Haematopoietic stem and progenitor cells (HSPCs) sustain haematopoiesis by generating precise numbers of mature blood cells throughout the lifetime of an individual. In vertebrates, HSPCs arise during embryonic development from a specialised endothelial cell population, the haemogenic endothelium (HE). Signalling by the Transforming Growth Factor β (TGFβ) pathway is key to regulate haematopoiesis in the adult bone marrow, but evidence for a role in the formation of HSPCs has only recently started to emerge. In this review, we examine recent work in various model systems that demonstrate a key role for TGFβ signalling in HSPC emergence from the HE. The current evidence underpins two seemingly contradictory views of TGFβ function: as a negative regulator of HSPCs by limiting haematopoietic output from HE, and as a positive regulator, by programming the HE towards the haematopoietic fate. Understanding how to modulate the requirement for TGFβ signalling in HSC emergence may have critical implications for the generation of these cells in vitro for therapeutic use.

TGFβ is a well-known regulator of adult haematopoietic stem cell (HSC) lineage determination, self-renewal and differentiation in development and in disease [1–4]. Its functions in adult haematopoiesis have been discussed in many excellent reviews [4–6] and will not be covered here. Here we will focus on the emerging role of TGFβ signalling in the formation of nascent HSPCs during embryonic development, the current views regarding TGFβ signal transduction and transcriptional responses in this context, and future research that can contribute to resolve the existing concerns and contradictions.

The fundamentals of TGFβ signalling — canonical and non-canonical signalling

TGFβ proteins are part of a large family of secreted growth factors with pleiotropic activities in diverse tissues ranging from proliferation, cell death and differentiation to lineage determination, organ morphogenesis and tissue homeostasis [5,7,8]. The family comprises over 30 members, including TGFβ1–3, Activins, Bone Morphogenetic Proteins (BMPs) and Growth and Differentiation Factors (GDFs) [9,10]. The fundamental components of this pathway have been elucidated over the years by many research groups and have been excellently reviewed by many colleagues. Briefly, in canonical signalling, TGFβ secreted factors bind to heteromeric tyrosine kinase type I and type II receptor complexes at the cell surface. Upon ligand binding, type II receptors phosphorylate type I receptors, which in turn phosphorylate intracellular mediators called Receptor-regulated Smad proteins (R-Smads). Subsequently, R-Smads then form a complex with the common Smad, Smad4, and translocate to the nucleus and bind to specific promoters and enhancers to regulate gene expression [9]. There are seven type I receptors (ALK1–ALK7) and five type II receptors (TGFβRII, BMPRII, ActRII, ActRIIB and AMHRII) and different TGFβ ligands bind different combinations of type I and type II receptors [11]. TGFβ, Nodal and Activin generally induce phosphorylation of Smad2 and Smad3 via ALK4 or ALK5, whereas BMPs phosphorylate Smad1, Smad5 or Smad8 through ALK2, ALK3 or ALK6 [7] (Figure 1). In endothelial cells, however, TGFβ can bind receptor complexes containing either ALK5 (TβRI) or ALK1 (ACVRL1) activating Smad2 and Smad3 via ALK5, but also Smad1 and Smad5 via ALK1 [12–14]. In addition to the canonical Smad-mediated pathways, there are several non-smad mediators of TGFβ signalling, including the c-Jun amino terminal kinase (JNK), p38 MAPK, Akt and others, collectively known as non-canonical signalling mediators and are reviewed elsewhere [6,15].

Schematic representation of TGFβ signalling.

Figure 1.
Schematic representation of TGFβ signalling.

The TGFβ ligands bind and activate type II receptors that phosphorylate type I receptors (ALK1–7). The ALKs in turn phosphorylate intracellular mediators (ALKs). Alk1/4/5 phosphorylate SMAD2/3, whereas Alk1/2/3/6 can phosphorylate SMAD1/4/8 in endothelial cells. The compound SB431542 can inhibit ALK5. The phosphorylated SMAD form a complex with the common SMAD4 to regulate gene transcription. Alternatively, SMAD2/3 can form a complex with YAP1 instead of SMAD4 to act as a chromatin pioneering factor. SMAD1/5/8 recruits HDAC proteins to the promoter erk1/2 gene. NOGGIN, BMPER and GREMLIN1a expression show an inverse correlation to pSMAD1/5/8 staining.

Figure 1.
Schematic representation of TGFβ signalling.

The TGFβ ligands bind and activate type II receptors that phosphorylate type I receptors (ALK1–7). The ALKs in turn phosphorylate intracellular mediators (ALKs). Alk1/4/5 phosphorylate SMAD2/3, whereas Alk1/2/3/6 can phosphorylate SMAD1/4/8 in endothelial cells. The compound SB431542 can inhibit ALK5. The phosphorylated SMAD form a complex with the common SMAD4 to regulate gene transcription. Alternatively, SMAD2/3 can form a complex with YAP1 instead of SMAD4 to act as a chromatin pioneering factor. SMAD1/5/8 recruits HDAC proteins to the promoter erk1/2 gene. NOGGIN, BMPER and GREMLIN1a expression show an inverse correlation to pSMAD1/5/8 staining.

Close modal

HSPCs arise from haemogenic endothelium during embryonic development

HSPCs sustain the blood system throughout life. Although they reside in the bone marrow of the adult, their origin can be traced to a region where the aorta, gonads and the mesonephros (AGM) meet in vertebrate embryos. Elegant time lapse imaging, transplantation assays, immuno-histochemistry for HSPC-associated markers, gene expression, and more recently single cell transcriptomics have convincingly demonstrated the existence of specialised endothelial cells with haematopoietic stem and progenitor cell (HSPC) potential, termed hemogenic endothelium (HE), that undergo a endothelial to haematopoietic transition (EHT) from the lining of the dorsal aorta and thereby progressively lose their endothelial identity and become haematopoietic [16–22]. These cells form intra-aortic haematopoietic clusters (IAHC) that appear associated with the ventral wall of the dorsal aorta in murine embryos starting between the embryonic days 10.25–12 (E10.25–E12). The emerging HSPCs then migrate to the foetal liver for maturation and expansion [23–27]. The sites of HSPC emergence and their migration between haematopoietic niches are very well conserved in vertebrates [28]. Based on transplantation assays performed at different time points within the window of HSPC emergence, early pre-HSC can readily contribute to the blood system of neonates, but not adult [29]. This potency is only evident in HSCs that are older than E11.5 and even then, only a very small fraction of these cells are functional HSCs [30–33]. Therefore, although there is consensus regarding the site of HSPC emergence, it is unclear whether HSPCs and HSC share the same HE precursors or if in fact the HE itself is heterogeneous, and which of the molecular pathways are unique to HSC emergence or shared amongst all HSPCs. In this review, we collectively discuss the role of TGFβ superfamily signalling in the emergence of all HSPC within the dorsal aorta, since there are no established makers to distinguish between haematopoietic progenitors and HSCs. In addition, EHT is a continuous process, meaning that the cells in the AGM are at different stages of generating HSPC. Adding to the complexity, EHT occurs at a developmental time when angiogenic processes are still taking place and vascular identity is being established. Therefore, it is highly likely that both these processes share common signalling pathways. In fact, despite some evidence suggesting that these two cell types may be independent from each other [34–36], it is now widely accepted that arterial specification of the aorta is a pre-requisite for HE specification and subsequent EHT of HSPCs [37–40].

