• No results found

Inhibition of signaling cascades in osteoblast differentiation and fibrosis

N/A
N/A
Protected

Academic year: 2021

Share "Inhibition of signaling cascades in osteoblast differentiation and fibrosis"

Copied!
13
0
0

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

Hele tekst

(1)

Inhibition of signaling cascades in osteoblast differentiation and fibrosis

Krause, C.

Citation

Krause, C. (2011, October 5). Inhibition of signaling cascades in osteoblast differentiation and fibrosis. Retrieved from https://hdl.handle.net/1887/17892

Version: Corrected Publisher’s Version

License: Licence agreement concerning inclusion of doctoral thesis in the Institutional Repository of the University of Leiden

Downloaded from: https://hdl.handle.net/1887/17892

Note: To cite this publication please use the final published version (if applicable).

(2)

Noggin

Krause C, Guzman A, Knaus P (2011) published

Int J Biochem Cell Biol. 2011 Apr;43(4):478-81.

(3)
(4)

Chapter 2

Noggin

2.1 Abstract

Metabologens initiate, promote and maintain morphogenesis and adult tissue home- ostasis. Bone morphogenetic proteins (BMPs) which belong to the transforming growth factor-β (TGF-β) superfamily, represent one of the major classes of metabologens that regulate ectoderm, mesoderm and endoderm derived tissue formation. In order to temporally and spatially control BMP initiated signaling cascades, a tight regulatory network is needed to maintain concinnity. There are a number of ways how BMP signaling can be inhibited or more likely be modified, among which the direct ex- tracellular inhibition through cysteine-knot containing proteins from the DAN-, the twisted gastrulation-, chordin- and Noggin-family is a classic. This review focuses on Noggin and its impact on the vast array of BMP driven actions and thereby invites the ever-growing BMP research field to (re-) investigate Noggin’s function in detail.

Keywords

BMP, metabologen, tissue patterning and homeostasis

2.2 Introduction

Noggin, encoded by the NOG gene, is a secreted homodimeric glycoprotein with a molecular mass of 64 kDa, and was discovered by R. Harland by its ability to induce secondary axis formation in Xenopus embryos [21]. Noggin rescues dorsal develop- ment in UV-induced ventralized Xenopus embryos and injection of the putative cDNA results in large heads, hence the name Noggin [21]. Nowadays, Noggin is known to regulate a major class of metabologens, the so called bone morphogenetic proteins (BMPs). It is suggested that due to excessive BMP action Noggin null mice display serious developmental abnormalities [12, 24].

23

(5)

24 Chapter 2. Noggin

LigandSynonymReceptorsBMPtissuelocationInduction ifNoggin expres- sion

Inhibition byNog- ginμCT BV/TV (%) BMP-2BMP-2AALK-3,-6;BMPR-II;En- doglin;TGF-RIII;Act- RIIA,-RIIB

intramembranousbone,bloodvessels,mus- cle,cartilage,teeth,liver,heart,sperm,hair follicle

(↑)YES BMP-3OsteogeninAlk-4;Act-RIIBlung,kidney,intramembranousbone,carti- lage,lung,teeth–NO BMP-4BMP-2BALK-3,-6;Act-RIIA; BMPR-II;TGF-RIIIintramembranousbone,muscle,uterus,car- tilage,teeth,kidney,gut,salivarygland, liver,pancreas,lung,heart,ovaries,sperm, hairfollicle

(↑)YES BMP-5––intramembranousbone,cartilage,kidney, ureter,pancreas,lung,heart–YES BMP-6Vgr-1ALK-2,-3,-6;Act-RIIA,- RIIB;BMPR-IIheart,cartilage,ureter,pancreas,heart, ovaries,epidermis,liver(↑)NO BMP-7Op-1Alk-3,-6;Act-RIIA,-RIIB; BMPR-IIkidney,intramembranousbone,cartilage, synovium,eye,salivarygland,liver,pan- creas,lung,heart,ovaries,sperm,epidermis

