• No results found

University of Groningen Non canonical Wnt ligands and cytokine-driven myelopoiesis Mastelaro de Rezende, Marina

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Non canonical Wnt ligands and cytokine-driven myelopoiesis Mastelaro de Rezende, Marina"

Copied!
9
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Non canonical Wnt ligands and cytokine-driven myelopoiesis

Mastelaro de Rezende, Marina

DOI:

10.33612/diss.118670709

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Mastelaro de Rezende, M. (2020). Non canonical Wnt ligands and cytokine-driven myelopoiesis. University

of Groningen. https://doi.org/10.33612/diss.118670709

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)
(3)

158 159 Final Discussion

7. FINAL DISCUSSION

Upstream effectors of intracellular signaling are similar for IL-3 and GM-CSF

Stem cells were first described in the hematopoietic tissue1-3, in which stem cell biology

has been studied extensively. Maintenance of primitive cell potential, while continuously producing specialized and fully mature blood cells, is dependent on proliferation and differentiation. These processes are main events during hematopoiesis and rely on intricate regulatory mechanisms. Alterations in these mechanisms can cause cellular disruption and disease onset (Chapter 1). The extracellular environment as well as intracellular molecules are is critical in guiding signaling events. Cytokines constitute the main class of extracellular molecules involved in hematopoietic regulation4,5;

initially they were denominated “colony-stimulating factors (CSF)”5-7, due to the

induction of the formation of conglomerates consisting of similar cells (i.e. ‘colonies’) in semi-solid medium6,7.

Colonies develop due to the clonal expansion of one cell (hence colonies are also referred to as “colony formation units” – CFU), which has primitive characteristics and gives rise to others8. Only primitive cells can form colonies, as they have the ability

to self-renew and harbor relevant proliferative potential9, which are key features of

undifferentiated cells.

Introduction of the colony formation assay (CFU assay) established the hierarchical relations of hematopoietic cells and their progenitors10-12, and later on provided

evidence for the existence of primitive characteristics in cells obtained from patients with leukemia, the disease in which cancer stem cell research was initiated (Chapters 1 and 3). In addition, the CFU assay demonstrated the requirement of CSFs for colony formation, as no colonies were formed in the absence of these factors8,12,13.

Among the earliest described CSFs, GM-CSF and IL-3 have a broader activity, stimulating committed, multi-, and bipotent progenitors, as compared to M- and G-CSF, which only target the committed progenitors of monocytes/macrophages and granulocytes, respectively6. In our studies, we observed similar colony formation

potential for these cytokines (Chapter 2). There is evidence suggesting that IL-3 targets a slightly more primitive population than GM-CSF14,15. This was confirmed by

the results of our replating CFU assay (Chapter 5), showing that IL-3 induced colony formation for longer periods of time.

Divergent biological roles, and to an even lesser extent the molecular mechanisms involved, for IL-3 and GM-CSF have not been completely established but unveiling fine myeloid regulation as well as population-specific mechanisms may provide insight in the therapeutic relevance of HSCs and/or myeloid imbalances. In fact, this knowledge could even provide tools to fight aging-related changes, as ‘inflamm-aging’16

is an established event characterized by the predominance of specific inflammatory cytokines, including IL-3 and GM-CSF, in the aging individual16. In this context, gathering

information on pharmacological inhibition and protein activation is helpful, as inhibition analysis can assert if and to what extent a pathway is activated.

Similar to the structure of trees, activation of intracellular signaling is outlined in a non-linear fashion, with embranchments representing activation of multiple pathways17, converging to downstream elements17. As funnels, upstream signals are

more likely to affect multiple and even distinct pathways and subsequent cellular outcomes than downstream signals.

JAK kinase activity targets cytoplasmic tyrosines in the unspecific receptor β-chain and its inhibition completely abrogates GM-CSF18,19 and IL-3 activity20-22. At this point,

no difference was observed between IL-3 and GM-CSF, presumably due to the crucial requirement of JAK kinase for both IL-3 and GM-CSF receptor activity and subsequent signaling.

Tyrosine residues in the cytoplasmic portions of the β-chain receptor (Tyr577, Tyr612, Tyr695, Tyr750, Tyr806, and Tyr866) can provide specificity to JAK signaling activation23. Dimerization of the β-chain is ligand-independent. However, only in the

presence of ligand, the α-chain couples to this complex, allowing JAK2 activation and signaling18,24. Thus, the short receptor subunit (the α chain) appears to be involved in

cytokine specificity and triggering of appropriate signaling pathways19,25-27. Interestingly,

in endothelial cells, GM-CSF activity seems to be related to α-chain activation, leading to p85 coupling and induction of PI3K signaling24,28. This evidence of PI3K activation

upstream of JAK2 is in line with our data, which revealed significant effects of PI3K inhibition on colony forming activities by IL-3 and GM-CSF (Chapter 2). Again, there were no apparent differences between IL-3 and GM-CSF, indicating similar routes of initial signal transduction.

Of note, wortmannin and Ly294002 have been described as STAT inhibitors as well, which might have a direct result of PI3K inhibition upstream of JAK24.

