From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

Blood First Edition Paper, prepublished online April 1, 2015; DOI 10.1182/blood-2014-12-618090

TGF-β signaling in the control of hematopoietic stem cells

Ulrika Blank and Stefan Karlsson

Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund

University Hospital, Lund, Sweden

Corresponding author:

Ulrika Blank

E-mail: [email protected]

Phone: +46-46-2220589

Fax: +46-46-2220568

BMC A12, 221 84 Lund, Sweden

β

Short title: Regulation of HSCs by TGF-

Copyright © 2015 American Society of Hematology

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

Abstract Blood is a tissue with high cellular turnover, and its production is a tightly

orchestrated process that requires constant replenishment. All mature blood

cells are generated from hematopoietic stem cells (HSCs), which are the self-

renewing units that sustain life-long hematopoiesis. HSC behavior, such as self-

renewal and quiescence, are regulated by a wide array of factors, including

external signaling cues present in the bone marrow. The Transforming Growth

β (TGF-β) family of cytokines constitutes a multifunctional signaling

Factor-

circuitry, which regulates pivotal functions related to cell fate and behavior in

β signaling

virtually all tissues of the body. In the hematopoietic system, TGF-

controls a wide spectrum of biological processes, from homeostasis of the

immune system to quiescence and self-renewal of HSCs. Here, we review key

β and downstream signaling

features and emerging concepts pertaining to TGF-

pathways in normal HSC biology, featuring aspects of aging, hematological

disease, and how this circuitry may be exploited for clinical purposes in the

future.

2

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

Introduction Blood cell production takes place in the bone marrow (BM) of adult individuals,

where it originates from a rare pool of tissue-specific stem cells known as

hematopoietic stem cells (HSCs). HSCs function to sustain and regenerate the

entire blood system in an unbroken fashion throughout life, as they have the dual

capacity to self-renew and differentiate to all blood cell lineages

1,2.

Self-renewal

pertains to the ability of a stem cell to duplicate itself without losing

developmental potential. This capacity is crucial for maintenance of the stem cell

pool and may occur in a symmetric or asymmetric fashion

3.

Furthermore,

quiescence, or withdrawal from the cell cycle, is a feature of HSCs that provides

the blood system with a dormant HSC reservoir that retains the ability to self-

renew and replenish all blood lineages. Hematopoiesis is hierarchically

organized, with the most immature pool of HSCs at the top, giving rise to various

progenitor cells that progressively lose self-renewal capacity as they form

differentiating progeny that proliferate extensively, finally generating functional

blood cells at the bottom of the hierarchy (Figure 1). To rapidly tailor blood

production to acute changes, such as bleeding and infection, HSC behavior is

tightly regulated yet highly flexible

4.

Signals that promote quiescence, and cues

that stimulate proliferation and differentiation, provide a fine-balanced system

that safeguards against depletion and overproduction of HSCs. Disruption of

these regulatory mechanisms can result in blood disease, such as BM failure or

β (TGF-β) is the founding member of a

leukemia. Transforming Growth Factor-

large family of secreted polypeptide growth factors, consisting of over 30

members in humans, including activins, Bone Morphogenetic Proteins (BMPs),

and others

5.

β family constitutes a multifunctional set of cytokines that

The TGF-

3

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

regulate a bewildering array of cellular processes during development and

β members regulate tissue homeostasis and

beyond. In the adult organism, TGF-

β plays an important role in

regeneration. With respect to hematopoiesis, TGF-

regulating HSC behavior, particularly quiescence and self-renewal. Although

many members of this family function to regulate hematopoiesis, we focus this

β.

review mainly on TGF-

The basic elements of TGF-β signaling TGF-β ligands signal through cell surface serine/threonine kinase receptors,

known as type I and type II receptors

6.

In vertebrates seven different type I

receptors (ALK1-7) and five distinct type II receptors have been identified,

serving the entire family of ligands

6.

β signals mainly via the type I receptor,

TGF-

βRII, both of which are required for signaling

ALK5, and the type II receptor, T

activation. Following ligand binding and receptor phosphorylation, the SMAD

signaling circuitry becomes activated (Figure 2)

6.

The SMAD proteins are a

family of transcription factors consisting of eight members, SMAD1-8, which are

further subdivided into three classes based on structural and functional

properties

7.

Receptor-regulated SMADs (R-SMADs), SMAD1, 2, 3, 5, and 8, are

the only SMADs directly phosphorylated and activated by the kinase domain of

type I receptors. Upon phosphorylation, R-SMADs form a complex with the

common-SMAD (Co-SMAD), SMAD4, resulting in nuclear accumulation of

activated complexes. In the nucleus, R-SMAD-SMAD4 complexes cooperate with

transcriptional co-regulators that further define target gene recognition and

transcriptional regulation

7.

The inhibitory SMADs (I-SMADs), SMAD6 and

SMAD7, constitute the third class, which function to inhibit TGF-β signaling. TGF-

β/activin/nodal and BMP/GDF employ different subsets of R-SMADs. R-

4

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

SMAD2/3 specifically relay signals from TGF-β and activin receptors, whereas R-

SMAD1/ 5/8 primarily operate downstream of BMP receptors

6.

Alternative pathways The SMAD pathway forms a fundamental signaling module and is the most well

β. However, activation of other pathways,

studied circuitry downstream of TGF-

β activated kinase 1 (TAK1), a component of the mitogen-

most notably TGF-

activated protein kinase (MAPK) pathway, has been observed in other cell types

8,9.

Depending on context, TAK1 activates a host of downstream transducers,

including p38 and JNK

10.

Conditional deletion of TAK1 in mice, using the

Mx-Cre

β has not been firmly

driver, results in hematopoietic failure, but the link to TGF-

established

11.

Other non-SMAD circuitries regulated by TGF-β include ERK,

PI3K-AKT-FOXO, and RHO-like small GTPases, though the contribution of most of

10.

these mechanisms are not well defined in HSCs (Figure 2)

In addition,

γ (TIF1γ) partners with SMAD2/3 (see

transcriptional intermediary factor-1

separate section below)

12.

The TGF-β pathway is intimately linked with other

signaling circuitries, for readers interested in crosstalk mechanisms we refer to

the following papers

13-17.

TGF-β: inducer of quiescence TGF-β is catalogued as one of the most potent inhibitors of HSC growth

and a variety of culture systems support this notion

18-21.

in vitro

During homeostatic

conditions, the majority of HSCs are in a quiescent cell-cycle state

22,23.

Quiescence is related to maintenance of the HSC pool and loss thereof results in

exhaustion and erosion of HSC function

24.

β has been

Naturally, TGF-

hypothesized to be a cardinal regulator of HSC dormancy, maintaining a pool of

quiescent HSCs

in vivo

. Indeed, neutralization of TGF-β

5

in vitro

releases early

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

hematopoietic progenitor cells from quiescence

25-27.

Several molecular

mechanisms have been proposed to account for TGF-β-mediated growth

inhibition, including alterations in cytokine receptor expression and

Ink4b,

upregulation of cyclin-dependent kinase inhibitors (CDKIs), such as p15

Cip1,

p21

Kip1 26,28-34.

and p27

β can exert growth inhibitory actions

However, TGF-

Cip1 and

Kip1 35.

independent of p21

p27

+ cells,

In human CD34

Kip2,

cell-cycle arrest occurs through upregulation of p57

CDKI family

36.

Kip2 is

β -mediated

TGF-

another member of the

-

Similarly, p57

+

-

+

highly enriched in mouse CD34 Kit Lin Sca1

+

(KLS) cells as opposed to the more mature and actively cycling CD34 KLS

fraction

37.

Kip2 correlates with

Interestingly, a high level of p57

the activation

-

status of SMAD2/3, which are uniquely phosphorylated in freshly isolated CD34

+

KLS cells but not in CD34 KLS progenitors

Kip2 in

p57

-

CD34 KLS cells

38.

