In this issue of Blood, Li et al report an unexpected but clinically relevant finding. They demonstrate that the mixed lineage leukemia (MLL1) gene acts independently from menin (Men1) in the hematopoietic system.1 

The MLL gene encodes a DNA-binding protein that methylates histone H3 lysine 4 (H3K4) and regulates gene expression including multiple Hox genes. Leukemogenic MLL translocations encode MLL fusion proteins (MLL-FPs) that have lost H3K4 methyltransferase activity. An important feature of MLL-FPs is their ability to transform hematopoietic cells into leukemia stem cells. At least 52 functionally diverse MLL-FPs have been described. Fusions with the nuclear proteins AF4, AF9, AF10, ENL, and ELL account for most of MLL-rearranged leukemia. Translocations disrupting the MLL1 gene occur frequently in infant and secondary acute leukemia. MLL1 translocations encode fusion proteins (MLL1-FPs) retaining the N terminus of MLL1, which interacts with the tumor suppressor menin.2  Indeed, several studies have demonstrated that MLL1-FPs require menin interaction for leukemogenesis. Mutations in the menin interaction motif of MLL1-ENL abolish its transforming activity in hematopoietic cells.3  Second, menin is required for MLL1-FPs to bind a PWWP domain–containing protein that participates in targeting of the MLL complex to chromatin.4  Finally, Men1-deficient bone marrow progenitors do not sustain leukemogenesis induced specifically by MLL1-FPs,4  thus defining Men1 as an essential oncogenic cofactor for MLL-associated leukemogenesis.

Men1 and Mll1 perform several similar functions, and loss of either gene in embryonic stem cells impairs hematopoietic differentiation.5  However, because of the discovery of menin in native MLL1 complexes,6  and the colocalization of menin, MLL1/MLL2, and H3K4Me3 at most genomic loci, it was predicted that menin would be a necessary cofactor for the function of the MLL1 complex, irrespective of tissue or target genes examined.7 

Nevertheless, until this study, it was unclear whether the normal functions of MLL1 required in hematopoiesis also depend on menin interaction. It was anticipated that disruption of this interaction with a fundamental MLL1 cofactor would abolish normal hematopoiesis because wild-type MLL1 plays a major function in sustaining hematopoietic stem cells (HSCs).

In a very elegant approach, the authors demonstrate the lack of genetic interaction between Men1 and MLL1 in steady-state or regenerative hematopoiesis and in B-cell differentiation despite the fact that MLL1 is critical for these processes. They show that Men1 loss affects only a subset of Mll1 target genes and, more importantly, that these gene expression changes are not sufficient to impact HSC homeostasis. To evaluate whether a single copy of Mll1 in the complete absence of Men1 was sufficient to support hematopoietic regeneration, competitive transplantation experiments using Men1−/−: Mll1+/− donor cells, they demonstrate that both proteins function additively through independent pathways, which is opposed to the view that Men1 serves as an essential cofactor for MLL1 in regulation of HSC homeostasis.8 

To determine the effect of disrupting the menin-MLL1 interaction on B-cell differentiation, they introduced a blocking peptide into primitive bone marrow cells and assessed B-cell growth and differentiation. They demonstrate that menin and MLL1 play important roles independently during B-cell differentiation but control largely independent genetic networks. Importantly, disrupting the menin-MLL1 interaction cannot recapitulate the block in B-cell differentiation observed in individual knockouts despite efficient reduction in shared target gene expression.

This study has important clinical implications and elucidates the importance of inhibition of the menin interaction with MLL-FPs.9  Specific inhibition could represent a very promising strategy to reverse the oncogenic activity of MLL1-FPs. These new results provide the rational and essential understanding to design safe inhibitors for effectively targeting the menin-MLL interaction in leukemia with minimal effects in the crucial physiological function of MLL1 in maintaining normal hematopoiesis. However, the Men1 tumor suppressive role in neuroendocrine cells could rely on the MLL-Men1 interaction. Disruption of this interaction could lead to the abrogation of Men1 tumor suppression activity in pancreatic cells. Therefore, it will be important to determine whether there is a risk associated with inhibiting the tumor suppression activity achieved by Men1.10 

Conflict-of-interest disclosure: The author declares no competing financial interests.

1
Li
 
BE
Gan
 
T
Meyerson
 
M
Rabbitts
 
TH
Ernst
 
P
Distinct pathways regulated by menin and by MLL1 in hematopoietic stem cells and developing B cells.
Blood
2013
, vol. 
122
 
12
(pg. 
2039
-
2046
)
2
Krivtsov
 
AV
Armstrong
 
SA
MLL translocations, histone modifications and leukaemia stem-cell development.
Nat Rev Cancer
2007
, vol. 
7
 
11
(pg. 
823
-
833
)
3
Yokoyama
 
A
Somervaille
 
TC
Smith
 
KS
Rozenblatt-Rosen
 
O
Meyerson
 
M
Cleary
 
ML
The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis.
Cell
2005
, vol. 
123
 
2
(pg. 
207
-
218
)
4
Yokoyama
 
A
Cleary
 
ML
Menin critically links MLL proteins with LEDGF on cancer-associated target genes.
Cancer Cell
2008
, vol. 
14
 
1
(pg. 
36
-
46
)
5
Maillard
 
I
Hess
 
JL
The role of menin in hematopoiesis.
Adv Exp Med Biol
2009
, vol. 
668
 (pg. 
51
-
57
)
6
Yokoyama
 
A
Wang
 
Z
Wysocka
 
J
, et al. 
Leukemia proto-oncoprotein MLL forms a SET1-like histone methyltransferase complex with menin to regulate Hox gene expression.
Mol Cell Biol
2004
, vol. 
24
 
13
(pg. 
5639
-
5649
)
7
Agarwal
 
SK
Jothi
 
R
Genome-wide characterization of menin-dependent H3K4me3 reveals a specific role for menin in the regulation of genes implicated in MEN1-like tumors.
PLoS ONE
2012
, vol. 
7
 
5
pg. 
e37952
 
8
Smith
 
E
Lin
 
C
Shilatifard
 
A
The super elongation complex (SEC) and MLL in development and disease.
Genes Dev
2011
, vol. 
25
 
7
(pg. 
661
-
672
)
9
Caslini
 
C
Yang
 
Z
El-Osta
 
M
Milne
 
TA
Slany
 
RK
Hess
 
JL
Interaction of MLL amino terminal sequences with menin is required for transformation.
Cancer Res
2007
, vol. 
67
 
15
(pg. 
7275
-
7283
)
10
Karnik
 
SK
Hughes
 
CM
Gu
 
X
, et al. 
Menin regulates pancreatic islet growth by promoting histone methylation and expression of genes encoding p27Kip1 and p18INK4c.
Proc Natl Acad Sci USA
2005
, vol. 
102
 
41
(pg. 
14659
-
14664
)
Sign in via your Institution