(↑)YES BMP-8Op-2, BMP8b–intramembranousbone,ovaries,sperm–– BMP-9ALK-1,-2;Act-RIIA,-RIIB; BMPR-II;Endoglinliver,CNS(↑)NO BMP-10–ALK-1,-3,-6;Act-RIIA,- RIIB;BMPR-IIheart–NO BMP-11GDF-11Alk-4,-5,-7;Act-RIIA,-RIIBCNS–NO BMP-12GDF-7, CDMP-3ALK-3,-6;Act-RIIA; BMPR-IItendons,CNS,cartilage–– BMP-13GDF-6, CDMP-2ALK-3,-6;BMPR-IItendons,cartilage–YES BMP-14GDF-5, CDMP-1Alk-6;Act-RIIA,-RIIB;BM- PRII;TGF-RIIIcartilage,synovium,eye(↑)YES BMP-15GDF-9bALK-6;Act-RIIA;BMPR-IIovaries–NO Table1:Overviewofmembersofthebonemorphogeneticprotein(BMP)family,theirknownBMPtypesIandBMPtypeIIreceptor andthedetectedtissueexpressionpatterns.WhethertherespectiveBMPisknowntoinduceNogginexpressionisindicatedwithan arrow()anditssusceptibilitytoNoggininhibitionisdepictedviaYES(inhibitionthroughNoggindetected)orNO(inhibitionthrough Nogginnotdetected).Activinreceptor-likekinase(ALK);BMPtypeIIreceptor(BMPR-II);activintypeIIreceptor(ActRII);transforming growthfactorreceptorIII(TGF-β-RIII);centralnervoussystem(CNS);unknown(–)

(6)

2.3. Structure 25

2.3 Structure

Noggin’s primary structure consists of an acidic amino-terminal and a cysteine-rich carboxy-terminal region. Through the formation of cystine knots, the carboxy-terminal region is used to classify BMP antagonists into three distinct subfamilies: CAN (eight membered ring), twisted gastrulation (nine-membered ring) as well as chordin and Noggin (ten-membered ring) [1]. The topology of Noggin resembles BMPs in a two- fold axis of symmetry. The BMP-dimer is shaped like a butterfly with wings extending from a core body that mediates dimerization. Noggin dimerizes via a core body, from which 2 pairs of β-strands extend preceding by a N-terminal segment of about 20 amino acids, the so called ’clip’ segment. This clip snakes around the BMP ligand and occludes the surfaces of the growth factor from binding to both the BMP type I and the type II receptors (Figure 1) [8]. This resembles a conserved mode of binding as this clip is also described for crossveinless-2, another member of the antagonist superfamily [27].

Figure 1: Schematic representation of the suggested Noggin-BMP-7 complex with indicated binding sites of BMP receptors (big circle = BMPRII; small circle = BMPRI). Noggin’s core body (orange) embraces the BMP-7 dimer (grey) allowing its amino-terminal extensions (orange) to snake around it and prevent the surfaces of the growth factor from binding to both BMP type I and type II receptors. Indicated is the suggested clip segment that creates high affinity BMP binding and shielding of the BMP type I receptor interface. Highlighted are Noggin point mutations and deletion of the heparin binding site (ΔB2) in Noggin (A) which possess lower BMP-7 binding capacity, and point mutations in BMP-14 (B) and BMP-7 (C), which are crucial for diminished susceptibility to Noggin inhibition.

(7)

26 Chapter 2. Noggin

Thus far four BMP type I receptors (activin receptor-like kinase 1 (ALK-1), Alk2, Alk3, and Alk6) and three BMP type II receptors (BMPR-II), activin type II receptor (ActR-II and ActR-IIB) have been found to be capable of specifically binding to cer- tain BMPs (Tab. 1) [15]. Binding of Noggin to some of the BMPs inhibits those from binding and therefore activation of BMP receptors, thus blocking Smad-dependent and non-Smad signaling [8]. In vitro experiments have shown that Noggin binds with varying affinities to BMP-2, -4, -5, -6, -7, -13, -14 [28, 22, 19]. Thereby it inhibits BMP- 2, -4, -5, -7, -13 and -14 mediated action, leaving BMP-3, -6, -9, -10 and -15 signaling unaffected (Tab. 1) [4, 22, 19]. Whether BMP binding and inhibition by Noggin does always follow the described BMP-7/Noggin structural mode is up to date uncertain.

Interestingly, binding of Noggin to BMP-6 does not coincide with diminishing BMP-6 activity in osteoblast differentiation in vitro [22].

2.4 Expression, activation and turnover

Noggin is pleiotropic factor which is expressed both early in development as well as in later stages. During early gastrulation Noggin is produced by the Spemann organizer and antagonizes the action of BMP-2 -4, -7, leading to a BMP gradient directed dorsal- ventral patterning with subsequent germ layer formation (Figure 2) [21, 28].

