46]
Asxl1−/− features; myeloid cell infiltration disease H3K27me3 in Asxl1−/−
in spleen and liver
Asxl1tm/+  + MN1 Promoted stem cell activities facilitates engraftment of cells None observed Increased H3K27me3 [49]
(mimics the most common (increased long-term colony overexpressing MN1
mutation c.1934dupG; cells
p.G646WfsX12)
Asxl1±;Nf1± Increased LT-HSC Hepatosplenomegaly, anemia, MDS, MDS/MPN, myeloid Increased H3K4me3 [110]
thrombocytopenia, myeloid leukemia
infiltration in spleen and liver
Asxl1Y588XTg Increased ST-HSC and LSK Anemia; myeloid cell infiltration AML, MPN, MDS, and MDS/ Increased H3K27ac and [47]
fractions; and enhanced HSC in spleen and liver; > 20% blast MPN H3K122ac
self-renewal cells in BM of leukemia mice
Asxl1-MT KI (mimics the well- Decreased HSPSC-LSK, MPP and Mild leukopenia, and anemia, CHIP Decreased H3K4me3, [50], [158]
described human ASXL1 muta- increased LT-HSC thrombocytosis, hypocellular H2AK119Ub, H3K27me3
tion, p.E635RfsX15) bone marrow, myeloid skewing
VavCre;Asxl1-MTfl/fl Decreased LT-HSC and LSK Decreased RBC and increased No disease alone Reduced H3K4me3 and [50]
fractions platelets H2AK119ub
Asxl1G643fs/+ Decreased LT-HSC and LSK Leukocytosis, anemia, thrombocy- MDS, MDS/MPN like disease Decreased H2AK119Ub [159]
tosis, disrupted splenic architec-
ture, myeloid and perivascular
infiltration in liver
F.-C. Yang, J. Agosto‑Peña
Epigenetic regulation by ASXL1 in myeloid malignancies

ASXL1 structural characterization Mutations in Asx enhance the phenotypes of both Poly-


comb group (PcG) and Thirotorax group (TrxG) gene
The human  ASXL  gene family consists of three mem- mutations [43]. PcG and TrxG proteins are regulated by
bers, ASXL1, ASXL2, and ASXL3, which encodes ASXL1, Enhancers of Trithorax and Polycomb (ETP) proteins,
ASXL2, and ASXL3 proteins, respectively. [15, 51, 52] which recruit PcG and TrxG complexes to target chromatin
Mutations in Asx enhance the phenotypes of both Poly- [53, 54]. PcG and TrxG proteins modulate the expression
comb group (PcG) and Thirotorax group (TrxG) gene of numerous genes by regulating histone methylation to
mutations [43]. PcG and TrxG proteins are regulated by maintain repressive and active chromatin states at target
Enhancers of Trithorax and Polycomb (ETP) proteins, loci, respectively. They preserve epigenetic memory and
which recruit PcG and TrxG complexes to target chroma- maintain the expression patterns of cell lineage-defining
tin [53, 54]. PcG and TrxG proteins modulate the expres- genes during cell division and replication. PcG proteins
sion of numerous genes by regulating histone methylation maintain the inactive state of Hox genes, while TrxG pro-
to maintain repressive and active chromatin states at tar- teins keep Hox genes activated [55].Hox genes are known
get loci, respectively. They preserve epigenetic memory to be critical for stem cell functions and dysregulated Hox
and maintain the expression patterns of cell lineage- protein expression results in leukemogenesis [55]. ASXL1
defining genes during cell division and replication. PcG contains 1541 amino acids [15], ASXL2 contains 1435
proteins maintain the inactive state of Hox genes, while amino acids [51], whereas, ASXL3 has 2248 amino acids
TrxG proteins keep Hox genes activated [55].Hox genes [52]. The three ASXL family member proteins share con-
are known to be critical for stem cell functions and, as served domains, including a N-terminal ASXN domain,
a result, dysregulated Hox protein expression results in an ASXH domain, ASXM1 and ASXM2 domains in the
leukemogenesis [55]. ASXL1 contains 1541 amino acids middle region and a PHD domain in the C-terminal region
[15], ASXL2 contains 1435 amino acids [51], whereas, [15, 51, 52]. While there is ∼40% amino acid homology
ASXL3 has 2248 amino acids [52]. The three ASXL fam- between ASXL1, ASXL2, and ASXL3, this homology
ily member proteins share conserved domains, including increases to ∼70% in the conserved domains of ASXL
a N-terminal ASXN domain, an ASXH domain, ASXM1 proteins including the ASXN, ASX homology (ASXH),
and ASXM2 domains in the middle region and a PHD ASXM1, and ASXM2 domains and the carboxy-terminal
domain in the C-terminal region [15, 51, 52]. While there cysteine cluster plant homedomain (PHD) [15, 51, 52].
is ∼40% amino acid homology between ASXL1, ASXL2, The ASXN domain is similar to the Forkhead-box (FOX)
and ASXL3, this homology increases to ∼70% in the con- domain, which is also known as the winged helix-turn-
served domains of ASXL proteins including the ASXN, helix domain, of FOXA3, FOXK1, FOXO1 and FOXO4.
ASX homology (ASXH), ASXM1, and ASXM2 domains [15, 51, 52, 56] The ASXH domain has been shown as a
and the carboxy-terminal cysteine cluster plant homedo- deubiquitinase adaptor domain (DEUBAD), spanning from
main (PHD) [15, 51, 52]. The ASXN domain is similar to 238 amino acid to 390 amino acid of ASXL1. The DEU-
the Forkhead-box (FOX) domain, which is also known as BAD domain is critical for the binding between ASXL1
the winged helix-turn-helix domain, of FOXA3, FOXK1, and the deubiquitinating enzyme (DUB) BRCA1-asso-
FOXO1 and FOXO4. [15, 51, 52, 56] The ASXH domain ciated protein 1 (BAP1) [57–60]. The C-terminal plant
has been shown as a deubiquitinase adaptor domain (DEU- homeodomain (PHD) finger is located at the very end of
BAD), spanning from 238 amino acid to 390 amino acid of the ASXL proteins and is a putative histone-interacting
ASXL1. The DEUBAD domain is critical for the binding PHD domain. [15, 51, 52]
between ASXL1 and the deubiquitinating enzyme (DUB)
BRCA1-associated protein 1 (BAP1) [57–60]. The C-ter-
minal plant homeodomain (PHD) finger is located at the ASXL1 interacts with other nuclear proteins
very end of the ASXL proteins and is a putative histone- to regulate hematopoiesis
interacting PHD domain. [15, 51, 52]
ASXL1 interacts with polycomb repressive complex
2 (PRC2)

ASXL1 structural characterization The polycomb repressive complex-2 (PRC2) complex


consists of four core components, including enhancer of
The human  ASXL  gene family consists of three mem- zeste homolog 1 (EZH1) or its paralog-EZH2, suppres-
bers, ASXL1, ASXL2, and ASXL3, which encodes ASXL1, sor of zeste 12 homolog (SUZ12), embryonic ectoderm
ASXL2, and ASXL3 proteins, respectively. [15, 51, 52] development (EED), and retinoblastoma-binding protein 4
and 7 (RBBP4/7) [61, 62]. As a component of the PRC2