Expression of TGFβ signalling pathway components during the formation of HSPCs

The expression of TGFβ superfamily members has been assessed in several in vivo and in vitro model systems for haematopoiesis. The TGFβ receptors TGFβRII, ALK1 and ALK5 are present in the endothelial cells and HE derived from Embryonic stem cells (ES) in vitro [41], zebrafish [42,43], mouse and chick embryos [41,44]. Endoglin, a type III co-receptor for both TGFβ and BMP9/BMP10 [13,45–47], is also expressed in endothelium [12,41,44]. Of the 3 TGFβ ligands, TGFβ1 (including tgfβ1a and tgfβ1b in zebrafish) is the most prominent one with high levels of expression in the mouse, chick, zebrafish and ESC-derived endothelium and HE [41,42,44,48]. Only very low levels of tgfβ2 have been detected in mouse endothelium, and tgfβ3 is hardly detectable in either the mouse or the chick endothelium [41,44]. However, both ligands are highly abundant in the sub-aortic mesenchyme and the notochord in chick [44] and in zebrafish [42]. In zebrafish, tgfβ3 was additionally detected in the endocardium, somites, floor plate and notochord [42].

The role of TGFβ signalling in the formation of HSPCs

Several labs have studied the involvement of TGFβ signalling in HE specification, EHT and IAHC formation and have come to seemingly contradictory conclusions. The signalling triggered by TGFβ ligands has been studied for its functional requirement during EHT in different models. Several studies have identified the expression of SMAD2/3 in the dorsal aorta and HE and provided evidence for activated SMAD2/3 (pSMAD2/3) in mouse HE/IAHC [49]. Both in mouse and chick embryos only a very discrete number of cells show pSMAD2/3 staining [44,49]. It is plausible that the snapshots provided by the IHC experiments do not represent the overall level of active SMAD2/3 induced by TGFβ ligands in HE since the trigger can by limited to a small window of time during HE specification and EHT.

Functional studies with morpholino mediated knock down of TgfβRII, TGFβ1a or TGFβ1b or using genetic tgfb1b mutants during zebrafish haematopoiesis leads to impaired specification of the HE [42,48]. Knockdown of TGFβ3 led to impaired EHT and decreased haematopoietic output from HSPCs [42]. In contrast, pharmacological inhibition of TGFβ signalling with the compound SB431542 increased haematopoietic output in a mouse embryonic stem cell (mESC) haematopoietic differentiation model [41]. This might seem contradictory at first, but a detailed study of the effects of SB431542 on pSMAD2/3 indicated that in cultures exceeding 24 h, the use of the inhibitor increases SMAD2/3 phosphorylation [49]. Indeed, overexpression of constitutively active SMAD2/3 resulted in the same phenotype as adding the inhibitor. It is unclear how the compound can increase the levels of pSMAD2/3 [50]. It's tempting to hypothesise that blocking the ALK5 receptor leads to over-activation of ALK4/7 that in turn phosphorylate SMAD2/3 as a fine balance is maintained between inputs from different ALKs in a cell [51]. A recent study using haematopoietic differentiation of human ES cells did not report an increased haematopoietic output when using the inhibitor in >24 h culture [52]. However, the authors used a lower concentration of the inhibitor compared with others and neither of these studies added the inhibitor at equivalent stages of in vitro differentiation (i.e. Bruveris et al. from the PDGFRα+ (mesodermal) stage, Thambyrajah et al. from the Flk1+ stage (mesodermal/haemangioblast) and finally Vargel et al. from the EHT stage (CD144+/CD41+) [41,49,52]. Beyond the issues with the mode of action of the inhibitor, these differences make direct comparisons to TGFβ function in vivo difficult. Vargel et al. also added the ligand TGFβ2 to Flk1+ cells sorted from embryoid body cultures which resulted in a decrease in VE-cad/CD41+ haematopoietic progenitor cells (HPCs). They therefore postulated that TGFβ signalling blocked the formation of blood cells and thus negatively regulates haematopoiesis [41]. In contrast, using TGFβ1 in a similar ESC differentiation model led to increased number of CD41+/CD117+ HSPCs [48]. Here it is important to note that TGFβ1 is the main ligand in angiogenesis and haematopoiesis [42,53–55]; although TGFβ2 is a key inducer of endothelial to mesenchymal transition (EndoMT) in the heart [56], it is not expressed in the aortic endothelium or HE [41,42,44]. In addition, mouse mutants for Tgfb2 don't show any obvious angiogenic or haematopoietic defects [57]. TGFβ2 knockdown had no effect on HE specification in zebrafish either [42].