2.4.1 Noggin expression in ectoderm derivatives

The presence of Noggin is essential in developmental structures derived from ec- toderm such as the neural tube, tooth, hair follicle and eye development [12]. Al- though neural tube induction occurs even in the absence of Noggin, it was shown to be crucial for neurogenesis [12, 10]. Noggin is expressed in the notochord and can be augmented upon noradrenalin exposure in ectodermal derivatives [12, 13]. There- upon, overexpression of Noggin counteracts BMP-4 activity on neural precursor cells causing over-proliferation of neural tissue (Figure 2) [2].

Noggin is also expressed in the dermal papilla and connective tissue of the hair follicle where it neutralizes BMP-4 hair follicle induction in embryonic skin organ culture (Figure 2) [3]. Interestingly, ectopic application of Noggin and subsequent BMP-4 inhibition leads to changes in tooth phenotype and to the development of mo- lars instead of incisors [23]. Moreover, a crucial role of Noggin has been implicated in eye development. Keratin 5 promoter-driven overexpression of Noggin in the epi- dermis led to reduced apoptosis and retardation of cell differentiation in the eyelid epithelium [20]. Noggin is furthermore detected in the lens, retina and periocular mesenchyme and rescues ablated epiblast cell induced eye defects [6].

2.4.2 Noggin expression in mesoderm derivatives

Noggin is modestly expressed in mesoderm derived tissue and is required for em- bryonic somites and skeleton patterning (Figure 2) [12]. In the medial somites Nog- gin expression is promoted by Wnt-1, which in turn antagonizes BMP-4 activity and

(8)

2.4. Expression, activation and turnover 27

Figure2:OverviewofNoggin’sfunctionduringgastrulationandtissuedevelopment.HighlightedistheantagonisticroleofNoggin (orange)onindicatedBMPproteinsduringdorsal-ventralpatterningofthegastrulaandgermlayerderivedtissuederivates.Asterisks indicatetissueswhereNogginhasbeendescribedtoplayaroleindevelopmentaland/oradulttissuehomeostasis.

(9)

28 Chapter 2. Noggin

therefore accelerates myogenesis [9]. Noggin is also critical for embryonic chrondro- osteogenesis and joint formation (Figure 2) [7, 24]. Noggin expression in osteoblasts is elevated in the presence of BMP-2, -4, -5, -6, -7[5, 22]. In chrondrocytes it has been described that Indian hedgehog induces Noggin expression [17]. In vivo, overexpress- ing of Noggin in mature mouse osteocalcin-positive osteoblasts revealed a dramatic decrease in bone mineral density and bone formation rate which is thought to be due to impaired osteoblast recruitment and function. Mouse studies have further- more shown that Noggin is expressed postnatally in the mesenchyme of patent, but not fusing, cranial sutures where Noggin transcripts are down-regulated by fibroblast growth factor-2 (FGF-2) and FGF-9. It is speculated that premature cranial suture fu- sion caused by constitutive activation of FGF receptors is the result of inappropriate inhibition of Noggin expression [26].

2.5 Biological Function

With the growing knowledge on BMPs and their respective tissue targets, the under- standing of Noggin’s functionality is also strengthend (Table 1). In Noggin null mice augmented BMP activity evokes a series of developmental abnormalities which in- clude failure of neural tube formation, hair-follicle retardation, dysmorphogenesis of the axial skeleton and joint lesions [12, 24]. Since Noggin null mice are embryonic lethal, the role of Noggin in adult tissue homeostasis is mostly undetermined. How important Noggin is in humans has been highlighted through the discovery of several heterozygous missense mutations of NOG with effect on joint morphogenesis, indicat- ing functional haploinsufficiency [11, 7]. Thereby, increased Noggin activity results in human skeletal dysplasia such as proximal symphalangism (SYM1) and multiple synostosis syndrome 1 (SYNS1). Beyond that, it has recently been shown that Nog- gin in combination with basic fibroblast growth factor (bFGF) is sufficent to maintain prolonged growth of human embryonic stem cells (hES) in vitro [25]. Additionally, Noggin has been reported to antagonize BMP signaling to regulate the stem cell niche during neurogenesis [10]. A novel role of Noggin in osteolytic prostate cancer cells has been recently implicated. Thereby it was found that Noggin expression is re- stricted to cell lines that induce osteolytic bone metastases. Re-expression of Noggin in prostate cancer cells reduced the osteosclerotic capacity and normalized the overall bone structural environment leading to balanced bone remodeling [18].