13
F.-C. Yang, J. Agosto‑Peña

complex, EZH2 is a histone methyltransferase that initi- proteins (i.e., STAG1, STAG2, PDS5A, PDS5B, NIPBL,
ates trimethylation of lysine 27 in histone 3 (H3K27me3), a WAPL, and Sororin) [82–84]. Cohesin protein complex
repressive chromatin mark [44, 57, 63]. The C-terminal SET forms a ring-shaped structure to embrace chromatin,
(Su(var)3–9, Enhancer-of-zeste and Trithorax) domain of cohere sister chromatids, and prevent their premature sepa-
EZH2 catalyzes the methylation of H3K27. SUZ12 and EED ration. [85, 86] It has been reported that cohesin complex
of the PRC2 complex are required for the catalytic activity proteins are also involved in DNA replication, DNA repair
of EZH2 [64–66]. RBBP4 and AEBP2 are two additional and dynamic restructuring of chromosomes during cell
subunits of the PRC2 complex, which maintain the integ- division, and gene regulation. [87–92]
rity of PRC2 by stimulating EZH2 methytransferate activ- Using mouse model system, we reported that ASXL1
ity on H3K27 [67]. The pathological role of deregulated interacts with the three core proteins (SMC1A, SMC3,
EZH2 has been well documented in both human and mice. and RAD21) of cohesin. [93] Deletion of Asxl1 increases
EZH2 overexpression are found in human cancers, such as frequency of cells with nuclear bridging and prominent
breast, prostate, and bladder tumors [68]. Coincidentally, disrupted sister chromatid separation in myeloid cells. We
somatic loss-of-function mutations of EZH2 occur in MDS also showed that expressing ASXL1 amino acids 401 to
[63, 69]. EZH2 mutations in lower-risk MDS are correlated 587 (i.e., the cohesin binding region of ASXL1) disrupts
with a significantly worse prognosis [70]. Deletion of Ezh2 cohesin and endogenous ASXL1 interaction and increases
in mice results in the development of multi-forms of hemato- the frequency of cells with nuclear bridging and prema-
logical malignancies, such as acute T-cell lymphoblastic leu- ture sister chromatid separation. Cohesin works together
kemia (T-ALL), lymphoma, and myeloid malignancies [71, with CCCTC-binding factor (CTCF) to facilitate long-
72, 73]. Loss of Ezh2 promotes JAK2V617F mutant-associated range interactions in the genome and gene regulation [94].
myelofibrosis [74–76]. These findings indicate that EZH2 The interaction between cohesin complex and CTCF is
plays a tumor suppressive role in MDS and MPN. required for CTCF-anchored loops and contributes to
ASXL1 has been reported to exert its epigenetic regula- the positioning of cohesin at CTCF binding sites [95]. It
tory function by activating or repressing the transcription of has also been reported that cohesin and NIPBL-MAU2
genes involved in proliferation and differentiation through its form an active holoenzyme that interacts with DNA either
cooperative effect with other chromatin modifiers to regu- pseudo-topologically or non-topologically to extrude
late histone modification [44, 57, 77]. ASXL1 is required to genomic interphase DNA into loops [96]. Our ChIP-seq
maintain homeotic gene activation and silencing. [15, 43] analysis on HSC/HPCs revealed a significant overlap of
Being a member of the enhancer of trithorax (trxG) and ASXL1/SMC1A/RAD21 binding sites, and Asxl1 loss
polycomb (PcG) group genes in Drosophila, Asx is associ- resulted in the decreased genomic occupancy of cohesin
ated with histone modification [14, 78]. PcG proteins sustain and the dysregulation of several ASXL1/SMC1A/RAD21
the ‘off state’ while trxG proteins sustain the ‘on state’ of common target genes, including those that have a role in
the clustered Hox (Homeotic box) genes [79]. It has been apoptosis, proliferation, and myelopoiesis and/or leukemo-
reported that the PcG and trxG genes are required for normal genesis (e.g., Stat3, Cbfb, and Fus) [93]. Our study dem-
hematopoiesis and mutations of these genes disturb self- onstrates that ASXL1-cohesin interaction is required to
renewal of hematopoietic stem cells (HSCs) in mammals maintain normal cell morphology, chromatid separation,
[80, 81]. Using myeloid leukemic cell lines, Abdel-Wahab and gene expression in hematopoietic cells [93]. Recent
and colleagues reported that that ASXL1 physically interacts studies indicate that cohesin mutations confer a clonal
with PRC2, which is likely important for the recruitment advantage by altering the chromatin and transcriptional
of the PRC2 complex [44]. Therefore, ASXL1 exerts the state of HSC/HPCs, perturbing the balance between self-
repressive regulation of its target genes through H3K27me3 renewal and differentiation and that cohesin mutations
by physically interacting with other PRC2 complex proteins could confer resistance to inflammatory signals, increased
and regulates epigenetic marks and expression of genes criti- self-renewal and clonal expansion [97, 98]. While clinical
cal for HSCs/hematopoietic progenitor cells (HPCs) through studies demonstate significant impact of somatic mutations
interaction with polycomb complex proteins and various of ASXL1 in clonal hematopoiesis and myeloid malig-
transcription activators and repressors. nancies [28, 29], the impact of cohesin complex in the
ASXL1 mutation-related clonal hematopoiesis remains to
ASXL1 interacts with cohesin protein complex be determined.
Chromatin looping is known to bring promoters and
Cohesin is an evolutionarily conserved macromolecu- enhancers in proximity to activate gene transcription [99].
lar complex. Cohesin protein complex is composed of Whether ASXL1 is also involved in chromatin looping via
multiple-subunit proteins, including three core subunits cohesin complex proteins to modulate the chromatin archi-
(SMC1A, SMC3, and RAD21), and multiple regulatory tecture and regulate gene expression remains to be explored.

13
Epigenetic regulation by ASXL1 in myeloid malignancies

ASXL1 interacts with BAP1 form a PR‑DUB complex well as a striking reduction of H3K27me3 [104]. Further
studies showed that BAP1 is required for the mutant ASXL1
BRCA1 associated protein 1  (BAP1) is a ubiquitin car- in the leukemogenesis [108], and deleting one allele of Bap1
boxyl-terminal hydrolase, and a member of the ubiquitin delay the truncated ASXL1-driven myeloid malignancies
C-terminal hydrolase (UCH) family [100]. BAP1 is located in vivo [109]. These data suggest that BAP1 has multifac-
in the nucleus and was originally discovered as a protein eted functions in malignant hematopoiesis. It is unclear how
interacting with the RING finger domain of BRCA1 [100]. much redundancy in function occurs between ASXL1 and
BAP1 has three critical domains, a UCH N-terminal cata- ASXL2 in their cooperative effect with BAP1.
lytic domain with enzymatic activity, a central linker region ASXL1 truncations are shown to confer an enhanced
which modulates binding to host cell factor-1, and a C-termi- activity on the ASXL1–BAP1 DUB complex [104], high-
nal ULD domain that potentially coordinates protein–protein lighting the significance of the ASXL1-BAP1 complex in
interactions. Loss-of-function BAP1 mutations have recently normal biological processes and cancer progression. We
been identified in a diverse array of solid tumor types [101, hypothesize that the A­ SXL1aa1−587 truncation mutant pro-
102]. Harbour and colleagues reported inactivating muta- motes myeloid malignancies by enhancing BAP1 DUB
tions in BAP1 occur in 47% of uveal melanomas [103]. activity. To test this hypothesis, we examined whether
They found a case with germline BAP1 mutation implying deleting one Bap1 allele can delay or even eradicate
gene susceptibility, and that BAP1 mutation was strongly ­ASXL1aa1−587-driven myeloid malignancies in vivo. We
associated with metastasis. These clinical data indicate a generated Mx1Cre;Bap1f/+;Asxl1Y588XTg mice. PI:pC was
tumor suppressive role of BAP1 in cancer. Surprisingly, injected into 7  month-old Asxl1 Y588XTg (disease estab-
unlike ASXL1 mutations, BAP1 is rarely mutated in myeloid lished) mice to induce a single Bap1 allele inactivation in
malignancies. the hematopoietic compartment. We analyzed the hemat-
PRC1 and PRC2 mediate mono-ubiquitination on lysine opoietic phenotypes at 3 months after pI:pC injection. Bap1
119 of histone H2A (H2AK119ub) and methylation on hemizygous deletion in Asxl1Y588XTg mice is sufficient to
lysine 27 of histone H3 (H3K27me1/me2/me3), respec- prevent the A ­ SXL1aa1−587-driven HSC/HPC dysregulation
tively [67]. BAP1 is activated by ASXL1 to deubiquitinate and myeloid malignancy. These data indicate that ASXL1
mono-ubiquitinated H2A at K119 during polycomb protein- truncation mutations confer gain-of-function in the patho-
mediated gene repression [60, 104]. ASXL1 and BAP1 form genesis of myeloid malignancies by increasing BAP1 DUB
a Polycomb-repressive deubiquitinase (PR-DUB) complex activity as reducing BAP1 activity ameliorates the abnormal
that removes monoubiquitin from H2AK119 [57]. ASXL1 hematopoietic phenotypes in Asxl1Y588XTg mice.
lacks a catalytic domain and generally exerts its functions
by assembling chromatin modification complexes and tran- ASXL1 interacts with RNAPII family members (Pol II)
scription factors. As a result, ASXL1 plays an important
role in epigenetic regulation by activating or repressing the ASXL1 was reported to bind to PRC2 complex and regu-
transcription of genes involved in proliferation and differen- late tri-methylation of histone H3 lysine 27 (H3K27me3)
tiation through its cooperative effect with other chromatin in myeloid cells [45, 46]. To determine the putative impact
modifiers to regulate histone modification [44, 57, 77]. As a of ASXL1-mediated H3K27me3 on the cell fates of bone
major component of PR-DUB complex, ASXL1 plays piv- marrow stromal cells (BMSCs), we performed H3K27me3
otal roles in controlling the levels of H2AK119ub and sub- and RNAPII ChIP-seq using WT and Asxl1−/− BMSCs
sequent gene regulation [57, 105]. ASXL1 binds to BAP1 [110]. Surprisingly, we didn’t observe a strong associa-
via the DEUBAD domain, to assemble a PR-DUB complex, tion between ASXL1 binding and H3K27me3 enrichment
which removes ubiquitin from H2AK119Ub [57–60]. Recent on the genome of BMSCs. There is a minimal correlation
biochemical characterization of BAP1 complexes suggests between H3K27me3 enrichment and RNAPII occupancy.
that BAP1 forms two mutually exclusive complexes with These results suggest ASXL1 regulates gene expression in
either ASXL1 or ASXL2 [58, 59], and both complexes can an H3K27me3-independent manner in BMSCs. To exam-
stimulate H2AK119 deubiquitination in vitro. [60] ine whether ASXL1 and RNAPII co-localize at the same
Dey and colleagues reported that conditional deletion of genomic regions, we performed integrative analyses to assess
Bap1 results in anemia, splenomegaly, leukocytosis, and the genome-wide distribution in BMSCs. The results showed
progenitor expansion, with decreased HoxA gene expres- a significantly positive correlation between ASXL1 binding
sion [106, 107]. Interestingly, deletion of Asxl1 or Asxl2 and RNAPII occupancy where ASXL1 promoter binding
increases HoxA gene expression due to chromatin altera- was highly overlapped with the RNAPII loading. Analyses
tions. Overexpression of truncated forms of ASXL1, but not of the coverage between ASXL1 and RNAPII bound genes
full-length ASXL1, in combination with overexpression of showed a high degree (88.51%) of the overlay, suggesting
BAP1 resulted in clear depletion of global H2AK119ub as a role for ASXL1 in gene regulation. Biochemistry studies