Further experimental evidence for a crucial involvement of SMAD2/3 in EHT was reported in two very recent studies. Here, they identify SMAD2/3 interactions with the chromatin as vital for transcription factors to access their target sites during EHT. In fact, in both studies, SMAD2/3 is discovered as a pioneering factor for opening the chromatin for cell fate change, including for the key haematopoietic transcription factor Runx1 [58,59]. Accordingly, HE and IAHC that are positive for pSMAD2/3 have been reported in the mouse AGM [49]. A more complex function for SMAD2/3 in EHT can be anticipated from studies on EndoMT. In EndoMT, the Hippo pathway member YAP1 can compete with Smad4 for complex formation with Smad3 during cardiac cushion formation [60]. The study finds that YAP1/Smad3 complexes have a stronger DNA binding affinity and drive stable transcription of downstream targets [60]. It´s noteworthy that YAP1 induced by shear stress is required for HSPC/HSC maintenance in the zebrafish AGM [61]. It will be interesting to examine if YAP1 interacts with Smad3 to induce and maintain HSPC/HSC fate during AGM haematopoiesis. Finally, several TGFβ regulatory proteins such as Smad6, Smad7, Bmper, Dach1, Crim1 and Ptpn14, are expressed in ES Cell-derived HE and are targets of the histone deacetylases Hdac1 and/or Hdac2 [49]. Interestingly, ChIP assays demonstrated that Runx1 binds to the Smad6 -57 enhancer and drives its expression in aortic endothelial cells [62]. Here, we hypothesise that Smad6 expression may be silenced in EHT-primed HE through histone deacetylases Hdac1/Hdac2. Histone silencing complexes can be recruited by transcription factors, including Gfi1 and its homologue Gfi1b [63–65]. Therefore, Runx1-expressing cells that start to express Gfi1/1B would recruit histone deacetylases (or other histone modifiers) to repress the negative regulators of TGFβ and permit TGFβ activity. Since epigenetic modifiers can be recruited by various transcription factors, it is possible that several HE and later HSPC-specific transcription factors fine tune the overall output of TGFβ signalling in each cell, and that the output can be varied depending on the cell type.

Evidence for non-canonical TGFβ signalling in HSPC emergence

Zhang and colleagues showed that Tgfβ1b regulated HSPC emergence by promoting gluconeogenesis, an effect mediated via the non-canonical JNK/c-jun pathway rather than canonical Smad2 phosphorylation [48]. They demonstrated that the expression of zebrafish JNK orthologues tak1, mapk8a, mapk9 and mapk10 was decreased in tgfb1b mutant endothelial cells, suggesting a role for JNK downstream of TGFβ signalling. Accordingly, chemical inhibition of JNK signalling phenocopied the loss of HSPCs found in tgfb1b mutants. They further identified that c-jun expression was decreased in the mutants [48]. Loss of tgfb1b or c-jun led to decreased g6pc3, an enzyme in the gluconeogenesis pathway that was specifically enriched in HSPCs. Addition of 1% glucose or re-expression of g6pc3 rescued expression of HSPC-specific markers runx1 and cmyb [48]. They proposed that a Tgfβ1b/JNK/c-jun/g6pc3 axis is required to maintain sufficient levels of gluconeogenesis to enable HSPC emergence from HE. Together with our previous study in zebrafish [42], these experiments indicate that TGFβ signalling is an inducer of HE and is critical for the formation of HSPCs (Figure 2).

Schematic representation of the stepwise development from mesodermal cells to HSPC/HSCs in the vertebrate embryo and the requirement of TGFβ signalling in these transitions based on current studies.

Figure 2.
Schematic representation of the stepwise development from mesodermal cells to HSPC/HSCs in the vertebrate embryo and the requirement of TGFβ signalling in these transitions based on current studies.

Decreasing BMP4 levels are needed from the mesodermal to a HE stage, and as a gradually decreasing gradient from the sub-aortic mesenchyme toward the ventral wall of the dorsal aorta. Within the ventral wall of the dorsal aorta, HE/HSPC/HSCs cells are activated by TGFβ1/3 and show presence of pSMAD2/3 and low pSMAD1/5/8. BMP4 is antagonised by BMPER, GREMLIN1a and NOGGIN. In our hypothesis, based on the published data, we postulate that SMAD2/3 is needed to open the chromatin for Runx1 to drive EHT, in a process similar to that seen in EndoMT. Please note that the scheme does not include other known regulators of Runx1 expression such as Notch or VegfA signalling.

Figure 2.
Schematic representation of the stepwise development from mesodermal cells to HSPC/HSCs in the vertebrate embryo and the requirement of TGFβ signalling in these transitions based on current studies.

Decreasing BMP4 levels are needed from the mesodermal to a HE stage, and as a gradually decreasing gradient from the sub-aortic mesenchyme toward the ventral wall of the dorsal aorta. Within the ventral wall of the dorsal aorta, HE/HSPC/HSCs cells are activated by TGFβ1/3 and show presence of pSMAD2/3 and low pSMAD1/5/8. BMP4 is antagonised by BMPER, GREMLIN1a and NOGGIN. In our hypothesis, based on the published data, we postulate that SMAD2/3 is needed to open the chromatin for Runx1 to drive EHT, in a process similar to that seen in EndoMT. Please note that the scheme does not include other known regulators of Runx1 expression such as Notch or VegfA signalling.

Close modal

The role of BMP signalling in the formation of HSCs

More research has been conducted to understand the role of BMP-induced signalling through ALK2, ALK3 or ALK6 in the generation of HSPCs. While BMP4 expression surrounds the mesenchyme around the dorsal aorta, BMP activity traced with a Smad1/5-responsive reporter mouse line is detected in cells of the dorsal aorta and HSPCs, with all HSC activity residing in the BMP activated cell AGM population [66]. This stark distinction is lost in the later stages of HSCs maturation where HSC activity resides in both the BMP-activated and non-activated cell fraction, although BMP activated HSCs are more enriched for myeloid biased HSCs [66]. BMP is required to establish the haematopoietic programme in definitive haemangioblasts (the precursors of the dorsal aorta and HE in Xenopus embryos), but not thereafter [67]. These observations tie in with reports that identified expression of the BMP antagonists, Noggin2, gremlin1a and Bmper, at an increasing gradient towards the aorta in the mesenchyme below. At least noggin2 and gremlin1a are induced by FGF signalling from the somites during zebrafish haematopoiesis [68]. Overexpression of noggin from pre-mesoderm stages in Xenopus embryos abrogates HE specification and blood development from the dorsal aorta but has no effect on haematopoiesis if induced after the event [67]. This indicates a time-limited requirement for BMP signalling during HSPC emergence (Figure 2). Similarly, the expression of Bmper increases just after the specification of HE, between E9.5 and E11.5 in a ventrally polarised manner in the AGM. When comparing the localisation of BMPER and BMP-activated cells (indicated by nuclear pSMAD1/5/8 immunostaining) within the AGM region, they showed a negatively correlated distribution, further indicating that Bmper restricts BMP4 activation in emerging IAHC [69]. Nevertheless, Bmper mRNA can be occasionally detected in some intra-aortic cells, including the early cells in the IAHC. Within the IAHC, Noggin expression also shows an inverse correlation with nuclear pSMAD1/5/8 immunostaining [69]. Notably, the TGFβ receptor(s) driving this Smad 1/5/8 phosphorylation in the AGM was not identified in these studies. It remains a possibility that next to BMP4, TGFβ ligands contribute this Smad activation, since they can phosphorylate Smad1 and Smad5 via ALK1 [12–14]. One of the experimentally validated downstream targets of active BMP4 signalling during HE specification and EHT is Erk1/2 signalling [70], a tyrosine kinase receptor activated pathway that controls proliferation and survival [71]. The balance between the Erk1/2/MAPK and the PI3K/AKT pathway determine the arterial versus venous specification [72] in angiogenesis. Zhang et al. [70] revealed that pSMAD1/5 accumulate at the promoter regions of erk1 and erk2 to repress gene expression through recruitment of epigenetic modifiers, including Hdac1. Here again, it is interesting that Erk activity is needed at early stages of HE/EHT. Altogether, these findings suggest an early need for BMP4 signalling prior to HE specification that becomes dispensable thereafter. In fact, pharmacological inhibition of Erk after this stage has the opposite effect, leading to the hypothesis that there is switch from a positive to negative requirement for Erk signalling during embryonic haematopoietic development [70].