IL-3 and GM-CSF induce a distinct pattern of Ca2+ signaling responses

U73122, 2APB and chelerythrine represent other pharmacological agents that had strong (more than 85%) inhibitory effects on colony formation induced by both cytokines (Chapter 2), suggesting important participation of PLC, the IP3R, and PKC, respectively. It was demonstrated that both IL-3 and GM-CSF activate PLCγ2 in Lin-

Sca-1+c-Kit+ cells29; however, activation was transient in response to IL-3, whereas GM-CSF

induced a more sustained activation (up to 30 minutes)29. It is unknown whether

transient and sustained PLC activation result in different downstream effects. There is evidence that, depending on the stimulation, DAG (downstream effector of PLC) can

(4)

Chapter 7 Final Discussion

follow Ca2+ oscillation signals; displaying an oscillatory pattern or exhibiting a sustained

signal, may reflect on PKC activation30. PKC inhibition (by GF109203X) significantly

reduced IL-3 and GM-CSF stimulated formation of subtype specific and total number of colonies, indicating roles for PKC in this event. In addition, we observed differential PKC activation after 15 minutes of stimulation with IL-3 and GM-CSF (Chapter 2). Both cytokines activate the threonine 514 region of PKC in both populations (progenitors and stem cells), indicating priming of PKC activation31, but only IL-3 induced PKCser660

phosphorylation, indicating a phosphosite specific activation by IL-3.

Considering Ca2+ signaling, the IP3R is importantly involved in the frequency and

modulation of the Ca2+ signals32, which can directly influence PKC and CaMKII signaling30,

as well as cellular outcomes33. Interestingly, inhibition due to interference with Ca2+

signaling (using 2APB, BAPTA, calmidazolium and KN-62) seems to be more effective when GM-CSF is used as a stimulus (Chapter 2). This is emphasized by the observed CaMKII phosphorylation, reinforcing the importance of Ca2+ in GM-CSF signaling and

suggesting another difference between GM-CSF and the IL-3 mediated response.

Differential Ca2+ signaling may be reflected by MAPK pathway activation

There is evidence from the literature to suggest that MAPK pathways are activated by PKC and CAMKII29. Since we observed differential PKC activation and CAMKII

phosphorylation for IL-3 and GM-CSF, we postulated this could influence MAPK activation. Transient MEK1/2 phosphorylation in response to IL-3 stimulation culminates in strong ERK1/2 activation, whereas sustained MEK1/2 phosphorylation by GM-CSF leads to decreased ERK1/2 activation29. Unfortunately, we could only

observe ERK1/2 activation after IL-3 stimulation (Chapter 2), which may indicate lower sensitivity of our analysis.

Interestingly, the sustained MEK1/2 activation (as described by Leon and coworkers in 2011), would take place in the same scenario we proposed to be CAMKII-dependent. Different from PKC, the CAMKII activation signal does not oscillate in response to [Ca2+]

i variations, exhibiting a progressive activation pattern30. Extrapolating this data,

we hypothesize that progressive CAMKII activation leads to consistent activation of MEK1/2, triggering self-inhibitory mechanisms, such as feedback loop activation, and resulting in decreased ERK1/2 activation. On the other hand, the oscillatory PKC activation pattern30 (putatively driven by IL-3 stimulation), would lead to a transient

MEK1/2 activation29, without activation of such self-inhibitory mechanisms, and

activation of ERK1/2.

Mild ERK1/2 activation in response to GM-CSF might be caused by modulatory mechanisms in response to sustained pathway activation, whereas strong ERK1/2 activation may reflect a sharper, more pinpointed mechanism of action, in which the

‘on and off’ status are changed faster without the activation of additional feedback loops. We observed robust p38 phosphorylation in response to both cytokines after 15 minutes of stimulation in all populations, except for progenitors in response to GM-CSF (Chapter 2). MAPK signaling has been described to vary between c-Kit+ and

c-Kit- cells, in addition to being PI3K dependent in response to stem cell factor34. Indeed,

after stimulation with either IL-3 or GM-CSF, we observed a stronger effect of PI3K inhibition than of MEK and ERK inhibition, which may indicate an upstream role for PI3K in the Raf/MEK/ERK cascade. In addition, PTEN phosphorylation suggested that Akt and PI3K were not being inhibited, further supporting PI3K participation. Akt activity may reflect GSK3β phosphorylation35, which tended to increase after IL-3 treatment in

the progenitor population; GSK3β is related to numerous intracellular pathways36-40,

including Wnt signaling41-43. Noteworthy, this “population-specific” response highlights

the importance of sensitive techniques, as has been discussed for the leukemic population (Chapter 3).

Figure 6 of Chapter 2 summarizes the main observed differences between IL-3 and GM-CSF treatment. STAT5-independent mechanisms have been implicated in JAK2 activity18,44; this could apply to our results in which little STAT5 phosphorylation

was observed (Chapter 2), despite published data indicating this signal transducer is activated by both IL-3 and GM-CSF26.

Conversely, STAT3 activation was observed under almost all conditions studied (with the exception of HSCs treated with GM-CSF); this was somewhat unexpected as cytokines other than IL-3 and GM-CSF have been more predominantly associated with STAT3 activation45. On the other hand, STAT3 activation by GM-CSF has been shown

to decrease apoptosis45, a well-known effect of this cytokine in the hematopoietic

system46-48.

Alterations in Wnt signaling can contribute to hematopoietic imbalances and malignancies (Chapters 3 and 4). Pathways involving PKC, Ca2+ and PI3K have been

linked to Wnt signaling49-52. Therefore, differences observed in these pathways were

hypothesized to regulate (or be regulated by) Wnt signaling.

The relative functional contribution of Wnt signaling may account for (some) differ-ences between IL-3 and GM-CSF driven signaling

Wnt signaling is increasingly associated with hematopoietic regulation, and its β-catenin independent branch appears to be particularly important for myelopoiesis and aging-related imbalances53-57. Chapters 4, 5 and 6 provide further evidence for

this functional association.