β upregulates

Additionally, TGF-

in vitro 38

. These findings point to a mechanism where

β functions to induce p57Kip2 within the most primitive HSC compartment,

TGF-

thus promoting their quiescent state

in vivo

(Figure 3).

Heterogeneity of the HSC pool Recently, evidence has accumulated suggesting that the adult HSC compartment

consists of a number of functionally distinct subsets with diverse self-renewal

and differentiation potentials

23,39-41.

Interestingly, discrete HSC subtypes

β, according to a model proposed by Challen and

respond differently to TGF-

colleagues

42.

β stimulates proliferation of myeloid-biased HSCs

Specifically, TGF-

(My-HSCs) whereas lymphoid-biased HSCs (Ly-HSCs) are growth inhibited

(Figure 3)

42.

β on HSCs is more nuanced than previously

Thus, the effect of TGF-

thought, such that proliferation is induced in certain HSC subtypes at certain

6

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

β represents a potential signal for

concentrations. However, though TGF-

differential regulation between HSC subtypes, there is currently no tangible

explanation for the underlying molecular mechanism. Another study found that

the RNA-binding protein MUSASHI-2 is required for HSCs to mount a

β at low concentrations 43. Loss of MUSASHI-2

proliferative response to TGF-

leads to impaired HSC quiescence, reduced My-HSC numbers and myeloid output

in vivo

Kip2 and

. These findings are coupled to significant reductions in p57

phosphorylated SMAD2/3 in HSCs, suggesting that MUSASHI-2 modulates the

β signaling circuitry 43. Most work regarding TGF-β in hematopoiesis has

TGF-

β1. However, TGF-β exists in three isoforms that are encoded by

focused on TGF-

separate genes,

Tgfb1-3

β 1-3 share significant sequence

. Although TGF-

homology and signal through the same receptor complex

44,45,

differences in

receptor affinity exist and varying responses have been reported. Most notably

β2, being growth inhibited at high

KLS cells exhibit a biphasic response to TGF-

doses and stimulated at low concentrations

46.

β

The bidirectional effects of TGF-

on proliferation vs. quiescence further substantiate the complexity of this

signaling pathway, and it remains to be clarified whether or not the SMAD

pathway is differentially regulated at high and low doses and between diverse

HSC subtypes. Additionally, it is conceivable that the bidirectional effects are a

β receptor expression among HSC subtypes,

function of contrasting TGF-

translating to dosage effects that generate qualitatively different responses. This

theory is corroborated by a recent study, which shows that a high level of ALK5

correlates with increased myeloid output

7

47.

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

TGF-β and the aging hematopoietic system The hematopoietic system declines functionally with age, a phenomenon that can

be traced back to alterations in the HSC compartment. The clonal composition of

the HSC pool changes over time, such that My-HSCs become more abundant at

the expense of Ly-HSCs

48.

Physiologically, this translates to a myeloid-biased

hematopoietic system with reduced lymphoid potential. Furthermore, the risk of

hematological malignancies, anemia, autoimmunity, and inflammatory disorders,

increases with age

48.

As TGF-β differentially regulates My- and Ly-HSCs, the

connection to aging is evident, especially since TGF-β is implicated in the aging

process of non-hematopoietic tissues

49,50.

A recent study, which profiled the

transcriptome, DNA methylome, and histone modifications in highly purified

young and old HSCs, found significant changes in the TGF-β pathway at the

transcriptional level

51.

In fact, the TGF-β pathway represented 19 percent of

differentially expressed genes in young vs. old HSCs. The study shows changes in

multiple layers of the TGF-β pathway, from extracellular regulators, to SMAD

transcription factors, and cofactors, resulting in an overall reduction of TGF-β

signaling in aged HSCs

51.

proliferation of My-HSCs

Since low concentrations of TGF-β stimulates

in vitro

, downsizing the TGF-β circuitry may provide a

more proliferative environment for My-HSCs, which can expand over time.

Similarly, the stimulatory effect of TGF-β2 on the much cruder KLS population

increases with age

52.

This is logical, as the aged KLS compartment should

contain a larger proportion of My-HSCs. However, the relationship between TGF-

β and aging is complex, as evidenced by a study of TIF1-γ. In HSCs, the

expression of TIF1-γ decreases with age, consistent with an accelerated aging

phenotype of TIF1-γ knockout mice

47.

Additionally, TIF1-γ controls the turnover

8

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

+ HSCs

of ALK5, such that ALK5

become more abundant with age

aged HSCs are more sensitive to the cytostatic effect of TGF-β

a reflection of more generous ALK5 expression.

47.

Interestingly,

in vivo,

possibly as

As TIF1-γ is itself part of the

signal transduction machinery downstream of TGF-β, the balance between TIF1-

γ and SMAD4-mediated transcriptional responses may play an additional role,

which was not investigated. In sum, perturbations of TGF-β signaling represent a

potential mechanism that contributes to HSC aging.

TGF-β and the bone marrow niche β in the regulation of HSCs, requires an

The quest to decipher the role of TGF-

understanding of the BM niches that house HSCs. Due to the anatomical

complexity of the BM, much of our knowledge concerning the HSC

microenvironment has for long remained nebulous and it is only in the last

decade that we have gained a more detailed appreciation of the regulatory

elements of the BM environment, although Ray Schofield proposed the concept

of the BM niche already as far back as 1978

53.

An increasing number of niche

components have now been identified, revealing a complex network of cell-cell

interactions, extracellular elements, signaling cues, and structures

54,55.

Together,

these entities create specialized niches that function to maintain and control self-

β is part of this microenvironment,

renewing HSCs in a coordinated manner. TGF-

as it is produced by a variety of cell types in the BM, and large quantities of latent

β are deposited into bone matrix 56,57. However, according to one study,

TGF-

surprisingly few BM cells exhibit significant phosphorylation of SMAD2/3 at

β is tightly regulated

steady state, suggesting that activation of latent TGF-

spatially

58.

Nonmyelinating Schwann cells, which ensheath peripheral nerves

and lay in parallel with blood vessels in the BM, are proposed as one of the major

9

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

β activation in the BM 58. These glial cells regulate the activation

sources for TGF-

β by binding latent TGF-β via integrin β8 on the cell surface, thus

process of TGF-

β to proteolytic cleavage by

promoting activation by exposing TGF-

metalloproteinases

58.

A sizable portion of HSCs is in direct contact with

nonmyelinating Schwann cells in the BM, and denervation results in loss of HSCs

β are megakaryocytes, a

and increased cell cycling. Another major source of TGF-

cell type that physically associates with approximately 20 percent of HSCs in the

BM

59,60.

HSCs in close proximity to megakaryocytes exhibit activation of

SMAD2/3 and ablation of megakaryocytes results in reduced phosphorylation of

SMAD2/3 as well as loss of quiescence and increased HSC proliferation

Importantly, conditional deletion of

increased cell cycling of HSCs

60.

Tgfb1

60.

in megakaryocytes results in

The anatomical relationship between

megakaryocytes and Schwann cells have not been investigated, but the

β provides an important

conclusion from these studies is that, indeed TGF-

quiescence signal to HSCs in the BM niche. Additionally, the conclusion must be

β production and

that more than one cell type in the BM contributes to TGF-

signaling in HSCs (Figure 4).

Lessons from in vivo models TGF-β can affect most cell types throughout the hematopoietic hierarchy, but the

response is modulated by context and differentiation stage of the target cell.

Therefore, TGF-β generates highly variable biological outcomes and can affect

proliferation, differentiation, and apoptosis in both positive and negative

directions

21,61-63.

β

Much of our knowledge of the physiological relevance of TGF-

has been gained by studying knockout mouse models. Based on these reports, we

know that TGF-β is a principal regulator of immune cell homeostasis and

10

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

function

in vivo

. Importantly, both

Tgfb1

-ligand and receptor knockout mice

develop a lethal inflammatory disorder

64-66.