13
F.-C. Yang, J. Agosto‑Peña

verified that ASXL1 interacts with RNAPII family mem- Cooperative leukemogenic effect of ASXL1
bers (POLR2A, POLR2B, and POLR2C) in the nuclei of alteration and co‑occurring somatic
BMSCs. We further found that loss of Asxl1 alters the tar- alterations
get gene expression by deregulating RNAPII transcriptional
activity in the nuclei of BMSCs. These studies demonstrate ASXL1 mutation co‑occurs with RAS pathway related
multifaceted functions of ASXL1 in gene regulation by gene mutations
assembling epigenetic regulators and transcription factors
at specific gene loci. These ASXL1 interacting proteins are Co-mutations of key genes are often in patients with mye-
summarized in Fig. 1. loid malignancies. ASXL1 is known to co-occur with other
gene mutations. The matched chronic and acute stages have
ASXL1 interacts with other nuclear proteins shown that additional alterations associate with disease pro-
gression. Studies with NGS provide evidence that multiple
Recent studies demonstrate a function of ASXL1 in the mutations can be identified in single cells of MDS patients.
modulation of H3K4 methylation, an active histone mark on These results imply a linear evolution of the disease and the
gene regulation [77]. Although phylogenetic analyses sug- existence of a dominant clone. Integrated genomic approach
gest that the PHD domain of ASXL1 may bind to H3K4me3 identified potential cooperating events in AML [112, 113],
[111], the exact function of the PHD domain of ASXL1 as Asxl1 mutations frequently co-occur with mutations
remains unclear. The role of ASXL1 in the connection of leading to hyperactivation of Ras in myeloid malignancies
promoters and enhancers remains to be explored. such as RAS, PTPN11, and NF1 mutations [4, 7, 114, 115].
Inactivating mutations of NF1 (neurofibromatosis type I)
are common genetic events in JMML and AML [116]. The
neurofibromin 1 gene (NF1) encodes neurofibromin, a RAS
GTPase activating protein, that when mutated, results in
hyperactive RAS signaling that is pro-leukemogenic [117].
NF1 alterations in signaling molecules can be grouped in

ASXL1 structure
238 390
DEUBAD
1 86 241 369 523 618 1099 1123 1506 1537
ASXL1-WT
ASXN ASXH NLS ASXM1 ASXM2 PHD
(1541aa)

238 390

BAP1
401 587
Not specified regions known:
1 248 Cohesin
(SMC1A,
OGT POLR2A
SMC3,
Rad21) POLR2B
N terminal
POLR2C
EZH2

1 86 241 369 523 618 646


ASXL1-MT
(range from ASXN ASXH ASXM1
~635-646aa)

1 200

BRD4

Fig. 1  Summary of wild type and mutant ASXL1 protein interactions

13
Epigenetic regulation by ASXL1 in myeloid malignancies

two major categories, a first one that is found in MPNs and The histological analyses of BM, spleen, and liver sections
affects oncogenic tyrosine kinases (ABL1, JAK2, FGFR1, of Asxl1±;Nf1± mice showed disrupted architecture with
PDGFRs) and the downstream JAK-STAT and/or PI3-kinase significant myeloid cell infiltration. The mouse models of
pathways, and a second one that is mutated in CMML and Asxl1 and Nf1 provide scientific evidence for the cooperative
affect the RAS-MAP kinase pathway (RAS, PTPN11, NF1). effect of key driver mutations in disease progression. RNA-
Expression of NRasG12D in combination with mASXL1 seq analyses on Asxl1±;Nf1± HSC/HPCs revealed aberrant
knockdown accelerated myeloproliferation and impaired sur- transcriptional activation of multiple pathways critical for
vival compared with mice transplanted with empty vector leukemogenesis, such as MYC, NRAS, and BRD4 [47],
expressing cells in vivo, demonstrating that ASXL1 loss, indicating a gain-of-function of the alterations of Asxl1 and
in combination with co-occurring oncogenic NRasG12D, Nf1 in epigenetic regulation. Significantly, pharmacologi-
can lead to hematopoietic transformation and increased self- cal inhibition of both BET bromodomain and the MAPK
renewal. [44] pathway prevents leukemia initiation and inhibits disease
In a cohort of 138 patients with myeloid malignancies progression [118]. This study provides a novel therapeutic
harboring ASXL1 mutations, we examined the gene muta- strategy for the treatment of myeloid malignancy patients
tional profiles of these patients based on targeted sequenc- with ASXL1 and RAS pathway gene mutations.
ing of 67 frequently mutated genes in myeloid malignan-
cies and identified 35 patients with gene mutations involving ASXL1 alteration cooperates with JAK2V617F
the RAS/MAPK signaling pathway, including NF1, NRAS, to accelerate myelofibrosis
KRAS, PTPN11 or CBL in these ASXL1-mutated patients
[118]. Malignancies in NF1 result from a combination of Myeloproliferative neoplasms (MPNs) are clonal hemat-
ubiquitous NF1 heterozygosity and somatic loss of the opoietic stem cell disorders. MPNs are characterized by
residual NF1 allele (i.e. loss of heterozygosity) [119, 120]. aberrant hematopoietic proliferation of one or more hemat-
Interestingly, we did not observe loss of heterozygosity opoietic cell lineages with increased risks of myelofibrosis
in both ASXL1 and RAS pathway genes in these patients. (MF) progression and leukemic transformation. [122, 123]
Of note, the incidence of AML was significantly higher in Somatic mutation of JAK2V617F is considered to be the
patients with co-mutations of ASXL1 and RAS pathway gene most notable landmark in the diagnosis of classic Philadel-
mutations than the cases without RAS pathway mutations. phia chromosome-negative MPNs and is present in > 95% of
According to the revised International Prognostic Scoring polycythemia vera patients and in ~ 50% of essential throm-
System (IPSS-R) [121], the ASXL1 mutated MDS patients bocythemia and primary myelofibrosis [124]. JAK2V617F
with RAS pathway gene mutations had a higher frequency activates pro-oncogenic signaling via diverse STAT-depend-
of very high-risk MDS compared to patients without a RAS ent and STAT-independent pathways [125]. JAK2 mutations
pathway gene mutation. These data demonstrate that con- are associated with age-related clonal hematopoiesis [29].
comitant mutations of ASXL1 and RAS pathway genes are Sidon et al. reported that JAK2V617F mutation was detected
likely associated with myeloid malignancies with worse in 10% of blood samples from healthy volunteers [126],
prognosis. underscoring the necessity of additional genetic alterations
To determine the functional significance of co-mutations for progression to an MPN.
of ASXL1 and NF1 in the disease progression of myeloid ASXL1 mutations co-occur with JAK2 mutations in patients
malignancies, we intercrossed Asxl1 heterozygous (Asxl1±) with myeloid malignancies [127, 128]. We reported that con-
mice with Nf1 ± mice and generated Asxl1 ±;Nf1 ± mice comitent mutations of ASXL1 and JAK2 results in decreased
[118]. The survival rate of Asxl1±;Nf1± mice was signifi- levels of hemoglobin (Hb), increased white blood cell (WBC)
cantly lower than those of the other 3 genotypes of mice. count, increased platelet (PLT) count, palpable splenomeg-
Examination of peripheral blood parameters showed that aly, and clonal abnormal karyotypes when compared with
a subset of Asxl1±;Nf1± mice developed profound anemia polycythemia vera (PV) patients with JAK2V617F mutation
and thrombocytopenia as compared to age-matched WT only in a cohort of 95 PV patients with JAK2V617F muta-
mice. In contrast, the survival of Asxl1± mice was 83% up tion [129]. In this cohort, we observed a higher proportion
to 600 days of age and the deceased Asxl1± mice died of of ASXL1 mutations in post-PV myelofibrosis (PPMF) (26%)
myeloid malignancies, such as MDS or MDS/MPN, which is than in PV patients (4%). PV patients with co-mutations of
consistent with our previous report. A subset of Asxl1±;Nf1± Asxl1 and JAK2V617F had a poor MF-free survival. Our study
mice developed myeloid leukemia. Morphologic analysis of demonstrated that JAK2V617F positive PV patients harbor-
cytospins prepared from the BM cells revealed an accumu- ing mutated ASXL1 exhibited poor myelofibrosis-free survival,
lation of blastic cells in Asxl1±;Nf1± mice [118]. Necropsy and that mutated ASXL1 was present in 25% of post-PV mye-
of the deceased/moribund Asxl1±;Nf1± mice demonstrated lofibrosis cases compared with only 4% of PV cases without
hepatosplenomegaly and pale foot pads, indicating cachexia.