Our comprehensive literature review (Chapter 4) revealed some intriguing elements about noncanonical Wnt signaling in the regulation of the hematopoietic

(5)

162 163

Chapter 7 Final Discussion

system. For example, it highlighted the increased response of LT-HSCs to Wnt5a, leading to quiescence in these cells58,59, which seems to be related to Cdc42 regulation

and apolarity development59. In addition, for the population representing the following

lower level of the hierarchic tree of differentiation, ST-HSCs respond to this ligand with proliferation57,60, which appears to be associated with the activation of canonical

rather than noncanonical Wnt signaling, although a myeloid skewing in differentiation, as occurs during aging, might be observed59,61. These differential cellular responses

indicate both canonical and noncanonical effects of Wnt5a, depending on the target population. As for Wnt ligand stimulation, the cytokines we studied also induced population-specific responses (Chapter 5), further reinforcing highly population-specific intracellular signaling. Interestingly, most studies on Wnt signaling in the hematopoietic system focus on the canonical branch, hampering the identification and establishment of roles for the noncanonical branch. Our review (Chapter 4) exposed numerous situations in which noncanonical Wnt signaling is active and could be of profound importance, as this pathway has relevant roles in hematopoietic ontogenesis, adult hematopoiesis and during aging, which is increasingly being associated with changes in Wnt signaling in this system.

In a heterogeneous environment as bone marrow, where a myriad of compounds simultaneously modulates cellular responses, it is essential that we have a complete overview of the occurring events. In this context, Chapter 5 describes the importance of the interaction between different compounds in modulating hematopoietic activity.

There is evidence to suggest that Wnt5a-induced signaling in the myeloid lineage is associated with Ca2+, PKC and PI3K activation, similar as to what we demonstrated

for IL-3 and GM-CSF. Therefore, we studied the effects of combined stimulation with Wnt ligands and cytokines on myeloid regulation. To our surprise, the prototypical noncanonical Wnt ligand (Wnt5a) showed no (additional) effects in the presence of IL-3 or GM-CSF in the CFU assay. Our investigation also included Wnt5b, as it is structurally close to Wnt5a62, although little is known about its roles in the

hematopoietic regulation. There is evidence for a role of Wnt5b in the regulation of megakaryopoiesis63,64, involving Ca2+ signaling63, but to our knowledge nothing is

currently known with regards to Wnt5b in the myelopoiesis field. Different from what we observed with Wnt5a, our results suggest striking effects of Wnt5b in the presence of IL-3 and GM-CSF in the CFU assay, in addition to potentially underpinning functional differences between IL-3 and GM-CSF (Chapter 5).

Our results not only indicate that the presence of Wnt5b determines the functional outcome of stimulation with cytokines, but also that there are differences between the impact of Wnt ligands and CT99021 (GSK3β inhibitor and β-catenin signaling activator) on the response induced by IL-3 and GM-CSF.

With IL-3 as stimulus, Wnt5b caused a decreased colony formation ratio, the opposite of what was observed in the presence of CT99021, suggesting that Wnt5b activates intracellular signaling not directly involving β-catenin. In addition, colonies could be successfully replated more times in the replating assay, indicative of primitive cell maintenance, which was confirmed by increased Ifitm-1 and GATA2 gene expression as compared to colonies exposed to IL-3 alone. On the other hand, stimulation with GM-CSF in the presence of Wnt5b resulted in the induction of differentiation, as demonstrated by an increase in colony formation and rapid exhaustion of cell potential. In addition, the Wnt5b effect was opposite to that observed in the presence of CT99021, again suggesting the involvement of β-catenin independent Wnt signaling. To our knowledge, this is the first study showing opposite effects of IL-3 and GM-CSF in myeloid regulation. It is likely that Wnt5b acts as a pivotal player underpinning our observed results by reinforcing pathways that are differentially activated by these cytokines.

In the presence of CT99021, IL-3 induced CFU growth, in a similar fashion as observed for GM-CSF+Wnt5b. This strongly suggests colony formation growth as a consequence of β-catenin signaling activation, which is in accordance with the literature60. It has been described that regarding IL-3, Wnt signaling is off65 and Wnt5b

does not seem to activate β-catenin. Conversely, with GM-CSF as a stimulus, Wnt5b may be activating β-catenin-dependent Wnt signaling on its own or sustaining GM-CSF-induced activity. Increased cyclinD1 gene expression in response to GM-CSF stimulation66 reinforces the hypothesis that GM-CSF activates β-catenin signaling, as

cyclinD1 is a well-known target gene downstream of β-catenin67-69.

It is debatable whether there is a differential distribution of cytokines and Wnt ligands in the bone marrow. In fact, even the existence of bone marrow niches (endosteal and vascular) is questionable70,71, with hematopoietic cells in diverse

maturation stages spreading across the bones71,72. In addition, active movement

by hematopoietic cells in the bone marrow73,74 hampers the understanding of how

extracellular signals modulate hematopoiesis. There is evidence that IL-3 has a higher activity on LT-HSCs in comparison with GM-CSF75,76, whereas GM-CSF is more active on

progenitors77,78. Therefore, together with our results, we propose that more primitive

HSCs, which are responsive to IL-3, have an intracellular environment unfavorable to β-catenin and Ca2+ signaling activation. On the other hand, with activation of Ca2+

signaling (as we observe after GM-CSF treatment), the intracellular environment of less primitive hematopoietic cells might be more favorable to Wnt/β-catenin independent signaling. This would explain why in the presence of Wnt5b, IL-3 drives maintenance of cell potential and GM-CSF triggers differentiation.

(6)

Chapter 7 Final Discussion

Wnt5 inhibition restores the potential of aged progenitors

The environment in which adult hematopoiesis occurs changes with aging; most of the hematopoietic balance is lost, with LT-HSCs prolonging quiescence and ST-HSCs proliferating to compensate for functional losses79,80. In addition, this environment

is characterized by an increase in β-catenin independent Wnt ligands, mainly the prototypical Wnt5a (Chapters 5 and 6), which really put our investigation on IL-3 and GM-CSF to the test.