This review highlights the pivotal roles played by the TGFβ family of signalling molecules during HE specification, EHT and HSPC/HSC emergence that have been characterised in the recent years. The evidence that has emerged from different model systems conclusively support a dynamic and carefully timed requirement for TGFβ signalling. Most reports clearly support a critical input from pSMAD2/3 and pSMAD1/5/8 for HSPC emergence and there is mounting evidence that non-canonical signalling may also play an important part in this process [48] (Figure 2). Remarkably, a novel interaction between Runx1 and SMAD2/3 has recently been discovered that highlights a key role for TGFβ signalling in promoting EHT by mediating chromatin accessibility of Runx1 target genes [58]. Transcription factor footprinting of haemogenic (E9.5) and non-haemogenic (E13.5) endothelium revealed higher enrichment of Smad2/3 binding motif next to the Runx1 motif at E9.5 [58]. Activation of Runx1 together with TGFβ3 was sufficient to induce HE activity in E13.5 endothelial cells, indicating that these cells progressively lose their plasticity as they mature. Accordingly, adult endothelium stayed refractory to transient induction of combined Runx1 and TGFβ3 expression [58], indicating that other factors might play a role in maintaining that haemogenic plasticity observed at earlier developmental stages. Because recent studies demonstrated the existence of combinatorial SMAD2/3 and Smad1/5-mediated signalling during EMT [14], we speculate that TGFβ-mediated Smad1/5 signalling may also contribute to HE specification and/or EHT. In this regard, in vitro differentiation of embryoid bodies from mouse ES cells towards blood showed increased numbers of CD45+ ALK1+/− cells compared with wild-type [44], suggesting a role for ALK1 in balancing the arterial versus haemogenic cell fate. This observation remains to be confirmed in vivo.

Overall, an abundant number of positive and negative TGFβ family regulators are expressed in the AGM region and therefore, further investigation is required to explain how the entire network is spatially, temporally and functionally coordinated during the stepwise development of HE to EHT and finally to HSPCs/HSCs. Linking data from the growing number of single cell transcriptional profiling studies in HE/HSPCs (e.g. [73–77]) with further studies on the exact composition of SMAD complexes will help address these questions. Chromatin binding data for the individual Smad proteins in HE or HSPC/HSCs would be desirable, but will remain a challenge to perform in low cell number samples until good antibodies and new alternatives to conventional ChIP such as Cut&Tag [78] are standardised. Moreover, Smad proteins only bind to DNA transiently and need transcription factors to stabilise these interactions that additionally increases the difficulty in identifying bona fide binding targets [60,79]. Finally, there is still a need to develop specific compounds to activate or inhibit specific TGFβ receptors and ligands since TGFβ signalling is aberrantly expressed in many diseases [80]. The first studies and clinical trial using the novel TGFβ1/3 inhibitor AVID200 in patients with advanced solid tumours show encouraging results [81,82]. The use of such compounds will help to broaden our understanding of TGFβ signalling in the development of HSPC from HE. In summary, we have identified and discussed mounting evidence for a central role for TGFβ signalling at the earliest stages of HSPC emergence. Which specific Smad complexes direct certain stages of HE specification, EHT and HSPC differentiation and which downstream targets are involved is yet to be fully unravelled.

  • Signalling by the Transforming Growth Factor β family is crucial for the establishment of haematopoietic stem and progenitor cells (HSPCs) from haemogenic endothelium in the developing embryo.

  • While there are controversial observations suggesting opposing roles for TGFβ in the formation of HSPCs, the consensus is emerging that TGFβ signalling is required to programme the embryonic arterial endothelium towards the haematopoietic fate.

  • A better understanding of the activities of TGFβ signalling that enable HSPC emergence will help deliver on the promise of generating these cells in vitro for personalised medicine applications. New cutting-edge technologies such as single cell transcriptomics and epigenomics and better techniques to interrogate transcription factor binding in very low cell numbers will provide a solid platform to achieve that goal.

The authors declare that there are no competing interests associated with the manuscript.

R.T. is a recipient of BP2016(00021) and BP/MSCA 2018(00034) fellowship programs from the Generalitat de Catalunya/Marie Skłodowska-Curie Actions. R.M. is funded by the University of Birmingham and supported by Cancer Research UK [C17422/A25154].

Open access for this article was enabled by the participation of University of Birmingham in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.

R.M. and R.T. conceived the outline, and RM and RT wrote the manuscript. Both authors participated in critical review and editing. Both authors have read and agreed to the published version of the manuscript.

ActRII

Activin receptor type II

AGM

Aorta-Gonad-Mesonephros

AKT

Ak strain transforming

ALK

Activin-like kinase

AMHRII

Anti-Mullerian Hormone receptor type II

BMP

Bone Morphogenetic Protein

BMPRII

BMP receptor type II

ChIP

Chromatin Immunoprecipitation

EHT

Endothelial to Haematopoietic Transition

EMT

Epithelial to Mesenchymal Transition

EndoMT

Endothelial to Mesenchymal Transition (EndoMT)

ERK

Extracellular Signal-Regulated Kinase.