13
F.-C. Yang, J. Agosto‑Peña

myelofibrosis, evidence of a worse prognosis in patients har- Mutual exclusive gene mutations
boring ASXL1 mutations in addition to JAK2V617F. between ASXL1 and other genes
To further study the impact of Asxl1 alteration on dis-
ease progression in JAK2V617F-mediated MPNs and Despite the high frequencies of several gene mutations
JAK2V617F-mutant hematopoietic stem and progenitor cell seen in MDS, MPN, AML and other blood malignancies,
(HSC/HPC) function, we assessed hematopoietic pheno- some of these common mutations are mutually exclu-
types in Asxl1±;JAK2V617F mice in vivo. [129] We found sive. In the case of ASXL1, it has been shown to be mutu-
that JAK2V617F;Asxl1± mice had significantly shorter mean ally exclusive with some common and other less common
survival rates (~ 60%) than JAK2V617F or Asxl1± mice. We mutations, such as DNMT3A, NPM1, WT1 and ASXL2.
found that mice with heterozygous loss of Asxl1 (Asxl1±) and [12, 21, 136–139]
JAK2V617F expression accelerated the development of bone As mentioned before, ASXL1 is part of the ASXL fam-
marrow fibrosis and some of JAK2V617F;Asxl1± mice pro- ily of proteins, along with ASXL2 and ASXL3. Interest-
gressed/transformed to secondary acute myeloid leukemia ingly, ASXL1 and ASXL2 mutations are mutually exclu-
(sAML) [124]. These results indicate that heterozygous dele- sive [21]. Given the high homology on several of their
tion of Asxl1 promoted MF in JAK2V617F-driven MPN in mice, domains and the fact that ASXL1 and ASXL2 mutations
and Asxl1 alteration cooperates with JAK2V617F mutation to are mutually exclusive with one another, it is likely that
accelerate myeloid leukemic transformation. these two genes' mutations share a common mechanism
of transformation in AML patients. These results raise
Co‑mutation of ASXL1 and SRSF2 in myeloid the possibility that ASXL1 and ASXL2 mutations are both
malignanices significant and distinct in AML patients. More studies
about the differences between ASXL1 and ASXL2 are
Concomitant mutations of ASXL1 and SRSF2 are common in critically needed to understand these genetically observed
MDS and CMML and are frequently found in patients with differences, and can probably unravel pathogenic routes
s-AML [16, 130–134]. AML with ASXL1 and SRSF2 muta- between ASXL1 mutations and ASXL2 mutations. Daou
tions has been associated with a dismal prognosis, similar to et al. reported that BAP1 forms two mutually exclusive
AML with high-risk cytogenetic features [133, 134]. John- complexes with the transcriptional regulators ASXL1 and
son and colleagues reported that ASXL1/SRSF2 co-mutated ASXL2, which are necessary for maintaining proper pro-
AML was significantly more likely to express 2 or more tein levels of BAP1 [59]. They also found that the interac-
monocytic markers, more likely to express aberrant CD2 and tion between ASXL1/2 and BAP1 requires ASXM, which
CD56, and significantly less likely to express the myeloblast is necessary for promoting BAP1-mediated deubiquitina-
markers CD34 and CD117. These data suggest that ASXL1/ tion of its physiological substrate H2AK119ub. The phys-
SRSF2 co-mutation results in a biased monocytic differentia- ical interaction between ASXL1/2 and BAP1 and DUB
tion [135]. ASXL1/SRSF2 co-mutated AML frequently arises activity are required for proper coordination of cell cycle
secondarily in patients with antecedent myeloid malignancy, progression. In contrast, Park and colleagues found that
and even when these mutations apparently occur “de novo,” while both ASXL1 and ASXL2 interact with peroxisome
patients may have unrecognized peripheral monocytosis prior proliferator-activated receptor γ, they play opposite roles
to leukemic onset. Their findings point to a possibility that in adipogenesis via reciprocal regulation of peroxisome
this subset of AML may often arise as a secondary AML proliferator-activated receptor γ. [140]
from an occult CMML-like MDS or MDS/MPN even in the Carbuccia and colleagues reported that ASXL1 and NPM1
absence of a known prior myeloid neoplasm. Richardson and mutations are mutually exclusive on a study of 63 AML
colleagues reported that ASXL1mutSRSF2mut AML, in contrast patients [138]. The mutual exclusive mutations of ASXL1
to ASXL1mutSRSF2wt AML, shares a mutational profile and and NPM1 have been also repored by Pratcorona [12, 136],
immunophenotype with CMML suggesting that this genomic which implies that this is a common phenomenom seen in
profile may identify patients with secondary AML (s-AML) different cohorts of ASXL1 mutated patients. ASXL1 and
from preexisting CMML [132]. Regardless of the clinical sig- NPM1 may function through a similar pathway in regulat-
nificance of the comutation of ASXL1 and SRSF2, the underly- ing hematopoietic cell functions. The authors reported that
ing mechanisms remains to be investigated. 9p INK4A-ARF-INK4B loci are regulated by both the poly-
comb complexes and the histone chaperone NPM1 [138].
These loci are key for the regulation of hematopoietic func-
tions, such as senescence and/or self-renewal.
In a cohort of CN-AML patients, Metzeler et  al.
reported that ASXL1  mutations were almost mutually

13
Epigenetic regulation by ASXL1 in myeloid malignancies

exclusive with  NPM1 mutations and  FLT3-ITD [141]. by RNA modification has the potential to be used in patients
Chou and colleagues also reported an inverse association to avoid long-term myelosuppression.
of ASXL1 mutation with WT1 mutation and FLT3-ITD Developing CAR T cells for AML has been hindered
[12]. Supprisingly, ASXL2, another member of ASXL fam- by a paucity of targets expressing on the blastic cells, but
ily, and FLT3-ITD mutations had a significant co-occur- not on hematopoietic progenitor cells. Evolutionarily con-
rence, completely opposite from the mutually exclusive served glucose-regulated-protein 78 (GRP78) is a key regu-
phenotype seen with ASXL1 and FLT3-ITD mutations lator on the unfolded protein response [155], and residing
[136]. However, the mechanism underlying the mutually in the endoplasmic reticulum makes it a promising target
exclusive mutations between ASXL1 and other genes in for AML-redirected CAR T cell therapy, since GRP78 is
leukemia remains to be further studied. translocated to the cell surface upon cell stress, a common
phenotype of malignant cells [156]. Hebbar et al. showed
the feasibility of targeting cell surface GRP78 in AML with
Challenges in therapy for ASXL1 mutated CAR T cells [157]. Hebbar and colleagues designed a panel
myeloid malignancies of GRP78-specific CARs (GRP78-CARs) using a peptide
that specifically binds to GRP78, and demonstrated that
ASXL1 mutation was an independent risk factor associated GRP78 is overexpressed in a variety of AML specimens.
with worse event-free survival in chronic phase CML [142]. The GRP78-CAR T cell cultures showed minimal fratri-
Although successful therapeutic strategies are developed cide and antigen-dependent T cell differentiation, and the
for several types of myeloid leukemia, including imatinib GRP78-CAR T cell have potent anti-AML activity without
in CML [143, 144], all trans retinoic acid (ATRA) in hematopoietic progenitor cell (HPC) toxicity [157]. Further
almost all the cases of acute promyelocytic leukemia (APL) investigation for developing CAR-T cells in myeloid leuke-
[145, 146], JAK2 inhibitors in myeloproliferative diseases mia patients with ASXL1 mutations is required.
[147–149], effective treatment are desperately needed for
most of the cases of myeloid malignancies, especially mye-
loid malignancies with ASXL1 mutations. ASXL1 muta- Open questions
tion frequency was significantly higher in TKI-resistant
patients. A study by Kim and colleagues revealed that While the advances in genomic and epigenetic research have
mutations in epigenetic modification pathway genes such uncovered a central role for aberrant epigenetic regulation
as DNMT3A and ASXL1 could be driver mutations in TKI in the pathogenesis of myeloid malignancies, the molecular
resistance-related progression of CML outside ABL1 KD mechanisms underlying the alterations of epigenetic modi-
mutations [150]. A study of comparative genomic land- fiers in myeloid malignancies remain largely unknown. Sev-
scape of AML by Tyner and colleagues with a large cohort eral studies suggest that macrophage driven inflammatory
of patients samples demonstrated that mutations in several responses are responsible for the accelerated atherosclerosis
genes, most notably ASXL1 or TP53, were associated with a seen in patients with clonal hematopoiesis, ASXL1 muta-
broad pattern of drug resistance. [151] tions induce a clonal advantage of hematopoietic cells and
Immunotherapy is one of the promising novel thera- subsequent CH development has not been elucidated. In
peutics targeting cancers [152]. Chimeric antigen recep- addition, how ASXL1 mutations induce a clonal advantage
tor (CAR) T cells using engineered T cells extracted from of hematopoietic cells and subsequent clonal hematopoie-
patients are able to target a specific antigen on the surface sis development has not been elucidated. Despite the asso-
of malignant cells [153]. Using humanized mouse model ciation of particular gene mutations observed early in the
system, Kenderian and colleagues developed CART cells course of disease and response to therapy and survival, it
to target CD33 (CART33) using the anti-CD33 single should be underscored that the complex dynamics of clonal
chain variable fragment used in gemtuzumab ozogamicin hematopoiesis are highly variable and not necessarily deter-
(clone My96) and tested the activity and toxicity of these minative. Close monitoring of clonal hematopoiesis by both
cells [154]. They found that CART33 exhibited significant deep sequencing and SNP-array will need to be combined
effector functions in vitro and resulted in eradication of with clinical evaluation for prognosis and to guide manage-
leukemia and prolonged survival in AML xenografts. How- ment of patients with aplastic anemia. It remains unknown
ever, CART33 also resulted in human lineage cytopenias whether the ASXL1 or ASXL2 PHD domain binds to non-
and reduction of myeloid progenitors in xenograft models histone proteins. Although observations in mice demonstrate
of hematopoietic toxicity, suggesting that CD33-specific dominance of DNMT3A, ASXL1 and other mutations, the
CART cells would have unacceptable toxicity. The authors exact mechanism of clonal selection of these mutations in
then designed a transiently expressed mRNA anti-CD33 aplastic anemia is unclear. Finally, given the poor progno-
CAR and found that transient expression of anti-CD33 CAR sis of myeloid malignancy patients with ASXL1 mutations,