2.6 Possible Medical and Industrial Applications

2.6.1 Noggin’s affinity to BMPs

Since Groppe and co-workers published the crystal structure of the BMP-7/Noggin- complex, a series of Noggin point mutations was engineered to evaluate their respec- tive binding affinities to BMP-7. Three Noggin mutations (L46D, E48K and I218E) revealed lower BMP-7 binding affinities compared to wild type Noggin, leading vice

(10)

2.6. Possible Medical and Industrial Applications 29

versa to the idea of tailored Noggin-insensitive and thus more potent BMPs (Fig- ure 1) [8]. However, because the binding sites for BMP type I and BMP type II re- ceptors and Noggin overlap on the surface of BMPs, the identification of BMP specific residues involved in binding to Noggin but not to BMP receptors is a critical determi- nant and still not resolved.

2.6.2 Susceptibility of BMPs to Noggin

Recently, two independent groups demonstrated that single amino acid changes in BMPs can determine the susceptibility to Noggin inhibition which provides insight on Noggin’s ligand selectivity. Mutations of BMP-14 at position N64 (N64K and N64T), which are linked to patients with SYM1, protects the ligand from Noggin antago- nism with subsequent elevated cartilage production in an in vivo chicken model (Fig- ure 1) [19]. A comparative analysis of BMP-6 and its close paralogue BMP-7 demon- strated that a single amino acid change at position K60 (BMP-6) to E60 (BMP-7) is mediating increased resistance to Noggin inhibition, thereby explaining its strong osteo-inductive properties [22]. Interestingly, the SYM1 associated mutations result in overactive BMP-14 due to altered BMP type I receptor binding specificity whereas the BMP-7/E60 Noggin complex is suggested to interfere with type II BMP receptor binding (Figure 1) [22, 19]. A systemic analysis of a panel of BMPs revealed that cru- cial amino acids for the determination of Noggin susceptibility are conserved among BMPs which enables a prediction on Noggin’s effect on yet not investigated BMP pro- teins. Both groups show that it is possible to engineer BMP variants, which overcome the Noggin regiment through amino acid substitutions. That allows for development of more effective, second generation BMP proteins (e.g.: BMP-2/P36K; BMP-7/K60E;

BMP- 14/N64K and N64T) with potential clinical applicability in spinal fusion, long bone non-union fracture treatment and osteoarthritis (Figure 1) [22, 19].

2.6.3 Noggin’s bioavailability

Additionally, it has been described that Noggin contains a central, highly basic heparin- binding segment that facilitates binding and storage in the extracellular matrix (ECM) [16]. Thereby, ECM bound Noggin plays an essential role in the formation of lateral BMP gradients, a prerequisite for cell polarization, embryogenesis and tissue pattern- ing (Figure 2). In fact, an area of ongoing research investigates the regulatory ca- pacity of bone matrix changing with environmental entities (e.g.: illness, aging) that interfere with the BMP and Noggin interplay. Recently, Noggin mutants lacking the heparin binding domain (hNogΔB2) where engineered. Thereby, systemic adminis- trated hNogΔB2 gave rise to reduced binding to heparin sulphate proteoglycans and revealed improved bio- availability / -activity, thus representing a potential candidate for gene therapy [16].

Even though the detailed functionality of Noggin is starting to be elucidated, this review indicates that there are still a lot of gaps to fill in order to get a complete view on Noggin’s action. In the future it will be interesting to see whether Noggin can act

(11)

30 Chapter 2. Noggin

independently of BMPs via binding to its own receptor, as it has been recently shown for the binding of gremlin to the VEGF receptor-2 [14]. Thereby the role of Noggin on BMPs in endoderm derivates and the regulation of in vivo adult tissue homeostasis will be particularly interesting.

2.7 Acknowledgments

This work was supported by the German Science Foundation (SFB 760, to P.K.). A.G.

is a member of the Berlin-Brandenburg School for Regenerative Therapies (BSRT;

GSC 230).

2.8 References

[1] O. Avsian-Kretchmer and A. J. Hsueh. Comparative genomic analysis of the eight-membered ring cystine knot-containing bone morphogenetic protein an- tagonists. Mol. Endocrinol., 18:1–12, Jan 2004.

[2] M. A. Bonaguidi, C. Y. Peng, T. McGuire, G. Falciglia, K. T. Gobeske, C. Czeisler, and J. A. Kessler. Noggin expands neural stem cells in the adult hippocampus.