In Chapter 6, we used CFU assays to show how Wnt5b treatment in young cells only partially mimicked the functional results of aged cells, and Wnt5 inhibition in aged cells mimicked the results observed in young cells just to a certain extent. Yet, Wnt5 inhibition in aged cells provided definitive evidence of this protein’s participation in myeloid regulation in the aging environment. Thus, Wnt5 inhibition by Box5 strikingly increased progenitor potential in aged cells, which seemed to be caused by upregulation of self-renewal and downregulation of commitment and differentiation.

Our results (Chapter 6) showed that Box5 induced upregulation of myc, E2F and Cdk1, which are genes associated with self-renewal81-84. Interestingly, both myc

and Cdk1 are targets of β-catenin signaling85,86, but they can be activated by other

pathways as well; this was likely the case in our experimental setup, since β-catenin was inhibited. Gene Set Enrichment Analysis of the obtained mRNA sequencing data also demonstrated that Box5 induced upregulation of the PI3K/Akt pathway, which may underpin its effects on gene expression85. Indeed, Akt is known to phosphorylate

and inhibit GSK3β40, and GSK3β inhibition is a hallmark for stem cell maintenance

(together with MTORC1 inhibition87).

p53 was amongst the most downregulated genes after Box5 treatment in old cells, and together with Notch1 and Fos, which were downregulated as well, it is involved in hematopoietic specification and lineage commitment88. In addition, STAT5,

a well-known driver of differentiation88,89, and mTORC1 were downregulated as well87.

Although strong evidence exists for differentiation arrest by Box5 treatment, other mechanisms might be involved in the maintenance of cell potential. Downregulation of hypoxia, TNFα and KRAS may be linked to β-catenin signaling inhibition, as these pathways are connected to Wnt/β-catenin in numerous systems90,91. β-Catenin

inhibition in aged animals treated with Box5 suggests activation of this pathway in the absence of the inhibitor, as proposed above, which also agrees with evidence from the literature indicating that activation of β-catenin is related to proliferation in hematopoietic progenitors92,93.

Noncanonical Wnt5b modulates myelopoiesis driven by IL-3 and GM-CSF

In conclusion, IL-3- and GM-CSF-driven myelopoiesis might not be as similar as it was considered to be over the last several years. Although they can produce similar cellular outcomes depending on the method used, specific intracellular signaling events triggered may differ at some point further up or down the tree, which can be highlighted in the presence of other compounds, such as Wnt ligands. These ligands appear to be important components of hematopoietic ontogenesis and modulation in adult life, as well as responsible for imbalances of senescence. Wnt5b is shown here as a critical modulator of myelopoiesis driven by IL-3 and GM-CSF stimulation. Of note, during aging, Wnt5a is likely to be involved in the developing imbalances as well. In addition, we reveal that activation of β-catenin signaling is not sufficient to fix the imbalances that occur during aging, as complete abrogation of Wnt5 activity was needed. Furthermore, the results provide evidence for the modulation of myelopoiesis by noncanonical Wnts, and point out differences between the response to cytokines and the distinct involvement of these Wnts. In addition, experimental tools and approaches implemented and/or introduced in our studies could prove useful for the investigation of hematopoietic senescence and (future) therapeutics.

(7)

166 167

Chapter 7 Final Discussion

CONCLUDING STATEMENTS

1. Hematopoietic regulation is essential for nutrient and oxygen distribution, as well as immune and inflammatory responses. These functions are exerted by short-lived cells, which need to be continuously produced by proliferation and differentiation of primitive cells;

2. Differences exist between the effects of IL-3 and GM-CSF on myelopoiesis, by targeting different hematopoietic cell fractions and distinct cellular outcomes;

3. Activation of PLC, PKC, Ca2+ and MEK/ERK signaling by IL-3 or GM-CSF stimulation

are involved in colony formation by these cytokines;

4. Wnt5a, the prototypical noncanonical ligand, is associated with Ca2+, PKC and

PI3K activation in the hematopoietic system;

5. Wnt5b, rather than Wnt5a, affects IL-3- and GM-CSF-driven myelopoiesis and is pivotal in the differential cellular outcomes primed by these cytokines;

6. Noncanonical Wnt signaling plays a central role in (the development of) hematopoietic imbalances in the aging individual;

7. Activation of canonical Wnt signaling (with CT99021) is not sufficient to completely abrogate the effects of aging on the cell potential of progenitors;

8. Abrogation of Wnt5 signaling (thus, both Wnt5a and Wnt5b) with Box5 completely restores progenitor cell potential in aged cells by stimulating self-renewal and inhibiting commitment and differentiation.

REFERENCES

1 Ford, C. E., Hamerton, J. L., Barnes, D. W. & LoutitO, J. F. Cytological identification of radiation-chimaeras. Nature 177, 452-454

(1956).

2 TILL, J. E. & McCULLOCH, E. A. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells.

Radiat Res 14, 213-222 (1961).

3 Becker, A. J., McCulloch, E. A. & Till, J. E. Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature

197, 452-454 (1963).

4 Rieger, M. A. & Schroeder, T. Hema-topoiesis. Cold Spring Harb Perspect Biol 4,

doi:10.1101/cshperspect.a008250 (2012). 5 Robb, L. Cy tokine receptors and

hematopoietic differentiation. Oncogene

26, 6715-6723, doi:10.1038/sj.onc.1210756

(2007).

6 Metcalf, D. The granulocyte-macrophage colony stimulating factors. Cell 43, 5-6

(1985).

7 Metcalf, D. Hematopoietic cytokines. Blood

111, 485-491,

doi:10.1182/blood-2007-03-079681 (2008).