13
F.-C. Yang, J. Agosto‑Peña

effort on developing effective treatment becomes more 14. Milne TA, Sinclair DA, Brock HW. The Additional sex combs
important. gene of drosophila is required for activation and repression of
homeotic loci, and interacts specifically with polycomb and
Acknowledgements  We apologize to the researchers whose work was super sex combs. Mol Gen Genet MGG. 1999;261:753–61.
not cited due to space limitations. We would also like to acknowledge 15. Fisher CL, Berger J, Randazzo F, Brock HW. A human homolog
the NCI Training Grant (T32 CA148724) given by the Mays Cancer of additional sex combs, ADDITIONAL SEX COMBS-LIKE
Center for their financial and professional support of the PhD student 1, maps to chromosome 20q11. Gene. 2003;306:115–26.
Joel Agosto-Peña. 16. Gelsi-Boyer V, et al. Mutations in ASXL1 are associated with
poor prognosis across the spectrum of malignant myeloid dis-
Funding  NIH/NCI, HL149318, Feng-Chun Yang, R01 HL158081, eases. J Hematol Oncol. 2012;5:12. https://​doi.​org/​10.​1186/​
Feng-Chun Yang,CA172408, Feng-Chun Yang, T32 CA148724, Joel 1756-​8722-5-​12.
Agosto-Peña. 17. Traina, F. et al. Single nucleotide polymorphism array lesions,
TET2, DNMT3A, ASXL1 and CBL mutations are present in
systemic mastocytosis. PLoS One 7, e43090, doi:https://​doi.​
Declarations  org/​10.​1371/​journ​al.​pone.​00430​90 (2012).
18. Damaj, G. et al. ASXL1 but Not TET2 Mutations Adversely
Conflict of interest  The authors declare that they have no conflict of Impact Overall Survival of Patients Suffering Systemic Mas-
interest. tocytosis with Associated Clonal Hematologic Non-Mast-Cell
Diseases. PLoS One 9, e85362, doi:https://​doi.​org/​10.​1371/​
journ​al.​pone.​00853​62 (2014).
19. Yoshizato T, et  al. Somatic Mutations and Clonal Hemat-
References opoiesis in Aplastic Anemia. N Engl J Med. 2015;373:35–47.
https://​doi.​org/​10.​1056/​NEJMo​a1414​799.
1. Hofmann WK, Koeffler HP. Myelodysplastic syndrome. Annu 20. Prebet T, et al. Concomitant germ-line RUNX1 and acquired
Rev Med. 2005;56:1–16. https://​doi.​org/​10.​1146/​annur​ev.​med.​ ASXL1 mutations in a T-cell acute lymphoblastic leukemia.
56.​082103.​104704. Eur J Haematol. 2013;91:277–9. https://​doi.​org/​10.​1111/​ejh.​
2. Nimer SD. Myelodysplastic syndromes. Blood. 2008;111:4841– 12147.
51. https://​doi.​org/​10.​1182/​blood-​2007-​08-​078139. 21. Micol JB, et al. Frequent ASXL2 mutations in acute myeloid
3. Bejar R, Levine R, Ebert BL. Unraveling the molecular patho- leukemia patients with t(8;21)/RUNX1-RUNX1T1 chromosomal
physiology of myelodysplastic syndromes. J Clin Oncol. translocations. Blood. 2014;124:1445–9. https://d​ oi.o​ rg/1​ 0.1​ 182/​
2011;29:504–15. https://​doi.​org/​10.​1200/​JCO.​2010.​31.​1175. blood-​2014-​04-​571018.
4. Bejar R, et al. Clinical effect of point mutations in myelodysplas- 22. Duployez N, et al. Comprehensive mutational profiling of core
tic syndromes. N Engl J Med. 2011;364:2496–506. https://​doi.​ binding factor acute myeloid leukemia. Blood. 2016;127:2451–9.
org/​10.​1056/​NEJMo​a1013​343. https://​doi.​org/​10.​1182/​blood-​2015-​12-​688705.
5. Thol F, et al. Prognostic significance of ASXL1 mutations in 23. Inoue, D. et al. Truncation mutants of ASXL1 observed in
patients with myelodysplastic syndromes. J Clin Oncol: Off J Am myeloid malignancies are expressed at detectable protein levels.
Soc Clin Oncol. 2011;29:2499–506. https://d​ oi.o​ rg/1​ 0.1​ 200/J​ CO.​ Experimental hematology 44, 172–176 e171, doi:https://​doi.​org/​
2010.​33.​4938. 10.​1016/j.​exphem.​2015.​11.​011 (2016).
6. Carbuccia N, et al. Mutations of ASXL1 gene in myeloprolifera- 24. Inoue D, et  al. Myelodysplastic syndromes are induced by
tive neoplasms. Leukemia. 2009;23:2183–6. https://​doi.​org/​10.​ histone methylation-altering ASXL1 mutations. J Clin Invest.
1038/​leu.​2009.​141. 2013;123:4627–40. https://​doi.​org/​10.​1172/​JCI70​739.
7. Brecqueville M, et  al. Mutation analysis of ASXL1, CBL, 25. Alvarez Argote, J. & Dasanu, C. A. ASXL1 mutations in myeloid
DNMT3A, IDH1, IDH2, JAK2, MPL, NF1, SF3B1, SUZ12, neoplasms: pathogenetic considerations, impact on clinical out-
and TET2 in myeloproliferative neoplasms. Genes Chromosomes comes and survival. Current Medical Research and Opinion 34,
Cancer. 2012;51:743–55. https://​doi.​org/​10.​1002/​gcc.​21960. 757–763, doi:https://​doi.​org/​10.​1080/​03007​995.​2016.​12768​96
8. Gelsi-Boyer V, et al. Mutations of polycomb-associated gene (2018).
ASXL1 in myelodysplastic syndromes and chronic myelomono- 26. Alberti MO, et al. Discriminating a common somatic ASXL1
cytic leukaemia. Br J Haematol. 2009;145:788–800. https://​doi.​ mutation (c.1934dup; p.G646Wfs*12) From artifact in myeloid
org/​10.​1111/j.​1365-​2141.​2009.​07697.x. malignancies using NGS. Leukemia. 2018;32:1874–8. https://​
9. Gelsi-Boyer V, et al. ASXL1 mutation is associated with poor doi.​org/​10.​1038/​s41375-​018-​0193-y.
prognosis and acute transformation in chronic myelomonocytic 27. Genovese G, et  al. Clonal hematopoiesis and blood-cancer
leukaemia. Br J Haematol. 2010;151:365–75. https://​doi.​org/​10.​ risk inferred from blood DNA sequence. N Engl J Med.
1111/j.​1365-​2141.​2010.​08381.x. 2014;371:2477–87. https://​doi.​org/​10.​1056/​NEJMo​a1409​405.
10. Stieglitz E, et al. The genomic landscape of juvenile myelomono- 28. Xie M, et al. Age-related mutations associated with clonal hemat-
cytic leukemia. Nat Genet. 2015;47:1326–33. https://​doi.​org/​10.​ opoietic expansion and malignancies. Nat Med. 2014;20:1472–8.
1038/​ng.​3400. https://​doi.​org/​10.​1038/​nm.​3733.
11. Caye A, et al. Juvenile myelomonocytic leukemia displays muta- 29. Jaiswal S, et al. Age-related clonal hematopoiesis associated with
tions in components of the RAS pathway and the PRC2 network. adverse outcomes. N Engl J Med. 2014;371:2488–98. https://d​ oi.​
Nat Genet. 2015;47:1334–40. https://​doi.​org/​10.​1038/​ng.​3420. org/​10.​1056/​NEJMo​a1408​617.
12. Chou WC, et al. Distinct clinical and biological features of de 30. Young NS. Current concepts in the pathophysiology and treat-
novo acute myeloid leukemia with additional sex comb-like 1 ment of aplastic anemia. Hematol Am Soc Hematol Educ Pro-
(ASXL1) mutations. Blood. 2010;116:4086–94. https://​doi.​org/​ gram. 2013;76–81:2013. https://​doi.​org/​10.​1182/​ashed​ucati​on-​
10.​1182/​blood-​2010-​05-​283291. 2013.1.​76.
13. Patel JP, et al. Prognostic relevance of integrated genetic profiling 31. Stanley N, Olson TS, Babushok DV. Recent advances in under-
in acute myeloid leukemia. N Engl J Med. 2012;366:1079–89. standing clonal haematopoiesis in aplastic anaemia. Br J Haema-
https://​doi.​org/​10.​1056/​NEJMo​a1112​304. tol. 2017;177:509–25. https://​doi.​org/​10.​1111/​bjh.​14510.