GDF

Growth and Differentiation Factor

HE

Haemogenic endothelium

HSC

Haematopoietic Stem Cell

HSPC

Haematopoietic Stem and Progenitor Cell

IAHC

Intra-aortic Haematopoietic Cluster

JNK

c-Jun amino terminal kinase

MAPK

Mitogen-activated protein kinase

mESC

mouse embryonic stem cell

PI3K

Phosphatidil inositol 3’-kinase

pSMAD1/5

phosphorylated SMAD1/5

pSMAD2/3

phosphorylated SMAD2/3

TGFβ

Transforming Growth Factor β

TGFBRII

TGFβ receptor type II

1
Challen
,
G.A.
,
Boles
,
N.C.
,
Chambers
,
S.M.
and
Goodell
,
M.A.
(
2010
)
Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-beta1
.
Cell Stem Cell
6
,
265
278
2
Wang
,
X.
,
Dong
,
F.
,
Zhang
,
S.
,
Yang
,
W.
,
Yu
,
W.
,
Wang
,
Z.
et al (
2018
)
TGF-beta1 negatively regulates the number and function of hematopoietic stem cells
.
Stem Cell Rep.
11
,
274
287
3
Yamazaki
,
S.
,
Ema
,
H.
,
Karlsson
,
G.
,
Yamaguchi
,
T.
,
Miyoshi
,
H.
,
Shioda
,
S.
et al (
2011
)
Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche
.
Cell
147
,
1146
1158
4
Blank
,
U.
and
Karlsson
,
S.
(
2015
)
TGF-beta signaling in the control of hematopoietic stem cells
.
Blood
125
,
3542
3550
5
Naka
,
K.
and
Hirao
,
A.
(
2017
)
Regulation of hematopoiesis and hematological disease by TGF-beta family signaling molecules
.
Cold Spring Harb. Perspect. Biol.
9
,
a027987
6
Hinge
,
A.
and
Filippi
,
M.D.
(
2016
)
Deconstructing the complexity of TGFbeta signaling in hematopoietic stem cells: quiescence and beyond
.
Curr. Stem Cell Rep.
2
,
388
397
7
Hata
,
A.
and
Chen
,
Y.G.
(
2016
)
TGF-beta signaling from receptors to Smads
.
Cold Spring Harb. Perspect. Biol.
8
,
a022061
8
Weiss
,
A.
and
Attisano
,
L.
(
2013
)
The TGFbeta superfamily signaling pathway
.
Wiley Interdiscip. Rev. Dev. Biol.
2
,
47
63
9
Massague
,
J.
(
1998
)
TGF-beta signal transduction
.
Annu. Rev. Biochem.
67
,
753
791
10
Morikawa
,
M.
,
Derynck
,
R.
and
Miyazono
,
K.
(
2016
)
TGF-beta and the TGF-beta family: context-dependent roles in cell and tissue physiology
.
Cold Spring Harb. Perspect. Biol.
8
,
a021873
11
Heldin
,
C.H.
and
Moustakas
,
A.
(
2016
)
Signaling receptors for TGF-beta family members
.
Cold Spring Harb. Perspect. Biol.
8
,
a022053
12
Oh
,
S.P.
,
Seki
,
T.
,
Goss
,
K.A.
,
Imamura
,
T.
,
Yi
,
Y.
,
Donahoe
,
P.K.
et al (
2000
)
Activin receptor-like kinase 1 modulates transforming growth factor-beta 1 signaling in the regulation of angiogenesis
.
Proc. Natl Acad. Sci. U.S.A.
97
,
2626
2631
13
Goumans
,
M.J.
,
Valdimarsdottir
,
G.
,
Itoh
,
S.
,
Rosendahl
,
A.
,
Sideras
,
P.
and
ten Dijke
,
P.
(
2002
)
Balancing the activation state of the endothelium via two distinct TGF-beta type I receptors
.
EMBO J.
21
,
1743
1753
14
Ramachandran
,
A.
,
Vizan
,
P.
,
Das
,
D.
,
Chakravarty
,
P.
,
Vogt
,
J.
,
Rogers
,
K.W.
et al (
2018
)
TGF-beta uses a novel mode of receptor activation to phosphorylate SMAD1/5 and induce epithelial-to-mesenchymal transition
.
eLife
7
,
e31756
15
Zhang
,
Y.E.
(
2017
)
Non-Smad signaling pathways of the TGF-beta family
.
Cold Spring Harb. Perspect. Biol.
9
,
a022129
16
Kissa
,
K.
and
Herbomel
,
P.
(
2010
)
Blood stem cells emerge from aortic endothelium by a novel type of cell transition
.
Nature
464
,
112
115
17
Bertrand
,
J.Y.
,
Chi
,
N.C.
,
Santoso
,
B.
,
Teng
,
S.
,
Stainier
,
D.Y.
and
Traver
,
D.
(
2010
)
Haematopoietic stem cells derive directly from aortic endothelium during development
.
Nature
464
,
108
111
18
Boisset
,
J.C.
,
van Cappellen
,
W.
,
Andrieu-Soler
,
C.
,
Galjart
,
N.
,
Dzierzak
,
E.
and
Robin
,
C.
(
2010
)
In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium
.
Nature
464
,
116
120
19
Zovein
,
A.C.
,
Hofmann
,
J.J.
,
Lynch
,
M.
,
French
,
W.J.
,
Turlo
,
K.A.
,
Yang
,
Y.
et al (
2008
)
Fate tracing reveals the endothelial origin of hematopoietic stem cells
.
Cell Stem Cell
3
,
625
636
20
Lam
,
E.Y.
,
Hall
,
C.J.
,
Crosier
,
P.S.
,
Crosier
,
K.E.
and
Flores
,
M.V.
(
2010
)
Live imaging of Runx1 expression in the dorsal aorta tracks the emergence of blood progenitors from endothelial cells
.
Blood
116
,
909
914
21
Dzierzak
,
E.
and
Bigas
,
A.
(
2018
)
Blood development: hematopoietic stem cell dependence and independence
.
Cell Stem Cell
22
,
639
651
22
Lancrin
,
C.
,
Sroczynska
,
P.
,
Serrano
,
A.G.
,
Gandillet
,
A.
,
Ferreras
,
C.
,
Kouskoff
,
V.
et al (
2010
)
Blood cell generation from the hemangioblast
.
J. Mol. Med. (Berl)
88
,
167
172
23
Medvinsky
,
A.
and
Dzierzak
,