8 Metcalf, D. The molecular biology and functions of the granulocyte-macrophage colony-stimulating factors. Blood 67,

257-267 (1986).

9 Wu, A. M., Till, J. E., Siminovitch, L. & McCulloch, E. A. Cytological evidence for a relationship between normal hemotopoietic colony-forming cells and cells of the lymphoid system. J Exp Med

127, 455-464 (1968).

10 Metcalf, D., Johnson, G. R. & Mandel, T. E. Colony formation in agar by multipotential hemopoietic cells. Journal of cellular

physiology 98, 401-420, doi:10.1002/

jcp.1040980216 (1979).

11 Abramson, S., Miller, R. G. & Phillips, R. A. The identification in adult bone marrow of pluripotent and restricted stem cells of the myeloid and lymphoid systems. J Exp Med

145, 1567-1579 (1977).

12 Bradley, T. R. & Metcalf, D. The growth of mouse bone marrow cells in vitro. Aust J

Exp Biol Med Sci 44, 287-299 (1966).

13 Pluznik, D. H. & Sachs, L. The induction of clones of normal mast cells by a substance from conditioned medium. Exp Cell Res 43,

553-563 (1966).

14 Donahue, R. E. et al. Human IL-3 and GM-CSF act synergistically in stimulating hematopoiesis in primates. Science 241,

1820-1823 (1988).

15 Saeland, S. et al. Combined and sequential effects of human IL-3 and GM-CSF on the proliferation of CD34+ hematopoietic cells from cord blood. Blood 73, 1195-1201

(1989).

16 Kovtonyuk, L. V., Fritsch, K., Feng, X., Manz, M. G. & Takizawa, H. Inflamm-Aging of Hematopoiesis, Hematopoietic Stem Cells, and the Bone Marrow Microenvironment.

Front Immunol 7, 502, doi:10.3389/

fimmu.2016.00502 (2016).

17 Rhee, A., Cheong, R. & Levchenko, A. Noise decomposition of intracellular biochemical signaling networks using nonequivalent reporters. Proc Natl Acad

Sci U S A 111, 17330-17335, doi:10.1073/

pnas.1411932111 (2014).

18 Watanabe, S., Itoh, T. & Arai, K. Roles of JAK kinases in human GM-CSF receptor signal transduction. J Allergy Clin Immunol 98,

S183-191 (1996).

19 D’Andrea, R. J. & Gonda, T. J. A model for assembly and activation of the GM-CSF, IL-3 and IL-5 receptors: insights from activated mutants of the common beta subunit. Exp Hematol 28, 231-243 (2000).

20 O’Shea, J. J., Kontzias, A., Yamaoka, K., Tanaka, Y. & Laurence, A. Janus kinase inhibitors in autoimmune diseases.

Ann Rheum Dis 72 Suppl 2, ii111-115,

doi:10.1136/annrheumdis-2012-202576 (2013).

21 Kovanen, P. E. et al. Regulation of Jak2 tyrosine kinase by protein kinase C during macrophage differentiation of IL-3-dependent myeloid progenitor cells. Blood

95, 1626-1632 (2000).

22 Leonard, W. J. & O’Shea, J. J. Jaks and STATs: biological implications. Annu Rev Immunol

16, 293-322, doi:10.1146/annurev.

immunol.16.1.293 (1998).

(8)

Chapter 7 Final Discussion

23 Guthridge, M. A. et al. Mechanism of activation of the GM-CSF, IL-3, and IL-5 family of receptors. Stem Cells 16, 301-313,

doi:10.1002/stem.160301 (1998).

24 Dhar-Mascareno, M., Pedraza, A. & Golde, D. W. PI3-kinase activation by GM-CSF in endothelium is upstream of Jak/Stat pathway: role of alphaGMR. Biochem

Biophys Res Commun 337, 551-556,

doi:10.1016/j.bbrc.2005.09.088 (2005). 25 Barry, S. C. et al. Roles of the N and C

terminal domains of the interleukin-3 receptor alpha chain in receptor function.

Blood 89, 842-852 (1997).

26 Hara, T. & Miyajima, A. Function and signal transduction mediated by the interleukin 3 receptor system in hematopoiesis.

Stem Cells 14, 605-618, doi:10.1002/

stem.140605 (1996).

27 Wittwer, N. L. et al. High CD123 levels enhance proliferation in response to IL-3, but reduce chemotaxis by downregulating CXCR4 expression. Blood Adv 1, 1067-1079,

doi:10.1182/bloodadvances.2016002931 (2017).

28 Perugini, M. et al. Alternative modes of GM-CSF receptor activation revealed using activated mutants of the common beta-subunit. Blood 115, 3346-3353,

doi:10.1182 /blood-20 09 - 08-2358 4 6 (2010).

29 Leon, C. M. et al. Requirement for PLCγ2 in IL-3 and GM-CSF-stimulated MEK/ERK phosphorylation in murine and human hematopoietic stem/progenitor cells. J

Cell Physiol 226, 1780-1792, doi:10.1002/

jcp.22507 (2011).

30 Cullen, P. J. Calcium signalling: the ups and downs of protein kinase C. Curr Biol 13,

R699-701 (2003).

31 Garczarczyk, D., Szeker, K., Galfi, P., Csordas, A. & Hofmann, J. Protein kinase Cgamma in colon cancer cells: expression, Thr514 phosphorylation and sensitivity to butyrate-mediated upregulation as related to the degree of differentiation. Chem

Biol Interact 185, 25-32, doi:10.1016/j.

cbi.2010.02.035 (2010).