13
Epigenetic regulation by ASXL1 in myeloid malignancies

32. Babushok DV, Olson TS, Bessler M. Somatic mutations 51. Katoh M, Katoh M. Identification and characterization of
and clonal hematopoiesis in aplastic anemia. N Engl J Med. ASXL2 gene in silico. Int J Oncol. 2003;23:845–50.
2015;373:1673. https://​doi.​org/​10.​1056/​NEJMc​15097​03. 52. Katoh M, Katoh M. Identification and characterization of
33. Babushok DV, et al. Emergence of clonal hematopoiesis in the ASXL3 gene in silico. Int J Oncol. 2004;24:1617–22.
majority of patients with acquired aplastic anemia. Cancer Genet. 53. Schuettengruber B, Cavalli G. Recruitment of polycomb group
2015;208:115–28. https://​doi.​org/​10.​1016/j.​cance​rgen.​2015.​01.​ complexes and their role in the dynamic regulation of cell fate
007. choice. Development. 2009;136:3531–42. https://​doi.​org/​10.​
34. Negoro E, et al. Origins of myelodysplastic syndromes after 1242/​dev.​033902.
aplastic anemia. Blood. 2017;130:1953–7. https://​doi.​org/​10.​ 54. Schuettengruber B, Chourrout D, Vervoort M, Leblanc B, Cav-
1182/​blood-​2017-​02-​767731. alli G. Genome regulation by polycomb and trithorax proteins.
35. Hoischen A, et al. De novo nonsense mutations in ASXL1 cause Cell. 2007;128:735–45. https://​doi.​org/​10.​1016/j.​cell.​2007.​02.​
Bohring-Opitz syndrome. Nat Genet. 2011;43:729–31. https://​ 009.
doi.​org/​10.​1038/​ng.​868. 55. Schuettengruber B, Martinez AM, Iovino N, Cavalli G. Trithorax
36. Magini P, et al. Two novel patients with Bohring-Opitz syn- group proteins: switching genes on and keeping them active. Nat
drome caused by de novo ASXL1 mutations. Am J Med Genet Rev Mol Cell Biol. 2011;12:799–814. https://​doi.​org/​10.​1038/​
A. 2012;158A:917–21. https://​doi.​org/​10.​1002/​ajmg.a.​35265. nrm32​30.
37. Russell B, et al. Clinical management of patients with ASXL1 56. Sanchez-Pulido L, Kong L, Ponting CP. A common ancestry for
mutations and Bohring-Opitz syndrome, emphasizing the BAP1 and Uch37 regulators. Bioinformatics. 2012;28:1953–6.
need for Wilms tumor surveillance. Am J Med Genet A. https://​doi.​org/​10.​1093/​bioin​forma​tics/​bts319.
2015;167A:2122–31. https://​doi.​org/​10.​1002/​ajmg.a.​37131. 57. Scheuermann JC, et  al. Histone H2A deubiquitinase activ-
38. Hastings R, et al. Bohring-Opitz (Oberklaid-Danks) syndrome: ity of the polycomb repressive complex PR-DUB. Nature.
clinical study, review of the literature, and discussion of possible 2010;465:243–7. https://​doi.​org/​10.​1038/​natur​e08966.
pathogenesis. Eur J Hum Genet. 2011;19:513–9. https://​doi.​org/​ 58. Peng H, et al. Familial and somatic BAP1 mutations inacti-
10.​1038/​ejhg.​2010.​234. vate ASXL1/2-mediated allosteric regulation of BAP1 deu-
39. Oberklaid F, Danks DM. The opitz trigonocephaly syndrome. a biquitinase by targeting multiple independent domains. Can
case report. Am J Dis Child. 1975;129:1348–9. Res. 2018;78:1200–13. https://​doi.​org/​10.​1158/​0008-​5472.​
40. Bohring A, Oudesluijs GG, Grange DK, Zampino G, Thierry CAN-​17-​2876.
P. New cases of Bohring-Opitz syndrome, update, and critical 59. Daou S, et al. The BAP1/ASXL2 histone H2A deubiquitinase
review of the literature. Am J Med Genet A. 2006;140:1257–63. complex regulates cell proliferation and is disrupted in cancer.
https://​doi.​org/​10.​1002/​ajmg.a.​31265. J Biol Chem. 2015;290:28643–63. https://​doi.​org/​10.​1074/​jbc.​
41. Katoh M. Functional and cancer genomics of ASXL family mem- M115.​661553.
bers. Br J Cancer. 2013;109:299–306. https://​doi.​org/​10.​1038/​ 60. Sahtoe DD, van Dijk WJ, Ekkebus R, Ovaa H, Sixma TK. BAP1/
bjc.​2013.​281. ASXL1 recruitment and activation for H2A deubiquitination. Nat
42. Baskind HA, et al. Functional conservation of Asxl2, a murine Commun. 2016;7:10292. https://d​ oi.o​ rg/1​ 0.1​ 038/n​ comms​ 10292.
homolog for the drosophila enhancer of trithorax and polycomb 61. Di Carlo V, Mocavini I, Di Croce L. Polycomb complexes in nor-
group gene Asx. PLoS One. 2009;4: e4750. https://​doi.​org/​10.​ mal and malignant hematopoiesis. J Cell Biol. 2019;218:55–69.
1371/​journ​al.​pone.​00047​50. https://​doi.​org/​10.​1083/​jcb.​20180​8028.
43. Fisher CL, et al. Additional sex combs-like 1 belongs to the 62. Hu D, Shilatifard A. Epigenetics of hematopoiesis and hemato-
enhancer of trithorax and polycomb group and genetically inter- logical malignancies. Genes Dev. 2016;30:2021–41. https://​doi.​
acts with Cbx2 in mice. Dev Biol. 2010;337:9–15. https://​doi.​ org/​10.​1101/​gad.​284109.​116.
org/​10.​1016/j.​ydbio.​2009.​10.​004. 63. Ernst T, et al. Inactivating mutations of the histone methyltrans-
44. Abdel-Wahab O, et al. ASXL1 mutations promote myeloid trans- ferase gene EZH2 in myeloid disorders. Nat Genet. 2010;42:722–
formation through loss of PRC2-Mediated gene repression. Can- 6. https://​doi.​org/​10.​1038/​ng.​621.
cer Cell. 2012;22:180–93. https://​doi.​org/​10.​1016/j.​ccr.​2012.​06.​ 64. Cao R, Zhang Y. SUZ12 is required for both the histone methyl-
032. transferase activity and the silencing function of the EED-EZH2
45. Abdel-Wahab O, et al. Deletion of Asxl1 results in myelodys- complex. Mol Cell. 2004;15:57–67. https://​doi.​org/​10.​1016/j.​
plasia and severe developmental defects in vivo. J Exp Med. molcel.​2004.​06.​020.
2013;210:2641–59. https://​doi.​org/​10.​1084/​jem.​20131​141. 65. Pasini D, Bracken AP, Jensen MR, Lazzerini Denchi E, Helin K.
46. Wang J, et al. Loss of Asxl1 leads to myelodysplastic syndrome- Suz12 is essential for mouse development and for EZH2 histone
like disease in mice. Blood. 2014;123:541–53. https://​doi.​org/​ methyltransferase activity. EMBO J. 2004;23:4061–71. https://​
10.​1182/​blood-​2013-​05-​500272. doi.​org/​10.​1038/​sj.​emboj.​76004​02.
47. Yang H, et al. Gain of function of ASXL1 truncating protein in 66. Montgomery ND, et al. The murine polycomb group protein Eed
the pathogenesis of myeloid malignancies. Blood. 2018;131:328– is required for global histone H3 lysine-27 methylation. Curr
41. https://​doi.​org/​10.​1182/​blood-​2017-​06-​789669. Biol: CB. 2005;15:942–7. https://d​ oi.o​ rg/1​ 0.1​ 016/j.c​ ub.2​ 005.0​ 4.​
48. Fisher CL, et al. Loss-of-function Additional sex combs like 1 051.
mutations disrupt hematopoiesis but do not cause severe myelo- 67. Margueron R, Reinberg D. The polycomb complex PRC2 and its
dysplasia or leukemia. Blood. 2010;115:38–46. https://​doi.​org/​ mark in life. Nature. 2011;469:343–9. https://​doi.​org/​10.​1038/​
10.​1182/​blood-​2009-​07-​230698. natur​e09784.
49. Hsu YC, et al. The distinct biological implications of Asxl1 68. Bachmann IM, et al. EZH2 expression is associated with high
mutation and its roles in leukemogenesis revealed by a knock-in proliferation rate and aggressive tumor subgroups in cutane-
mouse model. J Hematol Oncol. 2017;10:139. https://d​ oi.o​ rg/1​ 0.​ ous melanoma and cancers of the endometrium, prostate, and
1186/​s13045-​017-​0508-x. breast. J Clin Oncol. 2006;24:268–73. https://​doi.​org/​10.​1200/​
50. Nagase R, et al. Expression of mutant Asxl1 perturbs hemat- JCO.​2005.​01.​5180.
opoiesis and promotes susceptibility to leukemic transformation. 69. Nikoloski G, et al. Somatic mutations of the histone methyl-
J Exp Med. 2018;215:1729–47. https://​doi.​org/​10.​1084/​jem.​ transferase gene EZH2 in myelodysplastic syndromes. Nat Genet.
20171​151. 2010;42:665–7. https://​doi.​org/​10.​1038/​ng.​620.