E.
(
1996
)
Definitive hematopoiesis is autonomously initiated by the AGM region
.
Cell
86
,
897
906
24
Cumano
,
A.
,
Ferraz
,
J.C.
,
Klaine
,
M.
,
Di Santo
,
J.P.
and
Godin
,
I.
(
2001
)
Intraembryonic, but not yolk sac hematopoietic precursors, isolated before circulation, provide long-term multilineage reconstitution
.
Immunity
15
,
477
485
25
North
,
T.E.
,
de Bruijn
,
M.F.
,
Stacy
,
T.
,
Talebian
,
L.
,
Lind
,
E.
,
Robin
,
C.
et al (
2002
)
Runx1 expression marks long-term repopulating hematopoietic stem cells in the midgestation mouse embryo
.
Immunity
16
,
661
672
26
Jaffredo
,
T.
,
Gautier
,
R.
,
Eichmann
,
A.
and
Dieterlen-Lievre
,
F.
(
1998
)
Intraaortic hemopoietic cells are derived from endothelial cells during ontogeny
.
Development
125
,
4575
4583
27
de Bruijn
,
M.F.
,
Speck
,
N.A.
,
Peeters
,
M.C.
and
Dzierzak
,
E.
(
2000
)
Definitive hematopoietic stem cells first develop within the major arterial regions of the mouse embryo
.
EMBO J.
19
,
2465
2474
28
Ciau-Uitz
,
A.
,
Monteiro
,
R.
,
Kirmizitas
,
A.
and
Patient
,
R.
(
2014
)
Developmental hematopoiesis: ontogeny, genetic programming and conservation
.
Exp. Hematol.
42
,
669
683
29
Boisset
,
J.C.
,
Clapes
,
T.
,
Klaus
,
A.
,
Papazian
,
N.
,
Onderwater
,
J.
,
Mommaas-Kienhuis
,
M.
et al (
2015
)
Progressive maturation toward hematopoietic stem cells in the mouse embryo aorta
.
Blood
125
,
465
469
30
Medvinsky
,
A.
,
Rybtsov
,
S.
and
Taoudi
,
S.
(
2011
)
Embryonic origin of the adult hematopoietic system: advances and questions
.
Development
138
,
1017
1031
31
Rybtsov
,
S.
,
Sobiesiak
,
M.
,
Taoudi
,
S.
,
Souilhol
,
C.
,
Senserrich
,
J.
,
Liakhovitskaia
,
A.
et al (
2011
)
Hierarchical organization and early hematopoietic specification of the developing HSC lineage in the AGM region
.
J. Exp. Med.
208
,
1305
1315
32
Taoudi
,
S.
,
Gonneau
,
C.
,
Moore
,
K.
,
Sheridan
,
J.M.
,
Blackburn
,
C.C.
,
Taylor
,
E.
et al (
2008
)
Extensive hematopoietic stem cell generation in the AGM region via maturation of VE-cadherin+ CD45+ pre-definitive HSCs
.
Cell Stem Cell
3
,
99
108
33
Taoudi
,
S.
and
Medvinsky
,
A.
(
2007
)
Functional identification of the hematopoietic stem cell niche in the ventral domain of the embryonic dorsal aorta
.
Proc. Natl Acad. Sci. U.S.A.
104
,
9399
9403
34
Ditadi
,
A.
,
Sturgeon
,
C.M.
and
Keller
,
G.
(
2017
)
A view of human haematopoietic development from the petri dish
.
Nat. Rev. Mol. Cell Biol.
18
,
56
67
35
Ditadi
,
A.
,
Sturgeon
,
C.M.
,
Tober
,
J.
,
Awong
,
G.
,
Kennedy
,
M.
,
Yzaguirre
,
A.D.
et al (
2015
)
Human definitive haemogenic endothelium and arterial vascular endothelium represent distinct lineages
.
Nat. Cell Biol.
17
,
580
591
36
Elcheva
,
I.
,
Brok-Volchanskaya
,
V.
,
Kumar
,
A.
,
Liu
,
P.
,
Lee
,
J.H.
,
Tong
,
L.
et al (
2014
)
Direct induction of haematoendothelial programs in human pluripotent stem cells by transcriptional regulators
.
Nat. Commun.
5
,
4372
37
Bonkhofer
,
F.
,
Rispoli
,
R.
,
Pinheiro
,
P.
,
Krecsmarik
,
M.
,
Schneider-Swales
,
J.
,
Tsang
,
I.H.C.
et al (
2019
)
Blood stem cell-forming haemogenic endothelium in zebrafish derives from arterial endothelium
.
Nat. Commun.
10
,
3577
38
Uenishi
,
G.I.
,
Jung
,
H.S.
,
Kumar
,
A.
,
Park
,
M.A.
,
Hadland
,
B.K.
,
McLeod
,
E.
et al (
2018
)
NOTCH signaling specifies arterial-type definitive hemogenic endothelium from human pluripotent stem cells
.
Nat. Commun.
9
,
1828
39
Chen
,
I.I.
,
Caprioli
,
A.
,
Ohnuki
,
H.
,
Kwak
,
H.
,
Porcher
,
C.
and
Tosato
,
G.
(
2016
)
Ephrinb2 regulates the emergence of a hemogenic endothelium from the aorta
.
Sci. Rep.
6
,
27195
40
Slukvin
,
I.I.
and
Uenishi
,
G.I.
(
2019
)
Arterial identity of hemogenic endothelium: a key to unlock definitive hematopoietic commitment in human pluripotent stem cell cultures
.
Exp. Hematol.
71
,
3
12
41
Vargel
,
O.
,
Zhang
,
Y.
,
Kosim
,
K.
,
Ganter
,
K.
,
Foehr
,
S.
,
Mardenborough
,
Y.
et al (
2016
)
Activation of the TGFbeta pathway impairs endothelial to haematopoietic transition
.
Sci. Rep.
6
,
21518
42
Monteiro
,
R.
,
Pinheiro
,
P.
,
Joseph
,
N.
,
Peterkin
,
T.
,
Koth
,
J.
,
Repapi
,
E.
et al (
2016
)
Transforming growth factor beta drives hemogenic endothelium programming and the transition to hematopoietic stem cells
.
Dev. Cell
38
,
358
370
43
Roman
,
B.L.
,
Pham
,
V.N.
,
Lawson
,
N.D.
,
Kulik
,
M.
,
Childs
,
S.
,
Lekven
,
A.C.
et al (
2002
)
Disruption of acvrl1 increases endothelial cell number in zebrafish cranial vessels