32 Wieder, N., Fink, R. & von Wegner, F. Exact stochastic simulation of a calcium microdomain reveals the impact of Ca²+ fluctuations on IP₃R gating. Biophys J 108,

557-567, doi:10.1016/j.bpj.2014.11.3458 (2015).

33 Paredes-Gamero, E. J., Leon, C. M., Borojevic, R., Oshiro, M. E. & Ferreira, A. T. Changes in intracellular Ca2+ levels induced by cytokines and P2 agonists differentially modulate proliferation or commitment with macrophage differentiation in murine hematopoietic cells. J Biol Chem 283, 31909-31919,

doi:10.1074/jbc.M801990200 (2008). 34 Wandzioch, E., Edling, C. E., Palmer, R.

H., Carlsson, L. & Hallberg, B. Activation of the MAP kinase pathway by c-Kit is PI-3 kinase dependent in hematopoietic progenitor/stem cell lines. Blood 104,

51-57, doi:10.1182/blood-2003-07-2554 (2004).

35 Inuzuka, H. et al. Mcl-1 ubiquitination and destruction. Oncotarget 2, 239-244,

doi:10.18632/oncotarget.242 (2011). 36 Kitajima, K. et al. GSK3β inhibition activates

the CDX/HOX pathway and promotes hemogenic endothelial progenitor differentiation from human pluripotent stem cells. Exp Hematol 44, 68-74.e61-10,

doi:10.1016/j.exphem.2015.09.007 (2016). 37 Tang, X. L., Wang, C. N., Zhu, X. Y. & Ni,

X. Protein tyrosine phosphatase SHP-1 modulates osteoblast differentiation through direct association with and dephosphorylation of GSK3β. Mol Cell

Endocrinol 439, 203-212, doi:10.1016/j.

mce.2016.08.048 (2017).

38 Moon, J. B. et al. Akt induces osteoclast differentiation through regulating the GSK3β/NFATc1 signaling cascade. J

Immunol 188, 163-169, doi:10.4049/

jimmunol.1101254 (2012).

39 Morroni, F., Sita, G., Tarozzi, A., Rimondini, R. & Hrelia, P. Early effects of Aβ1-42 oligomers injection in mice: Involvement of PI3K/Akt/GSK3 and MAPK/ERK1/2 pathways. Behav Brain Res 314, 106-115,

doi:10.1016/j.bbr.2016.08.002 (2016).

40 Hermida, M. A., Dinesh Kumar, J. & Leslie, N. R. GSK3 and its interactions with the PI3K /AK T/mTOR signalling network.

Adv Biol Regul 65, 5-15, doi:10.1016/j.

jbior.2017.06.003 (2017).

41 Zhu, Z. et al. Lithium stimulates human bone marrow derived mesenchymal stem cell proliferation through GSK-3β-dependent β-catenin/Wnt pathway activation. FEBS J 281, 5371-5389,

doi:10.1111/febs.13081 (2014).

42 Wu, D. & Pan, W. GSK3: a multifaceted kinase in Wnt signaling. Trends Biochem Sci

35, 161-168, doi:10.1016/j.tibs.2009.10.002

(2010).

43 Duinhouwer, L. E. et al. Wnt3a protein reduces growth factor-driven expansion of human hematopoietic stem and progenitor cells in serum-free cultures.

PLoS One 10, e0119086, doi:10.1371/

journal.pone.0119086 (2015).

44 Ihle, J. N. STATs: signal transducers and activators of transcription. Cell 84,

331-334 (1996).

45 Gu, L., Chiang, K. Y., Zhu, N., Findley, H. W. & Zhou, M. Contribution of STAT3 to the activation of survivin by GM-CSF in CD34+ cell lines. Exp Hematol 35, 957-966,

doi:10.1016/j.exphem.2007.03.007 (2007). 46 Chiewchengchol, D. et al. The protective

effect of GM-CSF on serum-induced neutrophil apoptosis in juvenile systemic lupus erythematosus patients. Clin

Rheumatol 34, 85-91,

doi:10.1007/s10067-014-2800-2 (2015).

47 Wong, C. K. et al. MicroRNA-21* regulates the prosurvival effect of GM-CSF on human eosinophils. Immunobiology 218,

255-262, doi:10.1016/j.imbio.2012.05.019 (2013).

48 Huang, X. et al. GM-CSF inhibits apoptosis of neural cells via regulating the expression of apoptosis-related proteins.

Neurosci Res 58, 50-57, doi:10.1016/j.

neures.2007.01.015 (2007).

49 Kohn, A. D. & Moon, R. T. Wnt and calcium signaling: beta-catenin-independent pathways. Cell calcium 38, 439-446,

doi:10.1016/j.ceca.2005.06.022 (2005).

50 Kühl, M., Sheldahl, L. C., Park, M., Miller, J. R. & Moon, R. T. The Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes shape. Trends Genet 16, 279-283,

doi:10.1016/s0168-9525(00) 02028-x (2000).

51 Voskas, D., Ling, L. S. & Woodgett, J. R. Does GSK-3 provide a shortcut for PI3K activation of Wnt signalling? F1000 biology

reports 2, 82, doi:10.3410/B2-82 (2010).

52 Ling, L. S., Voskas, D. & Woodgett, J. R. Activation of PDK-1 maintains mouse embryonic stem cell self-renewal in a PKB-dependent manner. Oncogene 32,

5397-5408, doi:10.1038/onc.2013.44 (2013). 53 Austin, T. W., Solar, G. P., Ziegler, F. C., Liem,

L. & Matthews, W. A role for the Wnt gene family in hematopoiesis: expansion of multilineage progenitor cells. Blood 89,

3624-3635 (1997).

54 Baba, Y., Garrett, K. P. & Kincade, P. W. Constitutively active beta-catenin confers multilineage differentiation potential on lymphoid and myeloid progenitors.