With respect to hematopoiesis,

Tgfb1-

null mice exhibit enhanced myelopoiesis, suggesting that TGF-β acts as a

negative regulator of myelopoiesis

in vivo 65

. Upon analysis before the onset of

inflammation, a host of HSC properties are altered in

Most significantly, BM cells from

Tgfb1

knockout mice

67.

Tgfb1

-deficient neonates exhibit impaired

reconstitution ability upon transplantation, a finding attributed to defective

homing

67.

However, mice deficient in ALK5 display normal HSC self-renewal and

regenerative capacity

in vivo

, even under extreme hematopoietic stress with no

defects in homing capacity

68,69.

show increased HSC cell cycling

transplantation

58.

β RII conditional knockout mice

In contrast, T

in vivo

, and reduced regenerative capacity upon

Because of the severe inflammatory disease that develops in

β signaling deficient mouse models, most studies have employed different

TGF-

strategies to overcome disease progression, including a variety of immune-

deficient genetic backgrounds. This may account for the differences observed

βRII

between knockout models. Additionally, signals may emanate from T

independently of ALK5, as has been shown in non-hematopoietic cell types

where the cell polarity regulator partitioning defective 6 (PAR6) is

βRII 70,71. Further, the fact that TβRII is more highly

phosphorylated directly by T

expressed within HSCs compared to ALK5 might be an additional contributing

factor

57.

Thus, there are both overlapping and non-overlapping phenotypes

between knockout models and it appears to be critically important at which level

β signaling is disrupted. This notion is supported by a

and for how long TGF-

β was

recent study, in which a neutralizing antibody against active TGF-

administered to mice following 5-FU treatment

11

72.

β blockade after

TGF-

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

chemotherapy results in enhanced hematopoietic regeneration by delaying the

β-SMAD2-

return of HSCs to quiescence. These findings imply that the TGF-

Kip2 signaling axis

p57

is important for reestablishing quiescence of HSCs

following stress, once sufficient regeneration of the hematopoietic system has

been attained. Additionally, overexpression of SMAD4 sensitizes human cord

β , resulting in reduced regenerative

blood-derived candidate HSCs to TGF-

capacity

in vivo 73

. Together, these findings suggest that transient inhibition of

β might be a feasible strategy to enhance hematopoietic recovery in patients

TGF-

β coupled with a

following chemotherapy. Due to the multifaceted nature of TGF-

potentially complex set of redundant mechanisms, its role as a critical regulator

of HSC quiescence

in vivo

has been difficult to unveil. However, current dogma

β is indeed an important quiescence signal in vivo.

now suggests that TGF-

A role for SMAD signaling in self-renewal of HSCs β , two approaches

To block the SMAD signaling network downstream of TGF-

have been used: overexpression of the inhibitory SMAD7 and deletion of

Smad4

.

In murine HSCs, forced expression of SMAD7 results in increased self-renewal of

HSCs, indicating that the SMAD pathway negatively regulates self-renewal

74.

in vivo

Importantly, differentiation was unperturbed in this model, suggesting that

self-renewal is regulated independently of differentiation by SMAD signaling. In

contrast, overexpression of SMAD7 in human SCID repopulating cells (SRCs)

results in altered differentiation from lymphoid dominant engraftment toward

increased myeloid contribution

75.

Thus, in the xenograft model system, forced

expression of SMAD7 modulates differentiation of multipotent human SRCs.

Using a conditional knockout mouse model, disruption of the SMAD pathway at

the level of SMAD4 was investigated. Intriguingly,

12

Smad4

-deficient HSCs display

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

a reduced repopulative capacity of primary and secondary recipients, indicating

that SMAD4 is critical for HSC self-renewal

SMAD7 vs. deletion of

Smad4

in vivo 76

. Since overexpression of

is anticipated to yield similar hematopoietic

phenotypes, it is conceivable that SMAD4 functions as a positive regulator of self-

renewal independently of its role in the TGF-β pathway

13-15.

Alternatively,

SMAD7 may have unanticipated functions in an overexpression setting that

impinge positively on self-renewal.

Variations on TGF-β signaling: a role for TIF1γ SMAD4 has traditionally been viewed as the nexus of SMAD signaling as it

β/activin and BMP signaling

functions as a core component of both TGF-

γ 12. In human

branches. However, SMAD2/3 can also partner with TIF1

β balances erythroid differentiation

hematopoietic stem/progenitor cells, TGF-

with growth inhibition, in a mechanism dependent on competitive binding

γ to SMAD2/3 12. In response to TGF-β , the TIF1γ-

between SMAD4 and TIF1

SMAD2/3 complex stimulates erythroid differentiation whereas SMAD2/3 in

association with SMAD4 leads to growth inhibition of human hematopoietic

γ

progenitors (Figure 2). Interestingly, the zebrafish homolog of TIF1 , encoded by

moonshine

, is essential for blood formation with mutants displaying severe red

γ is required for erythroid development 77.

cell aplasia, indicating that TIF1

γ is implicated in regulation of transcription elongation of

Furthermore, TIF1

γ functions to release

erythroid genes. The proposed model suggests that TIF1

paused Pol II at erythroid genes by recruiting positive elongation factors to the

blood-specific transcriptional complex, thus promoting transcription

78.

also functions at the erythroid/myeloid lineage bifurcation by modulating

13

γ

TIF1

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

GATA1 and PU.1 expression

79.

γ and TGF-β signaling has

The link between TIF1

γ functions as a

been delineated in detail in embryonic stem cells. There, TIF1

γ

chromatin reader where TIF1 -SMAD2/3 recognizes certain repressive histone

marks, promoting a transition to open chromatin that allows SMAD4-SMAD2/3

to gain access to DNA

80.

γ is needed for certain SMAD4-SMAD2/3

Thus, TIF1

transcriptional responses.

TGF-β in hematopoietic disease

Leukemia β’s pronounced cytostatic effect on HSCs in vitro and the fact that

Despite TGF-

β pathway are frequently

mutations in genes encoding components of the TGF-

found in other neoplasms, such as pancreatic and colon cancer

81,82,

inactivating

mutations are relatively uncommon for this pathway in hematological

malignancies

SMAD4

and

83.

Nevertheless, a number of cases have been reported involving

TGFBR2

in patients with acute myelogenous leukemia (AML)

84-87.

For example, in a study investigating human AML genomes, various copy number

alterations were found recurrently modified, one of which represented the

SMAD4 88 TGFBR1 TGFBR2 deletion of

. Furthermore, reports of sporadic mutations in both

and

in lymphoid neoplasms exist, and loss of SMAD3 is

associated with T-cell acute lymphocytic leukemia (ALL)

for the SMAD3-deficiency is not known since the

no mutations could be detected in the

MADH3

SMAD3

89-91.

The mechanism

mRNA was present and

gene, which encodes SMAD3

91.

However, T-cell leukemogenesis is promoted in mice with haplo-insufficiency of

Smad3

and a complete deficiency of

because the

p27Kip1

p27Kip1 91

. These findings are interesting

gene is frequently mutated in pediatric ALL, due to

14

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

translocations and deletions or germline mutations

92,93.

β

Impaired TGF-

signaling in hematological malignancies can also be caused by suppression of

SMAD-dependent transcriptional responses by oncoproteins like TAX, EVI-1, and

AML1-ETO

94-96.

Similarly, downregulation of the transcription factor ZEB1 and

β 1-mediated growth

overexpression of SMAD7 contribute to resistance to TGF-

suppression in adult T-cell leukemia/lymphoma without known mutations in

β pathway genes 97. In addition, there are several reports on oncoproteins,

TGF-

which generate leukemia and simultaneously neutralize the growth inhibitory

signal of the SMAD pathway by binding to or interacting with SMADs. Fusion

oncoproteins, like TEL-AML1 and AML1-EVI1, bind to SMAD3, impairing both

β signaling and apoptosis of transduced HSCs in vitro 95,98-100. Furthermore,

TGF-

SMAD4 physically associates with HOXA9, reducing its ability to regulate

transcriptional targets in hematopoietic cells

in vitro 101 In vivo .

studies show

that in wild-type mice overexpressing HOXA9 or NUP98-HOXA9, SMAD4 binds

the oncoproteins and sequestrates them to the cytoplasm, suggesting that

SMAD4 plays a protective role against further promotion and growth of leukemic

cells

102.