13
F.-C. Yang, J. Agosto‑Peña

70. Bejar R, et al. Validation of a prognostic model and the impact EMBO J. 2009;28:2625–35. https://​d oi.​o rg/​1 0.​1 038/​e mboj.​
of mutations in patients with lower-risk myelodysplastic syn- 2009.​202.
dromes. J Clin Oncol. 2012;30:3376–82. https://​doi.​org/​10.​ 89. Horsfield JA, et  al. Cohesin-dependent regulation of Runx
1200/​JCO.​2011.​40.​7379. genes. Development. 2007;134:2639–49. https://​doi.​org/​10.​
71. Simon C, et al. A key role for EZH2 and associated genes in 1242/​dev.​002485.
mouse and human adult T-cell acute leukemia. Genes Dev. 90. Parelho V, et al. Cohesins functionally associate with CTCF on
2012;26:651–6. https://​doi.​org/​10.​1101/​gad.​186411.​111. mammalian chromosome arms. Cell. 2008;132:422–33. https://​
72. Souroullas GP, et al. An oncogenic Ezh2 mutation induces doi.​org/​10.​1016/j.​cell.​2008.​01.​011.
tumors through global redistribution of histone 3 lysine 27 91. Wendt KS, et al. Cohesin mediates transcriptional insulation
trimethylation. Nat Med. 2016;22:632–40. https://​doi.​org/​10.​ by CCCTC-binding factor. Nature. 2008;451:796–801. https://​
1038/​nm.​4092. doi.​org/​10.​1038/​natur​e06634.
73. Mochizuki-Kashio M, et al. Ezh2 loss in hematopoietic stem 92. Nasmyth K, Haering CH. Cohesin: its roles and mechanisms.
cells predisposes mice to develop heterogeneous malignan- Annu Rev Genet. 2009;43:525–58. https://​d oi.​o rg/​1 0.​1 146/​
cies in an Ezh1-dependent manner. Blood. 2015;126:1172–83. annur​ev-​genet-​102108-​134233.
https://​doi.​org/​10.​1182/​blood-​2015-​03-​634428. 93. Li Z, et  al. ASXL1 interacts with the cohesin complex to
74. Sashida G, et al. The loss of Ezh2 drives the pathogenesis of maintain chromatid separation and gene expression for normal
myelofibrosis and sensitizes tumor-initiating cells to bromo- hematopoiesis. Sci Adv. 2017;3: e1601602. https://​doi.​org/​10.​
domain inhibition. J Exp Med. 2016;213:1459–77. https://​doi.​ 1126/​sciadv.​16016​02.
org/​10.​1084/​jem.​20151​121. 94. Zuin J, et al. Cohesin and CTCF differentially affect chromatin
75. Shimizu T, et al. Loss of Ezh2 synergizes with JAK2-V617F in architecture and gene expression in human cells. Proc Natl
initiating myeloproliferative neoplasms and promoting myelofi- Acad Sci U S A. 2014;111:996–1001. https://​doi.​org/​10.​1073/​
brosis. J Exp Med. 2016;213:1479–96. https://​doi.​org/​10.​1084/​ pnas.​13177​88111.
jem.​20151​136. 95. Li Y, et al. The structural basis for cohesin-CTCF-anchored
76. Yang Y, Akada H, Nath D, Hutchison RE, Mohi G. Loss loops. Nature. 2020;578:472–6. https:// ​ d oi. ​ o rg/ ​ 1 0. ​ 1 038/​
of Ezh2 cooperates with Jak2V617F in the development of s41586-​019-​1910-z.
myelofibrosis in a mouse model of myeloproliferative neo- 96. Davidson IF, et al. DNA loop extrusion by human cohesin.
plasm. Blood. 2016;127:3410–23. https://​d oi.​o rg/​1 0.​1 182/​ Science. 2019;366:1338–45. https://​doi.​org/​10.​1126/​scien​ce.​
blood-​2015-​11-​679431. aaz34​18.
77. Inoue D, et al. A novel ASXL1-OGT axis plays roles in H3K4 97. Cuartero S, et al. Control of inducible gene expression links
methylation and tumor suppression in myeloid malignan- cohesin to hematopoietic progenitor self-renewal and differ-
cies. Leukemia. 2018;32:1327–37. https://​d oi.​o rg/​1 0.​1 038/​ entiation. Nat Immunol. 2018;19:932–41. https://​doi.​org/​10.​
s41375-​018-​0083-3. 1038/​s41590-​018-​0184-1.
78. LaJeunesse D, Shearn A. E(z): a polycomb group gene or a 98. Cuartero S, Innes AJ, Merkenschlager M. Towards a better
trithorax group gene? Development. 1996;122:2189–97. understanding of cohesin mutations in AML. Front Oncol.
79. Simon JA, Tamkun JW. Programming off and on states in 2019;9:867. https://​doi.​org/​10.​3389/​fonc.​2019.​00867.
chromatin: mechanisms of Polycomb and trithorax group com- 99. Hnisz D, Day DS, Young RA. Insulated neighborhoods: struc-
plexes. Curr Opin Genet Dev. 2002;12:210–8. tural and functional units of mammalian gene control. Cell.
80. Sauvageau M, Sauvageau G. Polycomb group genes: keeping 2016;167:1188–200. https://​d oi.​o rg/​1 0.​1 016/j.​c ell.​2 016.​1 0.​
stem cell activity in balance. PLoS Biol. 2008;6: e113. https://​ 024.
doi.​org/​10.​1371/​journ​al.​pbio.​00601​13. 100. Jensen DE, et al. BAP1: a novel ubiquitin hydrolase which
81. Vernimmen D, et  al. Polycomb eviction as a new distant binds to the BRCA1 RING finger and enhances BRCA1-medi-
enhancer function. Genes Dev. 2011;25:1583–8. https://​doi.​ ated cell growth suppression. Oncogene. 1998;16:1097–112.
org/​10.​1101/​gad.​16985​411. 101. Bott M, et al. The nuclear deubiquitinase BAP1 is commonly
82. Kon A, et al. Recurrent mutations in multiple components inactivated by somatic mutations and 3p21.1 losses in malig-
of the cohesin complex in myeloid neoplasms. Nat Genet. nant pleural mesothelioma. Nat Genet. 2011;43:668–72.
2013;45:1232–7. https://​doi.​org/​10.​1038/​ng.​2731. https://​doi.​org/​10.​1038/​ng.​855.
83. Seitan VC, et al. Cohesin-based chromatin interactions enable 102. Testa JR, et al. Germline BAP1 mutations predispose to malig-
regulated gene expression within preexisting architectural com- nant mesothelioma. Nat Genet. 2011;43:1022–5. https://​doi.​
partments. Genome Res. 2013;23:2066–77. https://​doi.​org/​10.​ org/​10.​1038/​ng.​912.
1101/​gr.​161620.​113. 103. Harbour JW, et al. Frequent mutation of BAP1 in metastasizing
84. Seitan VC, Merkenschlager M. Cohesin and chromatin organi- uveal melanomas. Science. 2010;330:1410–3. https://​doi.​org/​
sation. Curr Opin Genet Dev. 2012;22:93–100. https://​doi.​org/​ 10.​1126/​scien​ce.​11944​72.
10.​1016/j.​gde.​2011.​11.​003. 104. Balasubramani A, et al. Cancer-associated ASXL1 mutations
85. Onn I, Heidinger-Pauli JM, Guacci V, Unal E, Koshland DE. may act as gain-of-function mutations of the ASXL1-BAP1
Sister chromatid cohesion: a simple concept with a complex complex. Nat Commun. 2015;6:7307. https://​doi.​org/​10.​1038/​
reality. Annu Rev Cell Dev Biol. 2008;24:105–29. https://​doi.​ ncomm​s8307.
org/​10.​1146/​annur​ev.​cellb​io.​24.​110707.​175350. 105. Daou S, et  al. Monoubiquitination of ASXLs controls the
86. Losada A. The regulation of sister chromatid cohesion. Bio- deubiquitinase activity of the tumor suppressor BAP1.
chim Biophys Acta. 2008;1786:41–8. https://​doi.​org/​10.​1016/j.​ Nat Commun. 2018;9:4385. https:// ​ d oi. ​ o rg/ ​ 1 0. ​ 1 038/​
bbcan.​2008.​04.​003. s41467-​018-​06854-2.
87. Strom L, et al. Postreplicative formation of cohesion is required 106. Dey A, et al. Loss of the tumor suppressor BAP1 causes myeloid
for repair and induced by a single DNA break. Science. transformation. Science. 2012;337:1541–6. https://​doi.​org/​10.​
2007;317:242–5. https://​doi.​org/​10.​1126/​scien​ce.​11406​49. 1126/​scien​ce.​12217​11.
88. Watrin E, Peters JM. The cohesin complex is required for the 107. LaFave LM, et al. Loss of BAP1 function leads to EZH2-depend-
DNA damage-induced G2/M checkpoint in mammalian cells. ent transformation. Nat Med. 2015;21:1344–9. https://d​ oi.o​ rg/1​ 0.​
1038/​nm.​3947.