Immunity 23, 599-609, doi:10.1016/j.

immuni.2005.10.009 (2005).

55 Brandon, C., Eisenberg, L. M. & Eisenberg, C. A. WNT signaling modulates the diversification of hematopoietic cells.

Blood 96, 4132-4141 (2000).

56 Famili, F. et al. High Levels of Canonical Wnt Signaling Lead to Loss of Stemness and Increased Differentiation in Hematopoietic Stem Cells. Stem Cell Reports 6, 652-659,

doi:10.1016/j.stemcr.2016.04.009 (2016). 57 Nemeth, M. J. & Bodine, D. M. Regulation

of hematopoiesis and the hematopoietic stem cell niche by Wnt signaling pathways.

Cell Res 17, 746-758, doi:10.1038/cr.2007.69

(2007).

58 Povinelli, B. J. & Nemeth, M. J. Wnt5a regulates hematopoietic stem cell proliferation and repopulation through the Ryk receptor. Stem cells 32, 105-115,

doi:10.1002/stem.1513 (2014).

59 Florian, M. C. et al. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature

503, 392-396, doi:10.1038/nature12631

(2013).

(9)

170 171

Chapter 7 Final Discussion

60 Trowbridge, J. J., Guezguez, B., Moon, R. T. & Bhatia, M. Wnt3a activates dormant c-Kit(-) bone marrow-derived cells with short-term multilineage hematopoietic reconstitution capacity. Stem cells 28,

1379-1389, doi:10.1002/stem.457 (2010). 61 Xiao, J., Zhou, H., Wu, N. & Wu, L. The

non-canonical Wnt pathway negatively regulates dendritic cell differentiation by inhibiting the expansion of Flt3(+) l y m p h o c y te - p r im e d m ul t ip o ten t precursors. Cell Mol Immunol 13, 593-604,

doi:10.1038/cmi.2015.39 (2016).

62 Kessenbrock, K. et al. Diverse regulation of mammary epithelial growth and branching morphogenesis through noncanonical Wnt signaling. Proceedings of the National

Academy of Sciences of the United States of America 114, 3121-3126, doi:10.1073/

pnas.1701464114 (2017).

63 Louwette, S. et al. Regulator of G-protein signaling 18 controls megakaryopoiesis and the cilia-mediated vertebrate mechanosensory system. FASEB journal

: official publication of the Federation of American Societies for Experimental Biology 26, 2125-2136,

doi:10.1096/fj.11-198739 (2012).

64 Macaulay, I. C. et al. Canonical Wnt signaling in megakaryocytes regulates proplatelet formation. Blood 121, 188-196,

doi:10.1182/blood-2012-03-416875 (2013). 65 Nteliopoulos, G., Marley, S. B. & Gordon, M.

Y. Influence of PI-3K/Akt pathway on Wnt signalling in regulating myeloid progenitor cell proliferation. Evidence for a role of autocrine/paracrine Wnt regulation.

British journal of haematology 146,

637-651, doi:10.1111/j.1365-2141.2009.07823.x (2009).

66 Qiu, J., Papatsenko, D., Niu, X., Schaniel, C. & Moore, K. Divisional history and hematopoietic stem cell function during homeostasis. Stem Cell Reports 2, 473-490,

doi:10.1016/j.stemcr.2014.01.016 (2014).

67 Yamazaki, K. et al. Increased cyclin D1 expression in cancer of the ampulla of Vater: relevance to nuclear beta catenin accumulation and k-ras gene mutation.

Mol Pathol 56, 336-341, doi:10.1136/

mp.56.6.336 (2003).

68 Li, Y. J., Wei, Z. M., Meng, Y. X. & Ji, X. R. Beta-catenin up-regulates the expression of cyclinD1, c-myc and MMP-7 in human pancreatic cancer: relationships with carcinogenesis and metastasis. World J

Gastroenterol 11, 2117-2123, doi:10.3748/

wjg.v11.i14.2117 (2005).

69 Yang, Z., Kim, S., Mahajan, S., Zamani, A. & Faccio, R. Phospholipase Cγ1 (PLCγ1) Controls Osteoclast Numbers via Colony-stimulating Factor 1 (CSF-1)-dependent D ia c y lg l y ce ro l / β - C a te nin /Cy c lin D1 Pathway. J Biol Chem 292, 1178-1186,

doi:10.1074/jbc.M116.764928 (2017). 70 Hoggatt, J. & Scadden, D. T. The stem

cell niche: tissue physiology at a single cell level. J Clin Invest 122, 3029-3034,

doi:10.1172/JCI60238 (2012).

71 Nombela-Arrieta, C. & Manz, M. G. Quantification and three-dimensional microanatomical organization of the bone marrow. Blood Adv 1, 407-416, doi:10.1182/

bloodadvances.2016003194 (2017). 72 Kiel, M. J., Radice, G. L. & Morrison, S. J.

Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Cell Stem Cell 1, 204-217,

doi:10.1016/j.stem.2007.06.001 (2007). 73 Wright, D. E., Wagers, A. J., Gulati, A. P.,

Johnson, F. L. & Weissman, I. L. Physio-logical migration of hematopoietic stem and progenitor cells. Science 294,

1933-1936, doi:10.1126/science.1064081 (2001). 74 Nakamura-Ishizu, A. & Suda, T. Dynamic

Changes in the Niche with N-Cadherin Revisited: The HSC “Niche Herein”. Cell

Stem Cell 24, 355-356, doi:10.1016/j.

stem.2019.02.007 (2019).

75 Geijsen, N., Koenderman, L. & Coffer, P. J. Specificity in cytokine signal transduction: lessons learned from the IL-3/IL-5/GM-CSF receptor family. Cytokine & growth factor

reviews 12, 19-25 (2001).