Therefore, SMAD signaling can be reduced or neutralized in

hematopoietic malignancies, but in a majority of cases this is not due to

mutations in genes of the circuitry itself, but rather through altered expression

β

or function of co-factors and oncoproteins, or alternatively via loss of TGF-

target genes.

The diseased bone marrow microenvironment β plays a major role as tumor suppressor, TGF-β can paradoxically

Although TGF-

facilitate tumor growth, particularly in the later stages of disease. This is due to

effects on the tumor microenvironment and the immunosuppressive function of

15

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

β. Interestingly, recent findings show that there is considerable interplay

TGF-

between leukemic cells and the BM microenvironment. In fact, the BM

microenvironment undergoes substantial remodeling by signals from leukemic

cells

103.

Similarly, alterations in niche cells can trigger hematopoietic disease

and promote leukemia

104,105.

Furthermore, activation of the parathyroid

hormone (PTH) receptor specifically in osteoblastic cells results in remodeling of

the BM microenvironment

106.

The remodeled niche attenuates

BCR-ABL1

oncogene-induced chronic myeloid leukemia (CML)-like disease, via up-

β 1, whereas MLL-AF9 oncogene-induced AML is enhanced.

regulation of TGF-

These data differ from the findings by Naka

et al

β, via regulation

., in which TGF-

of AKT and FOXO3a, instead maintains leukemia-initiating cells in CML

107.

The

β dosage and context,

discrepancies may be attributed to differences in TGF-

β sensitivity across different types of leukemia vary.

indicating that TGF-

β is part of the leukemic BM

Nevertheless, the findings point to the fact that TGF-

niche and strategies to target the BM microenvironment may be a promising

approach to reduce certain types of leukemic stem cells in the future. On this

β has been proposed to contribute to myelofibrosis, which is a chronic

note, TGF-

myeloproliferative disease characterized by clonal proliferation and bone

marrow fibrosis

108.

Several cytokines are thought to contribute towards

accumulation of reticulin fibers in the BM of patients with myelofibrosis. These

β , fibroblast growth factor 2 (FGF-2), and platelet-derived growth

include TGF-

factor

109-112.

Some of the most compelling evidence for a prominent role of TGF-

β in myelofibrosis comes from a study using Tgfb1-null mice. To induce myelofibrosis, irradiated mice were transplanted with BM cells transduced with

16

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

vectors containing the thrombopoietin (TPO) gene. Importantly, prominent

myelofibrosis developed in mice receiving transduced wild-type cells but not

Tgfb1

-null cells

111.

β 1, produced by hematopoietic

These data indicate that TGF-

cells, is a crucial component in the development of myelofibrosis, and underlines

further the interplay between hematopoietic cells and the BM microenvironment.

TGF-β in the pathogenesis of MDS Myelodysplastic syndromes (MDSs) encompass a spectrum of clonal stem cell

disorders characterized by inefficient hematopoiesis and reduced peripheral

blood counts. Excessive activation of inhibitory pathways, such as TGF-β, has

been proposed to amplify the inefficient blood production inherent to MDS

+ BM

For example, SMAD2 is upregulated and over-activated in CD34

113.

progenitors

from MDS patients. Importantly, pharmacologic inhibition of the TGF-β pathway

in vivo

, using a small molecule inhibitor of ALK5, alleviates anemia in a mouse

model of MDS

114.

Furthermore, SMAD7 is reduced in BM progenitors from MDS

β signaling 115. The reduction of

patients, leading to over-activation of TGF-

SMAD7 is attributed to increased levels of microRNA-21 (miR-21), which binds

to the 3´UTR of

SMAD7 116

.

Administration of a chemically modified inhibitor of

miR-21 results in increased red blood cell counts, using the same mouse model

as mentioned above

116.

Together, these studies implicate TGF-β in the

pathogenesis of MDS and suggest that this pathway may be a realistic

therapeutic target in a subset of MDSs.

Therapeutic avenues and future perspectives HSCs are the therapeutic units of BM transplantations, and as such by far the

most widely used application of stem cell therapy to date. Despite this, strategies

to improve efficiency and applicability of HSC transplantations are needed,

17

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

including expansion of HSCs

ex vivo

and techniques to improve engraftment and

regeneration post transplantation. Although BMPs have been reported to

contribute to HSC expansion

ex vivo,

manipulating SMAD signaling alone is

117.

unlikely to result in effective expansion

Another approach, with substantial

β

promise that should be investigated further, is to transiently block TGF-

signaling

in vivo

chemotherapy

to boost regeneration and recovery of hematopoiesis following

72.

Similarly, a number of recent studies highlight the feasibility of

β pathway in hematopoietic disorders like MDS and

manipulating the TGF-

anemia

115,118,119.

In addition, the complexity of the SMAD pathway continues to

be exposed as new layers of regulatory mechanisms are found. For example, the

linker region of SMADs is subject to negative regulation by GSK3 (glycogen

synthase kinase 3), FGF, and EGF

120-123.

The importance of this type of crosstalk

in hematopoietic cells is unknown, but it is worth mentioning as megakaryocytes

β and FGF output, thereby regulating

were recently shown to balance TGF-

hematopoietic regeneration following 5-FU challenge

in vivo 60

. Thus,

β signaling is relevant to several aspects of hematopoiesis. In

manipulating TGF-

the future, more detailed mechanistic studies are required to precisely define

β signaling may be manipulated in relation to other signaling

how TGF-

circuitries, ultimately improving HSC regeneration and hematopoietic disease

vivo

.

18

in

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

Acknowledgments The authors apologize to those whose work was not cited due to space

limitations.

This work was supported by the European Commission (Stemexpand); Hemato-

Linné and Stemtherapy program project grants from the Swedish Research

Council; a project grant to S.K from the Swedish Research Council; Swedish

Cancer Society; Swedish Children Cancer Foundation; Clinical Research Award

from Lund University Hospital; and a grant from The Tobias Foundation

awarded by the Royal Academy of Sciences. (S.K.).

Authorship Contribution: U.B. and S.K. wrote the review, and U.B. designed the figures.

Conflict-of-interest disclosure: The authors declare no competing financial

interests.

Correspondence: Stefan Karlsson, Division of Molecular Medicine and Gene

Therapy, BMC A12, 221 84 Lund, Sweden; email: [email protected]; and

Ulrika Blank, Division of Molecular Medicine and Gene Therapy, BMC A12, 221

84 Lund, Sweden; email: [email protected].

19

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

References 1.

Blood

Ogawa M. Differentiation and proliferation of hematopoietic stem cells.

2.

Orkin SH, Zon LI. Hematopoiesis: an evolving paradigm for stem cell

. 1993;81(11):2844-2853.

biology. 3.

Cell Nature

. 2008;132(4):631-644.

Zon LI. Intrinsic and extrinsic control of haematopoietic stem-cell self-

renewal. 4.

. 2008;453(7193):306-313.

Rossi L, Lin KK, Boles NC, et al. Less is more: unveiling the functional core

Cell Stem Cell Nature reviews Molecular cell Annu Rev Biochem Genes Dev

of hematopoietic stem cells through knockout mice. 2012;11(3):302-317. 5.

Massague J. TGFbeta signalling in context.

6.

Massague J. TGF-beta signal transduction.

biology

. 2012;13(10):616-630.

1998;67:753-791. 7.

.

.

Massague J, Seoane J, Wotton D. Smad transcription factors.

.

2005;19(23):2783-2810. 8.

Blank U, Brown A, Adams DC, Karolak MJ, Oxburgh L. BMP7 promotes

proliferation of nephron progenitor cells via a JNK-dependent mechanism.

Development

. 2009;136(21):3557-3566.

9.