J. Neurosci., 28:9194–9204, Sep 2008.

[3] V. A. Botchkarev, N. V. Botchkareva, W. Roth, M. Nakamura, L. H. Chen, W. Her- zog, G. Lindner, J. A. McMahon, C. Peters, R. Lauster, A. P. McMahon, and R. Paus. Noggin is a mesenchymally derived stimulator of hair-follicle induc- tion. Nat. Cell Biol., 1:158–164, Jul 1999.

[4] L. W. Gamer, J. Nove, M. Levin, and V. Rosen. BMP-3 is a novel inhibitor of both activin and BMP-4 signaling in Xenopus embryos. Dev. Biol., 285:156–168, Sep 2005.

[5] E. Gazzerro, V. Gangji, and E. Canalis. Bone morphogenetic proteins induce the expression of noggin, which limits their activity in cultured rat osteoblasts. J.

Clin. Invest., 102:2106–2114, Dec 1998.

[6] J. Gerhart, J. Pfautz, C. Neely, J. Elder, K. DuPrey, A. S. Menko, K. Knudsen, and M. George-Weinstein. Noggin producing, MyoD-positive cells are crucial for eye development. Dev. Biol., 336:30–41, Dec 2009.

[7] Y. Gong, D. Krakow, J. Marcelino, D. Wilkin, D. Chitayat, R. Babul-Hirji, L. Hud- gins, C. W. Cremers, F. P. Cremers, H. G. Brunner, K. Reinker, D. L. Rimoin, D. H.

Cohn, F. R. Goodman, W. Reardon, M. Patton, C. A. Francomano, and M. L. War- man. Heterozygous mutations in the gene encoding noggin affect human joint morphogenesis. Nat. Genet., 21:302–304, Mar 1999.

[8] J. Groppe, J. Greenwald, E. Wiater, J. Rodriguez-Leon, A. N. Economides, W. Kwiatkowski, M. Affolter, W. W. Vale, J. C. Belmonte, and S. Choe. Structural

(12)

2.8. References 31

basis of BMP signalling inhibition by the cystine knot protein Noggin. Nature, 420:636–642, Dec 2002.

[9] E. Hirsinger, D. Duprez, C. Jouve, P. Malapert, J. Cooke, and O. Pourquie. Noggin acts downstream of Wnt and Sonic Hedgehog to antagonize BMP4 in avian somite patterning. Development, 124:4605–4614, Nov 1997.

[10] D. A. Lim, A. D. Tramontin, J. M. Trevejo, D. G. Herrera, J. M. Garcia-Verdugo, and A. Alvarez-Buylla. Noggin antagonizes BMP signaling to create a niche for adult neurogenesis. Neuron, 28:713–726, Dec 2000.

[11] J. Marcelino, C. M. Sciortino, M. F. Romero, L. M. Ulatowski, R. T. Ballock, A. N.

Economides, P. M. Eimon, R. M. Harland, and M. L. Warman. Human disease- causing NOG missense mutations: effects on noggin secretion, dimer forma- tion, and bone morphogenetic protein binding. Proc. Natl. Acad. Sci. U.S.A., 98:11353–11358, Sep 2001.

[12] J. A. McMahon, S. Takada, L. B. Zimmerman, C. M. Fan, R. M. Harland, and A. P. McMahon. Noggin-mediated antagonism of BMP signaling is required for growth and patterning of the neural tube and somite. Genes Dev., 12:1438–

1452, May 1998.

[13] N. J. Messenger, S. J. Rowe, and A. E. Warner. The neurotransmitter nora- drenaline drives noggin-expressing ectoderm cells to activate N-tubulin and be- come neurons. Dev. Biol., 205:224–232, Jan 1999.

[14] S. Mitola, C. Ravelli, E. Moroni, V. Salvi, D. Leali, K. Ballmer-Hofer, L. Zam- mataro, and M. Presta. Gremlin is a novel agonist of the major proangiogenic receptor VEGFR2. Blood, 116:3677–3680, Nov 2010.

[15] K. Miyazono, Y. Kamiya, and M. Morikawa. Bone morphogenetic protein recep- tors and signal transduction. J. Biochem., 147:35–51, Jan 2010.

[16] S. Paine-Saunders, B. L. Viviano, A. N. Economides, and S. Saunders. Heparan sulfate proteoglycans retain Noggin at the cell surface: a potential mechanism for shaping bone morphogenetic protein gradients. J. Biol. Chem., 277:2089–

2096, Jan 2002.