76 Manz, M. G., Miyamoto, T., Akashi, K. & Weissman, I. L. Prospective isolation of human clonogenic common myeloid progenitors. Proceedings of the National

Academy of Sciences of the United States of America 99, 11872-11877, doi:10.1073/

pnas.172384399 (2002).

77 Wognum, A. W., Westerman, Y., Visser, T. P. & Wagemaker, G. Distribution of receptors for granulocyte-macrophage colony-stimulating factor on immature CD34+ bone marrow cells, differentiating monomyeloid progenitors, and mature blood cell subsets. Blood 84, 764-774

(1994).

78 Rosas, M., Gordon, S. & Taylor, P. R. Characterisation of the expression and function of the GM-CSF receptor alpha-chain in mice. Eur J Immunol 37, 2518-2528,

doi:10.1002/eji.200636892 (2007). 79 Dykstra, B., Olthof, S., Schreuder, J.,

Ritsema, M. & de Haan, G. Clonal analysis reveals multiple functional defects of aged murine hematopoietic stem cells.

J Exp Med 208, 2691-2703, doi:10.1084/

jem.20111490 (2011).

80 Shlush, L. I. Age-related clonal hema-topoiesis. Blood 131, 496-504, doi:10.1182/

blood-2017-07-746453 (2018).

81 Viatour, P. Bridges between Cell Cycle Regulation and Self-Renewal Maintenance. Genes Cancer 3, 670-677,

doi:10.1177/1947601913481355 (2012). 82 Levi, B. P. & Morrison, S. J. Stem cells use

distinct self-renewal programs at different ages. Cold Spring Harb Symp Quant Biol

73, 539-553, doi:10.1101/sqb.2008.73.049

(2008).

83 Yeo, H. C. et al. Integrated transcriptome and binding sites analysis implicates E2F in the regulation of self-renewal in human pluripotent stem cells. PLoS One 6, e27231,

doi:10.1371/journal.pone.0027231 (2011). 84 Wilson, A. et al. c-Myc controls the balance

between hematopoietic stem cell self-renewal and differentiation. Genes Dev 18,

2747-2763, doi:10.1101/gad.313104 (2004).

85 de Cássia Viu Carrara, R. et al. Expression differences of genes in the PI3K/AKT, WNT/b-catenin, SHH, NOTCH and MAPK signaling pathways in CD34+ hematopoietic cells obtained from chronic phase patients with chronic myeloid leukemia and from healthy controls. Clin

Transl Oncol 20, 542-549, doi:10.1007/

s12094-017-1751-x (2018).

86 Lim, S. & Kaldis, P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation.

Development 140, 3079-3093, doi:10.1242/

dev.091744 (2013).

87 Nguyen-McCarty, M. & Klein, P. S. Autophagy is a signature of a signaling network that maintains hematopoietic stem cells. PLoS One 12, e0177054,

doi:10.1371/journal.pone.0177054 (2017). 88 Oommen, K. S. & Newman, A. P. Co-regulation by Notch and Fos is required for cell fate specification of intermediate precursors during C. elegans uterine development. Development 134,

3999-4009, doi:10.1242/dev.002741 (2007). 89 Schepers, H., Wierenga, A. T., Vellenga,

E. & Schuringa, J. J. STAT5-mediated self-renewal of normal hematopoietic and leukemic stem cells. JAKSTAT 1, 13-22,

doi:10.4161/jkst.19316 (2012).

90 Pacheco-Pinedo, E. C. & Morrisey, E. E. Wnt and Kras signaling-dark siblings in lung cancer. Oncotarget 2, 569-574,

doi:10.18632/oncotarget.305 (2011). 91 Jang, J. et al. WNT/β-catenin pathway

m o d ula te s t h e T N F - α - in d u ce d inflammatory response in bronchial epithelial cells. Biochem Biophys Res

Commun 484, 442-449, doi:10.1016/j.

bbrc.2017.01.156 (2017).

92 Staal, F. J., Chhatta, A. & Mikkers, H. Caught in a Wnt storm: Complexities of Wnt signaling in hematopoiesis. Exp

Hematol 44, 451-457, doi:10.1016/j.

exphem.2016.03.004 (2016).

93 Willert, K. et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 423, 448-452, doi:10.1038/

nature01611 (2003).

Referenties

GERELATEERDE DOCUMENTEN

Chondrocyte proliferation, matrix production and hypertrophy in the GP is responsible for the rate of longitudinal growth as well as for the ultimate length of all endochondral

Financial support for the costs associated with the publication of this thesis from the Depart- ment of Pediatrics of the Leiden University Medical Centre, the

Chondrocyte proliferation, matrix production and hypertrophy in the GP is responsible for the rate of longitudinal growth as well as for the ultimate length of all endochondral

(A-L) Immunostaining for β-catenin combined with Alcian blue (AB) staining (A,E), combined von Kossa-Toluidine blue staining (F), hematoxylin/eosin staining (G), gene expression

Relatively high levels of β-catenin signal- ing arising upon expression of Apc ∆15/1638N blocked the differentiation of SPC to both chondrocytes and osteoblasts,

KSFrt-Apc si cells displayed significantly increased endogenous levels of BMP signaling in comparison to control KSFrt-mtApc si cells (p < 0.01; Figure 5A). In these

The maintenance of progenitor cell phenotype by Wnt5b treatment in presence of IL-3 might explain why Wnt5b caused an initial decrease in the number of colonies in the CFU

- Non-canonical Wnt signalling is associated to hematopoietic senescence (this thesis) - Understanding the mechanisms involved in cytokine-driven myelopoiesis and