Yamaguchi K, Shirakabe K, Shibuya H, et al. Identification of a member of

the MAPKKK family as a potential mediator of TGF-beta signal transduction.

Science Res

. 1995;270(5244):2008-2011.

10.

Mu Y, Gudey SK, Landstrom M. Non-Smad signaling pathways.

Cell Tissue

. 2012;347(1):11-20.

11.

Tang M, Wei X, Guo Y, et al. TAK1 is required for the survival of

hematopoietic cells and hepatocytes in mice. 12.

J Exp Med

. 2008;205(7):1611-1619.

He W, Dorn DC, Erdjument-Bromage H, Tempst P, Moore MA, Massague J.

Hematopoiesis controlled by distinct TIF1gamma and Smad4 branches of the TGFbeta pathway. 13.

Cell

. 2006;125(5):929-941.

Nishita M, Hashimoto MK, Ogata S, et al. Interaction between Wnt and

TGF-beta signalling pathways during formation of Spemann's organizer.

Nature

.

2000;403(6771):781-785. 14.

Labbe E, Letamendia A, Attisano L. Association of Smads with lymphoid

enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-beta and wnt pathways.

A

Proc Natl Acad Sci U S

. 2000;97(15):8358-8363.

15.

Itoh F, Itoh S, Goumans MJ, et al. Synergy and antagonism between Notch

and BMP receptor signaling pathways in endothelial cells.

Embo J

.

2004;23(3):541-551. 16.

Emmrich S, Rasche M, Schoning J, et al. miR-99a/100~125b tricistrons

regulate hematopoietic stem and progenitor cell homeostasis by shifting the balance between TGFbeta and Wnt signaling. 17.

Genes Dev

. 2014;28(8):858-874.

Chabanon A, Desterke C, Rodenburger E, et al. A cross-talk between

stromal cell-derived factor-1 and transforming growth factor-beta controls the quiescence/cycling switch of CD34(+) progenitors through FoxO3 and mammalian target of rapamycin. 18.

Stem Cells

. 2008;26(12):3150-3161.

Garbe A, Spyridonidis A, Mobest D, Schmoor C, Mertelsmann R, Henschler

R. Transforming growth factor-beta 1 delays formation of granulocyte-

20

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

macrophage colony-forming cells, but spares more primitive progenitors during ex vivo expansion of CD34+ haemopoietic progenitor cells.

Br J Haematol

.

1997;99(4):951-958. 19.

Batard P, Monier MN, Fortunel N, et al. TGF-(beta)1 maintains

hematopoietic immaturity by a reversible negative control of cell cycle and induces CD34 antigen up-modulation. 20.

J Cell Sci

. 2000;113 ( Pt 3):383-390.

Sitnicka E, Ruscetti FW, Priestley GV, Wolf NS, Bartelmez SH.

Transforming growth factor beta 1 directly and reversibly inhibits the initial cell divisions of long-term repopulating hematopoietic stem cells.

Blood

.

1996;88(1):82-88. 21.

Fortunel NO, Hatzfeld A, Hatzfeld JA. Transforming growth factor-beta:

pleiotropic role in the regulation of hematopoiesis. 22.

Blood

. 2000;96(6):2022-2036.

Cheshier SH, Morrison SJ, Liao X, Weissman IL. In vivo proliferation and

cell cycle kinetics of long-term self-renewing hematopoietic stem cells.

Acad Sci U S A

Proc Natl

. 1999;96(6):3120-3125.

23.

Wilson A, Laurenti E, Oser G, et al. Hematopoietic stem cells reversibly

switch from dormancy to self-renewal during homeostasis and repair.

Cell

.

2008;135(6):1118-1129. 24.

Passegue E, Wagers AJ, Giuriato S, Anderson WC, Weissman IL. Global

analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates.

medicine

The Journal of experimental

. 2005;202(11):1599-1611.

25.

Soma T, Yu JM, Dunbar CE. Maintenance of murine long-term

repopulating stem cells in ex vivo culture is affected by modulation of transforming growth factor-beta but not macrophage inflammatory protein-1 alpha activities. 26.

Blood

. 1996;87(11):4561-4567.

Fortunel N, Batard P, Hatzfeld A, et al. High proliferative potential-

quiescent cells: a working model to study primitive quiescent hematopoietic cells. 27.

J Cell Sci

. 1998;111 ( Pt 13):1867-1875.

Hatzfeld J, Li ML, Brown EL, et al. Release of early human hematopoietic

progenitors from quiescence by antisense transforming growth factor beta 1 or Rb oligonucleotides. 28.

J Exp Med

. 1991;174(4):925-929.

Dao MA, Hwa J, Nolta JA. Molecular mechanism of transforming growth

factor beta-mediated cell-cycle modulation in primary human CD34(+) progenitors. 29.

Blood

. 2002;99(2):499-506.

Dao MA, Taylor N, Nolta JA. Reduction in levels of the cyclin-dependent

kinase inhibitor p27(kip-1) coupled with transforming growth factor beta neutralization induces cell-cycle entry and increases retroviral transduction of primitive human hematopoietic cells.

Proc Natl Acad Sci U S A

.

1998;95(22):13006-13011. 30.

Dubois CM, Ruscetti FW, Palaszynski EW, Falk LA, Oppenheim JJ, Keller JR.

Transforming growth factor beta is a potent inhibitor of interleukin 1 (IL-1) receptor expression: proposed mechanism of inhibition of IL-1 action.

J Exp Med

.

1990;172(3):737-744. 31.

Dubois CM, Ruscetti FW, Stankova J, Keller JR. Transforming growth

factor-beta regulates c-kit message stability and cell-surface protein expression in hematopoietic progenitors. 32.

Blood

. 1994;83(11):3138-3145.

Ducos K, Panterne B, Fortunel N, Hatzfeld A, Monier MN, Hatzfeld J.

p21(cip1) mRNA is controlled by endogenous transforming growth factor-beta1

21

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

in quiescent human hematopoietic stem/progenitor cells.

J Cell Physiol

.

2000;184(1):80-85. 33.

Jacobsen SE, Ruscetti FW, Dubois CM, Lee J, Boone TC, Keller JR.

Transforming growth factor-beta trans-modulates the expression of colony stimulating factor receptors on murine hematopoietic progenitor cell lines.

Blood

.

1991;77(8):1706-1716. 34.

Sansilvestri P, Cardoso AA, Batard P, et al. Early CD34high cells can be

separated into KIThigh cells in which transforming growth factor-beta (TGFbeta) downmodulates c-kit and KITlow cells in which anti-TGF-beta upmodulates c-kit. 35.

Blood

. 1995;86(5):1729-1735.

Cheng T, Shen H, Rodrigues N, Stier S, Scadden DT. Transforming growth

factor beta 1 mediates cell-cycle arrest of primitive hematopoietic cells independent of p21(Cip1/Waf1) or p27(Kip1). 36.

Blood

. 2001;98(13):3643-3649.

Scandura JM, Boccuni P, Massague J, Nimer SD. Transforming growth

factor beta-induced cell cycle arrest of human hematopoietic cells requires p57KIP2 up-regulation. 37.

Proc Natl Acad Sci U S A

. 2004;101(42):15231-15236.

Yamazaki S, Iwama A, Takayanagi S, et al. Cytokine signals modulated via

lipid rafts mimic niche signals and induce hibernation in hematopoietic stem cells.

Embo J

38.

. 2006;25(15):3515-3523.

Yamazaki S, Iwama A, Takayanagi S, Eto K, Ema H, Nakauchi H. TGF-beta

as a candidate bone marrow niche signal to induce hematopoietic stem cell hibernation. 39.

Blood

. 2009;113(6):1250-1256.

Dykstra B, Kent D, Bowie M, et al. Long-term propagation of distinct

hematopoietic differentiation programs in vivo.

Cell Stem Cell

. 2007;1(2):218-

229. 40.

Sieburg HB, Cho RH, Dykstra B, Uchida N, Eaves CJ, Muller-Sieburg CE. The

hematopoietic stem compartment consists of a limited number of discrete stem cell subsets. 41.