.
Development
129
,
3009
3019
44
Lempereur
,
A.
,
Canto
,
P.Y.
,
Richard
,
C.
,
Martin
,
S.
,
Thalgott
,
J.
,
Raymond
,
K.
et al (
2018
)
The TGFbeta pathway is a key player for the endothelial-to-hematopoietic transition in the embryonic aorta
.
Dev. Biol.
434
,
292
303
45
Lebrin
,
F.
,
Goumans
,
M.J.
,
Jonker
,
L.
,
Carvalho
,
R.L.
,
Valdimarsdottir
,
G.
,
Thorikay
,
M.
et al (
2004
)
Endoglin promotes endothelial cell proliferation and TGF-beta/ALK1 signal transduction
.
EMBO J.
23
,
4018
4028
46
Scharpfenecker
,
M.
,
van Dinther
,
M.
,
Liu
,
Z.
,
van Bezooijen
,
R.L.
,
Zhao
,
Q.
,
Pukac
,
L.
et al (
2007
)
BMP-9 signals via ALK1 and inhibits bFGF-induced endothelial cell proliferation and VEGF-stimulated angiogenesis
.
J. Cell Sci.
120
,
964
972
47
David
,
L.
,
Mallet
,
C.
,
Mazerbourg
,
S.
,
Feige
,
J.J.
and
Bailly
,
S.
(
2007
)
Identification of BMP9 and BMP10 as functional activators of the orphan activin receptor-like kinase 1 (ALK1) in endothelial cells
.
Blood
109
,
1953
1961
48
Zhang
,
C.Y.
,
Yin
,
H.M.
,
Wang
,
H.
,
Su
,
D.
,
Xia
,
Y.
,
Yan
,
L.F.
et al (
2018
)
Transforming growth factor-beta1 regulates the nascent hematopoietic stem cell niche by promoting gluconeogenesis
.
Leukemia
32
,
479
491
49
Thambyrajah
,
R.
,
Fadlullah
,
M.Z.H.
,
Proffitt
,
M.
,
Patel
,
R.
,
Cowley
,
S.M.
,
Kouskoff
,
V.
et al (
2018
)
HDAC1 and HDAC2 modulate TGF-beta signaling during endothelial-to-hematopoietic transition
.
Stem Cell Rep.
10
,
1369
1383
50
Ruetz
,
T.
,
Pfisterer
,
U.
,
Di Stefano
,
B.
,
Ashmore
,
J.
,
Beniazza
,
M.
,
Tian
,
T.V.
et al (
2017
)
Constitutively active SMAD2/3 are broad-scope potentiators of transcription-factor-mediated cellular reprogramming
.
Cell Stem Cell
21
,
791
805.e9
51
Lebrin
,
F.
,
Deckers
,
M.
,
Bertolino
,
P.
and
Ten Dijke
,
P.
(
2005
)
TGF-beta receptor function in the endothelium
.
Cardiovasc. Res.
65
,
599
608
52
Bruveris
,
F.F.
,
Ng
,
E.S.
,
Stanley
,
E.G.
and
Elefanty
,
A.G.
(
2021
)
VEGF, FGF2, and BMP4 regulate transitions of mesoderm to endothelium and blood cells in a human model of yolk sac hematopoiesis
.
Exp. Hematol.
103
,
30
39.e2
53
Akhurst
,
R.J.
,
Lehnert
,
S.A.
,
Faissner
,
A.
and
Duffie
,
E.
(
1990
)
TGF beta in murine morphogenetic processes: the early embryo and cardiogenesis
.
Development
108
,
645
656
54
ten Dijke
,
P.
and
Arthur
,
H.M.
(
2007
)
Extracellular control of TGFbeta signalling in vascular development and disease
.
Nat. Rev. Mol. Cell Biol.
8
,
857
869
55
Goumans
,
M.J.
and
Mummery
,
C.
(
2000
)
Functional analysis of the TGFbeta receptor/Smad pathway through gene ablation in mice
.
Int. J. Dev. Biol.
44
,
253
265
PMID:
[PubMed]
56
Sabbineni
,
H.
,
Verma
,
A.
and
Somanath
,
P.R.
(
2018
)
Isoform-specific effects of transforming growth factor beta on endothelial-to-mesenchymal transition
.
J. Cell Physiol.
233
,
8418
8428
57
Sanford
,
L.P.
,
Ormsby
,
I.
,
Gittenberger-de Groot
,
A.C.
,
Sariola
,
H.
,
Friedman
,
R.
,
Boivin
,
G.P.
et al (
1997
)
TGFbeta2 knockout mice have multiple developmental defects that are non-overlapping with other TGFbeta knockout phenotypes
.
Development
124
,
2659
2670
58
Howell
,
E.D.
,
Yzaguirre
,
A.D.
,
Gao
,
P.
,
Lis
,
R.
,
He
,
B.
,
Lakadamyali
,
M.
et al (
2021
)
Efficient hemogenic endothelial cell specification by RUNX1 is dependent on baseline chromatin accessibility of RUNX1-regulated TGFbeta target genes
.
Genes Dev.
35
,
1475
1489
59
Hass
,
M.R.
,
Brissette
,
D.
,
Parameswaran
,
S.
,
Pujato
,
M.
,
Donmez
,
O.
,
Kottyan
,
L.C.
et al (
2021
)
Runx1 shapes the chromatin landscape via a cascade of direct and indirect targets
.
PLoS Genet.
17
,
e1009574
60
Savorani
,
C.
,
Malinverno
,
M.
,
Seccia
,
R.
,
Maderna
,
C.
,
Giannotta
,
M.
,
Terreran
,
L.
et al (
2021
)
A dual role of YAP in driving TGFbeta-mediated endothelial-to-mesenchymal transition
.
J. Cell Sci.
134
,
jcs251371
61
Lundin
,
V.
,
Sugden
,
W.W.
,
Theodore
,
L.N.
,
Sousa
,
P.M.
,
Han
,
A.
,
Chou
,
S.
et al (
2020
)
YAP regulates hematopoietic stem cell formation in response to the biomechanical forces of blood flow
.
Dev. Cell
52
,
446
60.e5
62
Knezevic
,
K.
,
Bee
,
T.
,
Wilson
,
N.K.
,
Janes
,
M.E.
,
Kinston
,
S.
,
Polderdijk
,
S.
et al (
2011
)
A Runx1-Smad6 rheostat controls Runx1 activity during embryonic hematopoiesis
.
Mol. Cell. Biol.
31
,
2817
2826
63
Duan
,
Z.
,
Zarebski
,
A.
,
Montoya-Durango
,
D.
,
Grimes
,
H.L.
and
Horwitz
,
M.
(