13
Epigenetic regulation by ASXL1 in myeloid malignancies

108. Asada S, et al. Mutant ASXL1 cooperates with BAP1 to pro- 126. Sidon P, El Housni H, Dessars B, Heimann P. The JAK2V617F
mote myeloid leukaemogenesis. Nat Commun. 2018;9:2733. mutation is detectable at very low level in peripheral blood of
https://​doi.​org/​10.​1038/​s41467-​018-​05085-9. healthy donors. Leukemia. 2006;20:1622. https://​doi.​org/​10.​
109. Guo Y, et  al. Reduced BAP1 activity prevents ASXL1 1038/​sj.​leu.​24042​92.
truncation-driven myeloid malignancy in  vivo. Leukemia. 127. Shih AH, Abdel-Wahab O, Patel JP, Levine RL. The role of muta-
2018;32:1834–7. https://​doi.​org/​10.​1038/​s41375-​018-​0126-9. tions in epigenetic regulators in myeloid malignancies. Nat Rev
110. Zhang P, et al. Loss of ASXL1 in the bone marrow niche dys- Cancer. 2012;12:599–612. https://​doi.​org/​10.​1038/​nrc33​43.
regulates hematopoietic stem and progenitor cell fates. Cell 128. Brecqueville M, et al. Array comparative genomic hybridization
Discov. 2018;4:4. https://​doi.​org/​10.​1038/​s41421-​017-​0004-z. and sequencing of 23 genes in 80 patients with myelofibrosis at
111. Katoh M. Functional proteomics of the epigenetic regulators chronic or acute phase. Haematologica. 2014;99:37–45. https://​
ASXL1, ASXL2 and ASXL3: a convergence of proteomics and doi.​org/​10.​3324/​haema​tol.​2013.​091454.
epigenetics for translational medicine. Expert Rev Proteom- 129. Guo Y, et al. ASXL1 alteration cooperates with JAK2V617F
ics. 2015;12:317–28. https://​doi.​org/​10.​1586/​14789​450.​2015.​ to accelerate myelofibrosis. Leukemia. 2019. https://​doi.​org/​10.​
10334​09. 1038/​s41375-​018-​0347-y.
112. Hatlen MA, et al. Integrative genetic analysis of mouse and 130. Zhang SJ, et  al. Genetic analysis of patients with leukemic
human AML identifies cooperating disease alleles. J Exp Med. transformation of myeloproliferative neoplasms shows recur-
2016;213:25–34. https://​doi.​org/​10.​1084/​jem.​20150​524. rent SRSF2 mutations that are associated with adverse out-
113. Cancer Genome Atlas Research Network, et al. Genomic and come. Blood. 2012;119:4480–5. https://​ d oi. ​ o rg/ ​ 1 0. ​ 1 182/​
epigenomic landscapes of adult de novo acute myeloid leuke- blood-​2011-​11-​390252.
mia. New Engl J Med. 2013;368:2059–74. https://​doi.​org/​10.​ 131. Yoshizato T, et al. Genetic abnormalities in myelodysplasia and
1056/​NEJMo​a1301​689. secondary acute myeloid leukemia: impact on outcome of stem
114. Rocquain J, et al. Combined mutations of ASXL1, CBL, FLT3, cell transplantation. Blood. 2017;129:2347–58. https://​doi.​org/​
IDH1, IDH2, JAK2, KRAS, NPM1, NRAS, RUNX1, TET2 10.​1182/​blood-​2016-​12-​754796.
and WT1 genes in myelodysplastic syndromes and acute mye- 132. Richardson DR, et al. Genomic characteristics and prognostic
loid leukemias. BMC Cancer. 2010;10:401. https://​doi.​org/​10.​ significance of co-mutated ASXL1/SRSF2 acute myeloid leu-
1186/​1471-​2407-​10-​401. kemia. Am J Hematol. 2021;96:462–70. https://​doi.​org/​10.​1002/​
115. Fernandez-Mercado M, et al. Mutation patterns of 16 genes in ajh.​26110.
primary and secondary acute myeloid leukemia (AML) with 133. Papaemmanuil E, Dohner H, Campbell PJ. Genomic classifica-
normal cytogenetics. PLoS One. 2012;7:e42334. https://​doi.​ tion in acute myeloid leukemia. N Engl J Med. 2016;375:900–1.
org/​10.​1371/​journ​al.​pone.​00423​34. https://​doi.​org/​10.​1056/​NEJMc​16087​39.
116. Ward AF, Braun BS, Shannon KM. Targeting oncogenic Ras 134. Papaemmanuil E, et al. Genomic classification and prognosis
signaling in hematologic malignancies. Blood. 2012;120:3397– in acute myeloid leukemia. N Engl J Med. 2016;374:2209–21.
406. https://​doi.​org/​10.​1182/​blood-​2012-​05-​378596. https://​doi.​org/​10.​1056/​NEJMo​a1516​192.
117. Bollag G, et al. Loss of NF1 results in activation of the Ras 135. Johnson SM, et al. Acute myeloid leukemia with co-mutated
signaling pathway and leads to aberrant growth in haematopoi- ASXL1 and SRSF2 exhibits monocytic differentiation and has
etic cells. Nat Genet. 1996;12:144–8. https://​doi.​org/​10.​1038/​ a mutational profile overlapping with chronic myelomonocytic
ng0296-​144. leukemia. Hemasphere. 2019;3:e292. https://​doi.​org/​10.​1097/​
118. Zhang P, et al. Chromatin regulator Asxl1 loss and Nf1 hap- HS9.​00000​00000​000292.
loinsufficiency cooperate to accelerate myeloid malignancy. J 136. Pratcorona M, et al. Acquired mutations in ASXL1 in acute mye-
Clin Invest. 2018;128:5383–98. https://​doi.​org/​10.​1172/​JCI12​ loid leukemia: prevalence and prognostic value. Haematologica.
1366. 2012;97:388–92. https://d​ oi.o​ rg/1​ 0.3​ 324/h​ aemat​ ol.2​ 011.0​ 51532.
119. Gottfried ON, Viskochil DH, Couldwell WT. Neurofibromatosis 137. Haferlach T, et al. Landscape of genetic lesions in 944 patients
type 1 and tumorigenesis: molecular mechanisms and therapeutic with myelodysplastic syndromes. Leukemia. 2014;28:241–7.
implications. Neurosurg Focus. 2010;28:E8. https://​doi.​org/​10.​ https://​doi.​org/​10.​1038/​leu.​2013.​336.
3171/​2009.​11.​FOCUS​09221. 138. Carbuccia N, et  al. Mutual exclusion of ASXL1 and NPM1
120. Staser K, Yang FC, Clapp DW. Plexiform neurofibroma genesis: mutations in a series of acute myeloid leukemias. Leukemia.
questions of Nf1 gene dose and hyperactive mast cells. Curr Opin 2010;24:469–73. https://​doi.​org/​10.​1038/​leu.​2009.​218.
Hematol. 2010;17:287–93. https://​doi.​org/​10.​1097/​MOH.​0b013​ 139. Schnittger S, et al. ASXL1 exon 12 mutations are frequent in
e3283​39511b. AML with intermediate risk karyotype and are independently
121. Greenberg PL, et al. Revised international prognostic scoring associated with an adverse outcome. Leukemia. 2013;27:82–91.
system for myelodysplastic syndromes. Blood. 2012;120:2454– https://​doi.​org/​10.​1038/​leu.​2012.​262.
65. https://​doi.​org/​10.​1182/​blood-​2012-​03-​420489. 140. Park UH, Yoon SK, Park T, Kim EJ, Um SJ. Additional sex
122. Campbell PJ, et al. Mutation of JAK2 in the myeloproliferative comb-like (ASXL) proteins 1 and 2 play opposite roles in adi-
disorders: timing, clonality studies, cytogenetic associations, pogenesis via reciprocal regulation of peroxisome proliferator-
and role in leukemic transformation. Blood. 2006;108:3548–55. activated receptor {gamma}. J Biol Chem. 2011;286:1354–63.
https://​doi.​org/​10.​1182/​blood-​2005-​12-​013748. https://​doi.​org/​10.​1074/​jbc.​M110.​177816.
123. Campbell PJ, Green AR. The myeloproliferative disorders. N 141. Metzeler KH, et  al. ASXL1 mutations identify a high-
Engl J Med. 2006;355:2452–66. https://​doi.​org/​10.​1056/​NEJMr​ risk subgroup of older patients with primary cytogeneti-
a0637​28. cally normal AML within the ELN Favorable genetic cat-
124. Guo Y, et al. ASXL1 alteration cooperates with JAK2V617F to egory. Blood. 2011;118:6920–9. https:// ​ d oi. ​ o rg/ ​ 1 0. ​ 1 182/​
accelerate myelofibrosis. Leukemia. 2019;33:1287–91. https://​ blood-​2011-​08-​368225.
doi.​org/​10.​1038/​s41375-​018-​0347-y. 142. Bidikian A, et al. Prognostic impact of ASXL1 mutations in
125. Chen E, Mullally A. How does JAK2V617F contribute to the chronic phase chronic myeloid leukemia. Blood Cancer J.
pathogenesis of myeloproliferative neoplasms? Hematol Am Soc 2022;12:144. https://​doi.​org/​10.​1038/​s41408-​022-​00742-1.
Hematol Educ Program. 2014;268–276:2014. https://​doi.​org/​10.​ 143. Druker BJ, et al. Efficacy and safety of a specific inhibitor of the
1182/​ashed​ucati​on-​2014.1.​268. BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl

13
F.-C. Yang, J. Agosto‑Peña

J Med. 2001;344:1031–7. https://​doi.​org/​10.​1056/​NEJM2​00104​ 154. Kenderian SS, et al. CD33-specific chimeric antigen receptor
05344​1401. T cells exhibit potent preclinical activity against human acute
144. Druker BJ, et al. Activity of a specific inhibitor of the BCR-ABL myeloid leukemia. Leukemia. 2015;29:1637–47. https://​doi.​org/​
tyrosine kinase in the blast crisis of chronic myeloid leukemia 10.​1038/​leu.​2015.​52.
and acute lymphoblastic leukemia with the Philadelphia chromo- 155. Ting J, Lee AS. Human gene encoding the 78,000-dalton glu-
some. N Engl J Med. 2001;344:1038–42. https://d​ oi.o​ rg/1​ 0.1​ 056/​ cose-regulated protein and its pseudogene: structure, conserva-
NEJM2​00104​05344​1402. tion, and regulation. DNA. 1988;7:275–86. https://​doi.​org/​10.​
145. Huang ME, et al. Use of all-trans retinoic acid in the treatment 1089/​dna.​1988.7.​275.
of acute promyelocytic leukemia. Blood. 1988;72:567–72. 156. Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D.
146. Wang ZY, Chen Z. Acute promyelocytic leukemia: from highly Dynamic interaction of BiP and ER stress transducers in the
fatal to highly curable. Blood. 2008;111:2505–15. https://​doi.​ unfolded-protein response. Nat Cell Biol. 2000;2:326–32. https://​
org/​10.​1182/​blood-​2007-​07-​102798. doi.​org/​10.​1038/​35014​014.
147. Harrison C, et  al. JAK inhibition with ruxolitinib ver- 157. Hebbar N, et al. CAR T cells redirected to cell surface GRP78
sus best available therapy for myelofibrosis. N Engl J Med. display robust anti-acute myeloid leukemia activity and do
2012;366:787–98. https://​doi.​org/​10.​1056/​NEJMo​a1110​556. not target hematopoietic progenitor cells. Nat Commun.
148. Verstovsek S, et al. A double-blind, placebo-controlled trial of 2022;13:587. https://​doi.​org/​10.​1038/​s41467-​022-​28243-6.
ruxolitinib for myelofibrosis. N Engl J Med. 2012;366:799–807. 158. Fujino T, Goyama S, Sugiura Y, et al. Mutant ASXL1 induces
https://​doi.​org/​10.​1056/​NEJMo​a1110​557. age-related expansion of phenotypic hematopoietic stem cells
149. Pardanani A, Tefferi A. Targeting myeloproliferative neoplasms through activation of Akt/mTOR pathway. Nat Commun.
with JAK inhibitors. Curr Opin Hematol. 2011;18:105–10. 2021;12:1826. https://​doi.​org/​10.​1038/​s41467-​021-​22053-y.
https://​doi.​org/​10.​1097/​MOH.​0b013​e3283​439964. 159. Uni M, Masamoto Y, Sato T, et al. Modeling ASXL1 muta-
150. Kim T, et al. Exome sequencing reveals DNMT3A and ASXL1 tion revealed impaired hematopoiesis caused by derepression
variants associate with progression of chronic myeloid leukemia of p16Ink4a through aberrant PRC1-mediated histone modifi-
after tyrosine kinase inhibitor therapy. Leuk Res. 2017;59:142–8. cation. Leukemia. 2019;33:191–204. https://​doi.​org/​10.​1038/​
https://​doi.​org/​10.​1016/j.​leukr​es.​2017.​06.​009. s41375-​018-​0198-6.
151. Tyner JW, et al. Functional genomic landscape of acute myeloid
leukaemia. Nature. 2018;562:526–31. https://​doi.​org/​10.​1038/​ Publisher's Note Springer Nature remains neutral with regard to
s41586-​018-​0623-z. jurisdictional claims in published maps and institutional affiliations.
152. Kraehenbuehl L, Weng CH, Eghbali S, Wolchok JD, Merghoub
T. Enhancing immunotherapy in cancer by targeting emerging Springer Nature or its licensor (e.g. a society or other partner) holds
immunomodulatory pathways. Nat Rev Clin Oncol. 2022;19:37– exclusive rights to this article under a publishing agreement with the
50. https://​doi.​org/​10.​1038/​s41571-​021-​00552-7. author(s) or other rightsholder(s); author self-archiving of the accepted
153. Yu S, Yi M, Qin S, Wu K. Next generation chimeric antigen manuscript version of this article is solely governed by the terms of
receptor T cells: safety strategies to overcome toxicity. Mol Can- such publishing agreement and applicable law.
cer. 2019;18:125. https://​doi.​org/​10.​1186/​s12943-​019-​1057-4.

13

View publication stats

You might also like