Blood

. 2006;107(6):2311-2316.

Copley MR, Beer PA, Eaves CJ. Hematopoietic stem cell heterogeneity

Cell Stem Cell

takes center stage. 42.

. 2012;10(6):690-697.

Challen GA, Boles NC, Chambers SM, Goodell MA. Distinct hematopoietic

stem cell subtypes are differentially regulated by TGF-beta1.

Cell Stem Cell

.

2010;6(3):265-278. 43.

Park SM, Deering RP, Lu Y, et al. Musashi-2 controls cell fate, lineage bias,

and TGF-beta signaling in HSCs.

The Journal of experimental medicine

.

2014;211(1):71-87. 44.

Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane

to the nucleus. 45.

Cell

. 2003;113(6):685-700.

Piek E, Heldin CH, Ten Dijke P. Specificity, diversity, and regulation in

TGF-beta superfamily signaling. 46.

FASEB J

. 1999;13(15):2105-2124.

Langer JC, Henckaerts E, Orenstein J, Snoeck HW. Quantitative trait

analysis reveals transforming growth factor-beta2 as a positive regulator of early hematopoietic progenitor and stem cell function. 47.

J Exp Med

. 2004;199(1):5-14.

Quere R, Saint-Paul L, Carmignac V, et al. Tif1gamma regulates the TGF-

beta1 receptor and promotes physiological aging of hematopoietic stem cells.

Proceedings of the National Academy of Sciences of the United States of America

.

2014;111(29):10592-10597. 48. aging.

Beerman I, Rossi DJ. Epigenetic regulation of hematopoietic stem cell

Exp Cell Res

. 2014;329(2):192-199.

22

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

49.

Blaney Davidson EN, Scharstuhl A, Vitters EL, van der Kraan PM, van den

Berg WB. Reduced transforming growth factor-beta signaling in cartilage of old mice: role in impaired repair capacity. 50.

Arthritis Res Ther

. 2005;7(6):R1338-1347.

Loffredo FS, Steinhauser ML, Jay SM, et al. Growth differentiation factor

11 is a circulating factor that reverses age-related cardiac hypertrophy.

Cell

.

2013;153(4):828-839. 51.

Sun D, Luo M, Jeong M, et al. Epigenomic profiling of young and aged HSCs

reveals concerted changes during aging that reinforce self-renewal.

Cell Stem Cell

.

2014;14(5):673-688. 52.

Henckaerts E, Langer JC, Orenstein J, Snoeck HW. The positive regulatory

effect of TGF-beta2 on primitive murine hemopoietic stem and progenitor cells is dependent on age, genetic background, and serum factors.

J Immunol

.

2004;173(4):2486-2493. 53.

Schofield R. The relationship between the spleen colony-forming cell and

the haemopoietic stem cell.

Blood Cells

. 1978;4(1-2):7-25.

54.

Lo Celso C, Scadden DT. The haematopoietic stem cell niche at a glance.

55.

Wang LD, Wagers AJ. Dynamic niches in the origination and

Journal of cell science

. 2011;124(Pt 21):3529-3535.

differentiation of haematopoietic stem cells.

biology

Nature reviews Molecular cell

. 2011;12(10):643-655.

56.

Pfeilschifter J, Diel I, Scheppach B, et al. Concentration of transforming

growth factor beta in human bone tissue: relationship to age, menopause, bone

Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research turnover, and bone volume.

. 1998;13(4):716-

730. 57.

Utsugisawa T, Moody JL, Aspling M, Nilsson E, Carlsson L, Karlsson S. A

road map toward defining the role of Smad signaling in hematopoietic stem cells.

Stem Cells

. 2006;24(4):1128-1136.

58.

Yamazaki S, Ema H, Karlsson G, et al. Nonmyelinating Schwann cells

maintain hematopoietic stem cell hibernation in the bone marrow niche.

Cell

.

2011;147(5):1146-1158. 59.

Bruns I, Lucas D, Pinho S, et al. Megakaryocytes regulate hematopoietic

stem cell quiescence through CXCL4 secretion.

Nature medicine

.

2014;20(11):1315-1320. 60.

Zhao M, Perry JM, Marshall H, et al. Megakaryocytes maintain homeostatic

quiescence and promote post-injury regeneration of hematopoietic stem cells.

Nature medicine

. 2014;20(11):1321-1326.

61.

Soderberg SS, Karlsson G, Karlsson S. Complex and context dependent

regulation of hematopoiesis by TGF-beta superfamily signaling.

Ann N Y Acad Sci

.

2009;1176:55-69. 62.

Larsson J, Karlsson S. The role of Smad signaling in hematopoiesis.

63.

Ruscetti FW, Akel S, Bartelmez SH. Autocrine transforming growth factor-

Oncogene

. 2005;24(37):5676-5692.

beta regulation of hematopoiesis: many outcomes that depend on the context.

Oncogene

. 2005;24(37):5751-5763.

64.

Yaswen L, Kulkarni AB, Fredrickson T, et al. Autoimmune manifestations

in the transforming growth factor-beta 1 knockout mouse. 1996;87(4):1439-1445.

23

Blood

.

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

65.

Letterio JJ, Geiser AG, Kulkarni AB, et al. Autoimmunity associated with

TGF-beta1-deficiency in mice is dependent on MHC class II antigen expression.

Clin Invest

J

. 1996;98(9):2109-2119.

66.

Leveen P, Larsson J, Ehinger M, et al. Induced disruption of the

transforming growth factor beta type II receptor gene in mice causes a lethal inflammatory disorder that is transplantable. 67.

Blood

. 2002;100(2):560-568.

Capron C, Lacout C, Lecluse Y, et al. A major role of TGF-{beta}1 in the

homing capacities of murine hematopoietic stem cell/ progenitors. 68.

Blood

. 2010.

Larsson J, Blank U, Helgadottir H, et al. TGF-beta signaling-deficient

hematopoietic stem cells have normal self-renewal and regenerative ability in vivo despite increased proliferative capacity in vitro.

Blood

. 2003;102(9):3129-

3135. 69.

Larsson J, Blank U, Klintman J, Magnusson M, Karlsson S. Quiescence of

hematopoietic stem cells and maintenance of the stem cell pool is not dependent on TGF-beta signaling in vivo. 70.

Exp Hematol

. 2005;33(5):592-596.

Ozdamar B, Bose R, Barrios-Rodiles M, Wang HR, Zhang Y, Wrana JL.

Regulation of the polarity protein Par6 by TGFbeta receptors controls epithelial cell plasticity. 71.

Science

. 2005;307(5715):1603-1609.

Viloria-Petit AM, David L, Jia JY, et al. A role for the TGFbeta-Par6 polarity

Proceedings of the National Academy of Sciences of the United States of America The Journal of experimental medicine

pathway in breast cancer progression.

. 2009;106(33):14028-14033.

72.

Brenet F, Kermani P, Spektor R, Rafii S, Scandura JM. TGFbeta restores

hematopoietic homeostasis after myelosuppressive chemotherapy. . 2013;210(3):623-639.

73.

Rorby E, Hagerstrom MN, Blank U, Karlsson G, Karlsson S. Human

hematopoietic stem/progenitor cells overexpressing Smad4 exhibit impaired reconstitution potential in vivo. 74.

hematopoietic stem cells. 75.

Blood

. 2012;120(22):4343-4351.

Blank U, Karlsson G, Moody JL, et al. Smad7 promotes self-renewal of

Blood

. 2006;108(13):4246-4254.

Chadwick K, Shojaei F, Gallacher L, Bhatia M. Smad7 alters cell fate

decisions of human hematopoietic repopulating cells.

Blood

. 2005;105(5):1905-

1915. 76.

Karlsson G, Blank U, Moody JL, et al. Smad4 is critical for self-renewal of

hematopoietic stem cells. 77.

J Exp Med

. 2007;204(3):467-474.