2005
)
Gfi1 coordinates epigenetic repression of p21Cip/WAF1 by recruitment of histone lysine methyltransferase G9a and histone deacetylase 1
.
Mol. Cell. Biol.
25
,
10338
10351
64
Helness
,
A.
,
Fraszczak
,
J.
,
Joly-Beauparlant
,
C.
,
Bagci
,
H.
,
Trahan
,
C.
,
Arman
,
K.
et al (
2021
)
GFI1 tethers the NuRD complex to open and transcriptionally active chromatin in myeloid progenitors
.
Commun. Biol.
4
,
1356
65
Saleque
,
S.
,
Kim
,
J.
,
Rooke
,
H.M.
and
Orkin
,
S.H.
(
2007
)
Epigenetic regulation of hematopoietic differentiation by Gfi-1 and Gfi-1b is mediated by the cofactors CoREST and LSD1
.
Mol. Cell
27
,
562
572
66
Crisan
,
M.
,
Kartalaei
,
P.S.
,
Vink
,
C.S.
,
Yamada-Inagawa
,
T.
,
Bollerot
,
K.
,
van
,
I.W.
et al (
2015
)
BMP signalling differentially regulates distinct haematopoietic stem cell types
.
Nat. Commun.
6
,
8040
67
Kirmizitas
,
A.
,
Meiklejohn
,
S.
,
Ciau-Uitz
,
A.
,
Stephenson
,
R.
and
Patient
,
R.
(
2017
)
Dissecting BMP signaling input into the gene regulatory networks driving specification of the blood stem cell lineage
.
Proc. Natl Acad. Sci. U.S.A.
114
,
5814
5821
68
Pouget
,
C.
,
Peterkin
,
T.
,
Simoes
,
F.C.
,
Lee
,
Y.
,
Traver
,
D.
and
Patient
,
R.
(
2014
)
FGF signalling restricts haematopoietic stem cell specification via modulation of the BMP pathway
.
Nat. Commun.
5
,
5588
69
McGarvey
,
A.C.
,
Rybtsov
,
S.
,
Souilhol
,
C.
,
Tamagno
,
S.
,
Rice
,
R.
,
Hills
,
D.
et al (
2017
)
A molecular roadmap of the AGM region reveals BMPER as a novel regulator of HSC maturation
.
J. Exp. Med.
214
,
3731
3751
70
Zhang
,
C.
,
Lv
,
J.
,
He
,
Q.
,
Wang
,
S.
,
Gao
,
Y.
,
Meng
,
A.
et al (
2014
)
Inhibition of endothelial ERK signalling by Smad1/5 is essential for haematopoietic stem cell emergence
.
Nat. Commun.
5
,
3431
71
Meloche
,
S.
and
Pouyssegur
,
J.
(
2007
)
The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1- to S-phase transition
.
Oncogene
26
,
3227
3239
72
Hong
,
C.C.
,
Peterson
,
Q.P.
,
Hong
,
J.Y.
and
Peterson
,
R.T.
(
2006
)
Artery/vein specification is governed by opposing phosphatidylinositol-3 kinase and MAP kinase/ERK signaling
.
Curr. Biol.
16
,
1366
1372
73
Hou
,
S.
,
Li
,
Z.
,
Zheng
,
X.
,
Gao
,
Y.
,
Dong
,
J.
,
Ni
,
Y.
et al (
2020
)
Embryonic endothelial evolution towards first hematopoietic stem cells revealed by single-cell transcriptomic and functional analyses
.
Cell Res.
30
,
376
392
74
Xia
,
J.
,
Kang
,
Z.
,
Xue
,
Y.
,
Ding
,
Y.
,
Gao
,
S.
,
Zhang
,
Y.
et al (
2021
)
A single-cell resolution developmental atlas of hematopoietic stem and progenitor cell expansion in zebrafish
.
Proc. Natl Acad. Sci. U.S.A.
118
,
e2015748118
75
Dignum
,
T.
,
Varnum-Finney
,
B.
,
Srivatsan
,
S.R.
,
Dozono
,
S.
,
Waltner
,
O.
,
Heck
,
A.M.
et al (
2021
)
Multipotent progenitors and hematopoietic stem cells arise independently from hemogenic endothelium in the mouse embryo
.
Cell Rep.
36
,
109675
76
Canu
,
G.
,
Athanasiadis
,
E.
,
Grandy
,
R.A.
,
Garcia-Bernardo
,
J.
,
Strzelecka
,
P.M.
,
Vallier
,
L.
et al (
2020
)
Analysis of endothelial-to-haematopoietic transition at the single cell level identifies cell cycle regulation as a driver of differentiation
.
Genome Biol.
21
,
157
77
Ulloa
,
B.A.
,
Habbsa
,
S.S.
,
Potts
,
K.S.
,
Lewis
,
A.
,
McKinstry
,
M.
,
Payne
,
S.G.
et al (
2021
)
Definitive hematopoietic stem cells minimally contribute to embryonic hematopoiesis
.
Cell Rep.
36
,
109703
78
Kaya-Okur
,
H.S.
,
Janssens
,
D.H.
,
Henikoff
,
J.G.
,
Ahmad
,
K.
and
Henikoff
,
S.
(
2020
)
Efficient low-cost chromatin profiling with CUT&Tag
.
Nat. Protoc.
15
,
3264
3283
79
Hill
,
C.S.
(
2016
)
Transcriptional control by the SMADs
.
Cold Spring Harb. Perspect. Biol.
8
,
a022079
80
Massague
,
J.
(
2012
)
TGF-beta signaling in development and disease
.
FEBS Lett.
586
,
1833
81
Varricchio
,
L.
,
Iancu-Rubin
,
C.
,
Upadhyaya
,
B.
,
Zingariello
,
M.
,
Martelli
,
F.
,
Verachi
,
P.
et al (
2021
)
TGF-beta1 protein trap AVID200 beneficially affects hematopoiesis and bone marrow fibrosis in myelofibrosis
.
JCI Insight
6
,
e145651
82
Kim
,
B.G.
,
Malek
,
E.
,
Choi
,
S.H.
,
Ignatz-Hoover
,
J.J.
and
Driscoll
,
J.J.
(
2021
)
Novel therapies emerging in oncology to target the TGF-beta pathway
.
J. Hematol. Oncol.
14
,
55
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY). Open access for this article was enabled by the participation of University of Birmingham in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.