Ransom DG, Bahary N, Niss K, et al. The zebrafish moonshine gene

encodes transcriptional intermediary factor 1gamma, an essential regulator of hematopoiesis. 78.

PLoS Biol

. 2004;2(8):E237.

Bai X, Kim J, Yang Z, et al. TIF1gamma controls erythroid cell fate by

regulating transcription elongation. 79.

Cell

. 2010;142(1):133-143.

Monteiro R, Pouget C, Patient R. The gata1/pu.1 lineage fate paradigm

varies between blood populations and is modulated by tif1gamma.

Embo J

.

2011;30(6):1093-1103. 80.

Xi Q, Wang Z, Zaromytidou AI, et al. A poised chromatin platform for TGF-

beta access to master regulators. 81.

Cell

. 2011;147(7):1511-1524.

Grady WM, Myeroff LL, Swinler SE, et al. Mutational inactivation of

transforming growth factor beta receptor type II in microsatellite stable colon cancers.

Cancer Res

. 1999;59(2):320-324.

24

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

82.

Villanueva A, Garcia C, Paules AB, et al. Disruption of the antiproliferative

TGF-beta signaling pathways in human pancreatic cancer cells.

Oncogene

.

1998;17(15):1969-1978. 83.

Kim SJ, Letterio J. Transforming growth factor-beta signaling in normal

and malignant hematopoiesis. 84.

Leukemia

. 2003;17(9):1731-1737.

Imai Y, Kurokawa M, Izutsu K, et al. Mutations of the Smad4 gene in acute

myelogeneous leukemia and their functional implications in leukemogenesis.

Oncogene

. 2001;20(1):88-96.

85.

Yang L, Wang N, Tang Y, Cao X, Wan M. Acute myelogenous leukemia-

derived SMAD4 mutations target the protein to ubiquitin-proteasome degradation. 86.

Hum Mutat

. 2006;27(9):897-905.

Molenaar JJ, Gerard B, Chambon-Pautas C, et al. Microsatellite instability

and frameshift mutations in BAX and transforming growth factor-beta RII genes are very uncommon in acute lymphoblastic leukemia in vivo but not in cell lines.

Blood

. 1998;92(1):230-233.

87.

Scott S, Kimura T, Ichinohasama R, et al. Microsatellite mutations of

transforming growth factor-beta receptor type II and caspase-5 occur in human precursor T-cell lymphoblastic lymphomas/leukemias in vivo but are not

Leuk Res

associated with hMSH2 or hMLH1 promoter methylation.

.

2003;27(1):23-34. 88.

Walter MJ, Payton JE, Ries RE, et al. Acquired copy number alterations in

adult acute myeloid leukemia genomes.

Sciences of the United States of America

Proceedings of the National Academy of

. 2009;106(31):12950-12955.

89.

Knaus PI, Lindemann D, DeCoteau JF, et al. A dominant inhibitory mutant

of the type II transforming growth factor beta receptor in the malignant progression of a cutaneous T-cell lymphoma.

Mol Cell Biol

. 1996;16(7):3480-

3489. 90.

Schiemann WP, Pfeifer WM, Levi E, Kadin ME, Lodish HF. A deletion in the

gene for transforming growth factor beta type I receptor abolishes growth regulation by transforming growth factor beta in a cutaneous T-cell lymphoma.

Blood

. 1999;94(8):2854-2861.

91.

Wolfraim LA, Fernandez TM, Mamura M, et al. Loss of Smad3 in acute T-

cell lymphoblastic leukemia. 92.

N Engl J Med Neoplasia

acute lymphoblastic leukemia. 93.

. 2004;351(6):552-559.

Komuro H, Valentine MB, Rubnitz JE, et al. p27KIP1 deletions in childhood . 1999;1(3):253-261.

Takeuchi C, Takeuchi S, Ikezoe T, Bartram CR, Taguchi H, Koeffler HP.

Germline mutation of the p27/Kip1 gene in childhood acute lymphoblastic leukemia. 94.

Leukemia

. 2002;16(5):956-958.

Jakubowiak A, Pouponnot C, Berguido F, et al. Inhibition of the

transforming growth factor beta 1 signaling pathway by the AML1/ETO leukemia-associated fusion protein. 95.

J Biol Chem

. 2000;275(51):40282-40287.

Kurokawa M, Mitani K, Imai Y, Ogawa S, Yazaki Y, Hirai H. The t(3;21)

fusion product, AML1/Evi-1, interacts with Smad3 and blocks transforming growth factor-beta-mediated growth inhibition of myeloid cells.

Blood

.

1998;92(11):4003-4012. 96.

Lee DK, Kim BC, Brady JN, Jeang KT, Kim SJ. Human T-cell lymphotropic

virus type 1 tax inhibits transforming growth factor-beta signaling by blocking the association of Smad proteins with Smad-binding element. 2002;277(37):33766-33775.

25

J Biol Chem

.

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

97.

Nakahata S, Yamazaki S, Nakauchi H, Morishita K. Downregulation of

ZEB1 and overexpression of Smad7 contribute to resistance to TGF-beta1mediated growth suppression in adult T-cell leukemia/lymphoma.

Oncogene

.

2010;29(29):4157-4169. 98.

Mitani K. Molecular mechanisms of leukemogenesis by AML1/EVI-1.

99.

Ford AM, Palmi C, Bueno C, et al. The TEL-AML1 leukemia fusion gene

Oncogene

. 2004;23(24):4263-4269.

dysregulates the TGF-beta pathway in early B lineage progenitor cells.

Invest

J Clin

. 2009;119(4):826-836.

100.

Sood R, Talwar-Trikha A, Chakrabarti SR, Nucifora G. MDS1/EVI1

enhances TGF-beta1 signaling and strengthens its growth-inhibitory effect but the leukemia-associated fusion protein AML1/MDS1/EVI1, product of the t(3;21), abrogates growth-inhibition in response to TGF-beta1.

Leukemia

.

1999;13(3):348-357. 101.

Wang N, Kim HG, Cotta CV, et al. TGFbeta/BMP inhibits the bone marrow

transformation capability of Hoxa9 by repressing its DNA-binding ability.

Embo J

.

2006;25(7):1469-1480. 102.

Quere R, Karlsson G, Hertwig F, et al. SMAD4 Sequestrates HOXA9 to

Protect Hematopoietic Stem Cells Against Leukemia Transformation.

Annual Meeting Abstracts)

Blood (ASH

. 2010;116:3153.

103.

Schepers K, Pietras EM, Reynaud D, et al. Myeloproliferative neoplasia

remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. 104.

Cell Stem Cell

. 2013;13(3):285-299.

Raaijmakers MH, Mukherjee S, Guo S, et al. Bone progenitor dysfunction

induces myelodysplasia and secondary leukaemia.

Nature

. 2010;464(7290):852-

857. 105.

Walkley CR, Olsen GH, Dworkin S, et al. A microenvironment-induced

myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency.

Cell

. 2007;129(6):1097-1110.

106.

Krause DS, Fulzele K, Catic A, et al. Differential regulation of myeloid

leukemias by the bone marrow microenvironment.

Nature medicine

.

2013;19(11):1513-1517. 107.

Naka K, Hoshii T, Muraguchi T, et al. TGF-beta-FOXO signalling maintains

leukaemia-initiating cells in chronic myeloid leukaemia.

Nature

.

2010;463(7281):676-680. 108.

Dong M, Blobe GC. Role of transforming growth factor-beta in hematologic

malignancies. 109.

Blood

. 2006;107(12):4589-4596.

Shehata M, Schwarzmeier JD, Hilgarth M, Hubmann R, Duechler M,

Gisslinger H. TGF-beta1 induces bone marrow reticulin fibrosis in hairy cell leukemia. 110.

J Clin Invest

. 2004;113(5):676-685.

Le Bousse-Kerdiles MC, Chevillard S, Charpentier A, et al. Differential

expression of transforming growth factor-beta, basic fibroblast growth factor, and their receptors in CD34+ hematopoietic progenitor cells from patients with myelofibrosis and myeloid metaplasia. 111.

Blood

. 1996;88(12):4534-4546.

Chagraoui H, Komura E, Tulliez M, Giraudier S, Vainchenker W, Wendling

F. Prominent role of TGF-beta 1 in thrombopoietin-induced myelofibrosis in mice.

Blood

. 2002;100(10):3495-3503.

26

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

112.

Vannucchi AM, Bianchi L, Paoletti F, et al. A pathobiologic pathway linking

thrombopoietin, GATA-1, and TGF-beta1 in the development of myelofibrosis.

Blood

. 2005;105(9):3493-3501.

113.

Bachegowda L, Gligich O, Mantzaris I, et al. Signal transduction inhibitors

J Hematol Oncol Blood

in treatment of myelodysplastic syndromes. 114.

Zhou L, Nguyen AN, Sohal D, et al. Inhibition of the TGF-beta receptor I

kinase promotes hematopoiesis in MDS. 115.

. 2013;6:50.

. 2008;112(8):3434-3443.

Zhou L, McMahon C, Bhagat T, et al. Reduced SMAD7 leads to

overactivation of TGF-beta signaling in MDS that can be reversed by a specific inhibitor of TGF-beta receptor I kinase. 116.

Cancer research Blood

. 2011;71(3):955-963.

Bhagat TD, Zhou L, Sokol L, et al. miR-21 mediates hematopoietic

suppression in MDS by activating TGF-beta signaling.

. 2013;121(15):2875-

2881. 117.

Bhatia M, Bonnet D, Wu D, et al. Bone morphogenetic proteins regulate

J Exp Med

the developmental program of human hematopoietic stem cells.

.

1999;189(7):1139-1148. 118.

Suragani RN, Cadena SM, Cawley SM, et al. Transforming growth factor-

beta superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. 119.

Nature medicine

. 2014;20(4):408-414.

Dussiot M, Maciel TT, Fricot A, et al. An activin receptor IIA ligand trap

corrects ineffective erythropoiesis in beta-thalassemia.

Nature medicine

.

2014;20(4):398-407. 120.

Kretzschmar M, Doody J, Massague J. Opposing BMP and EGF signalling

pathways converge on the TGF-beta family mediator Smad1.

Nature

.

1997;389(6651):618-622. 121.

Kretzschmar M, Doody J, Timokhina I, Massague J. A mechanism of

repression of TGFbeta/ Smad signaling by oncogenic Ras.

Genes Dev

.

1999;13(7):804-816. 122.

Fuentealba LC, Eivers E, Ikeda A, et al. Integrating patterning signals:

Wnt/GSK3 regulates the duration of the BMP/Smad1 signal.

Cell

.

2007;131(5):980-993. 123.

Pera EM, Ikeda A, Eivers E, De Robertis EM. Integration of IGF, FGF, and

anti-BMP signals via Smad1 phosphorylation in neural induction. 2003;17(24):3023-3028.

27

Genes Dev

.

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

Figure legends Figure 1. The hematopoietic hierarchy

. Hematopoiesis is organized in a

hierarchical manner, with rare HSCs at the top that give rise to various types of

progenitor cells, which proliferate extensively, finally generating mature blood

cells at the bottom of the hierarchy. Long-term HSC (LT-HSC); short-term HSC

(ST-HSC); multipotent progenitor (MPP); common myeloid progenitor (CMP);

common lymphoid progenitor (CLP); granulocyte-macrophage-lymphocyte

progenitor (GMLP); granulocyte-macrophage progenitor (GMP); megakaryocyte-

erythrocyte progenitor (MEP); natural killer (NK). Red arrows indicate

stimulation of proliferation by TGF-β, whereas inhibition signs point to TGF-β’s

growth inhibitory effect, in specific cell types or lineages.

Figure 2. TGF-β signaling pathways.

β ligands bind type I and type II

TGF-

receptors at the cell surface. Subsequently, the type I receptor (ALK5) becomes

phosphorylated by the type II receptor. This leads to phosphorylation of SMAD2

and SMAD3, which form a complex with SMAD4. Activated complexes

accumulate in the nucleus where they cooperate with DNA-binding cofactors to

regulate target gene transcription. SMAD2 and SMAD3 also bind to TIF1γ. In ES

cells, SMAD2/3-TIF1γ recognizes specific chromatin marks, promoting access of

SMAD2/3-SMAD4 to otherwise repressed targets. TIF1-γ-SMAD2/3 promotes

erythroid differentiation whereas SMAD4-SMAD2/SMAD3 complexes inhibit

proliferation. In certain cell types Jun N-terminal kinase (JNK) and p38 are

phosphorylated by TAK1 and constitute, together with the phosphatidyl inositol

3-kinase (PI3K)-AKT-FOXO axis, extracellular signal-regulated kinase (ERK), and

β. Dashed line

PAR6, so-called non-canonical signaling responses to TGF-

indicates unclear molecular mechanism.

28

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

Figure 3. Bidirectional effects of TGF-β.

Low concentrations of TGF-β

stimulate proliferation of My-HSCs, whereas Ly-HSCs are growth inhibited. A

higher dose of TGF-β inhibits proliferation, irrespective of HSC subtype, and

Kip2.

induces quiescence via SMAD2/3-SMAD4-dependent expression of p57

Figure 4. TGF-β in the bone marrow niche.

HSCs reside in the bone marrow, in

β is activated

specialized niches that support and regulate HSC fate options. TGF-

β are

by Schwann cells that ensheath sympathetic nerves. Large amounts of TGF-

also produced by megakaryocytes. HSCs in close proximity to Schwann cells and

β and exhibit activation of SMAD2/3.

megakaryocytes become exposed to TGF-

29

Self-renewal

Figure 1

LT-HSC Self-renewal

Symmetric

ST-HSC Asymmetric

TGFß

MPP

Differentiation: Lineage specification

CMP

MEP

GMLP

GMP

CLP

Granulocyte

Erythrocyte Platelets

Dendritic cell Monocyte

NK cell

T lymphocyte B lymphocyte

Figure 2

TGFß

Cell surface

TßRII ALK5

P

RHO

PAR6

P

P

P

PI3K

P

SMAD2/3

SMAD7

Cytoplasm

ERK

TAK1 P

AKT TIF1γ

P

SMAD4

JNK

P

p38

P FOXO

Erythroid differention

Co-factor

SMAD4

SMAD2/3

TIF1γ

Nucleus

Growth inhibition

Figure 3 My-HSC

TGFβ concentration Low

High

Ly-HSC

HSC

SMAD4

Co-factor

SMAD2/3

Nucleus

p57Kip2

Quiescence

Figure 4

Blood vessel

Sympathetic nerve

Schwann cell

TGFβ HSC

TGFβ

Bone

ce

s

Osteoblast

Q uie

Megakaryocyte

nce

Support cell

From www.bloodjournal.org by guest on May 11, 2015. For personal use only.

Prepublished online April 1, 2015; doi:10.1182/blood-2014-12-618090

TGF-β signaling in the control of hematopoietic stem cells Ulrika Blank and Stefan Karlsson

Information about reproducing this article in parts or in its entirety may be found online at: http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests Information about ordering reprints may be found online at: http://www.bloodjournal.org/site/misc/rights.xhtml#reprints Information about subscriptions and ASH membership may be found online at: http://www.bloodjournal.org/site/subscriptions/index.xhtml

Advance online articles have been peer reviewed and accepted for publication but have not yet appeared in the paper journal (edited, typeset versions may be posted when available prior to final publication). Advance online articles are citable and establish publication priority; they are indexed by PubMed from initial publication. Citations to Advance online articles must include digital object identifier (DOIs) and date of initial publication.

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036. Copyright 2011 by The American Society of Hematology; all rights reserved.

TGF-β signaling in the control of hematopoietic stem cells.

Blood is a tissue with high cellular turnover, and its production is a tightly orchestrated process that requires constant replenishment. All mature b...
313KB Sizes 0 Downloads 9 Views