[17] S. Pathi, J. B. Rutenberg, R. L. Johnson, and A. Vortkamp. Interaction of Ihh and BMP/Noggin signaling during cartilage differentiation. Dev. Biol., 209:239–253, May 1999.

[18] R. Schwaninger, C. A. Rentsch, A. Wetterwald, G. van der Horst, R. L. van Be- zooijen, G. van der Pluijm, C. W. Lowik, K. Ackermann, W. Pyerin, F. C. Hamdy, G. N. Thalmann, and M. G. Cecchini. Lack of noggin expression by cancer cells is a determinant of the osteoblast response in bone metastases. Am. J. Pathol., 170:160–175, Jan 2007.

(13)

32 Chapter 2. Noggin

[19] P. Seemann, A. Brehm, J. Konig, C. Reissner, S. Stricker, P. Kuss, J. Haupt, S. Ren- ninger, J. Nickel, W. Sebald, J. C. Groppe, F. Ploger, J. Pohl, M. Schmidt-von Kegler, M. Walther, I. Gassner, C. Rusu, A. R. Janecke, K. Dathe, and S. Mundlos.

Mutations in GDF5 reveal a key residue mediating BMP inhibition by NOGGIN.

PLoS Genet., 5:e1000747, Nov 2009.

[20] A. A. Sharov, L. Weiner, T. Y. Sharova, F. Siebenhaar, R. Atoyan, A. M. Reginato, C. A. McNamara, K. Funa, B. A. Gilchrest, J. L. Brissette, and V. A. Botchkarev.

Noggin overexpression inhibits eyelid opening by altering epidermal apoptosis and differentiation. EMBO J., 22:2992–3003, Jun 2003.

[21] W. C. Smith and R. M. Harland. Expression cloning of noggin, a new dorsalizing factor localized to the Spemann organizer in Xenopus embryos. Cell, 70:829–

840, Sep 1992.

[22] K. Song, C. Krause, S. Shi, M. Patterson, R. Suto, L. Grgurevic, S. Vukicevic, M. van Dinther, D. Falb, P. Ten Dijke, and M. H. Alaoui-Ismaili. Identification of a key residue mediating bone morphogenetic protein (BMP)-6 resistance to noggin inhibition allows for engineered BMPs with superior agonist activity. J.

Biol. Chem., 285:12169–12180, Apr 2010.

[23] A. S. Tucker, K. L. Matthews, and P. T. Sharpe. Transformation of tooth type induced by inhibition of BMP signaling. Science, 282:1136–1138, Nov 1998.

[24] P. Tylzanowski, L. Mebis, and F. P. Luyten. The Noggin null mouse phenotype is strain dependent and haploinsufficiency leads to skeletal defects. Dev. Dyn., 235:1599–1607, Jun 2006.

[25] G. Wang, H. Zhang, Y. Zhao, J. Li, J. Cai, P. Wang, S. Meng, J. Feng, C. Miao, M. Ding, D. Li, and H. Deng. Noggin and bFGF cooperate to maintain the pluripotency of human embryonic stem cells in the absence of feeder layers.

Biochem. Biophys. Res. Commun., 330:934–942, May 2005.

[26] S. M. Warren, L. J. Brunet, R. M. Harland, A. N. Economides, and M. T. Longaker.

The BMP antagonist noggin regulates cranial suture fusion. Nature, 422:625–

629, Apr 2003.

[27] J. L. Zhang, L. Y. Qiu, A. Kotzsch, S. Weidauer, L. Patterson, M. Hammer- schmidt, W. Sebald, and T. D. Mueller. Crystal structure analysis reveals how the Chordin family member crossveinless 2 blocks BMP-2 receptor binding. Dev.

Cell, 14:739–750, May 2008.

[28] L. B. Zimmerman, J. M. De Jesus-Escobar, and R. M. Harland. The Spemann organizer signal noggin binds and inactivates bone morphogenetic protein 4.

Cell, 86:599–606, Aug 1996.

Referenties

GERELATEERDE DOCUMENTEN

License: Licence agreement concerning inclusion of doctoral thesis in the Institutional Repository of the University of Leiden Downloaded.

II Inhibition of Signaling Cascades in Fibrosis 117 7 Signal transduction cascades controlling fibrosis in Dupuytren’s Disease 121 7.1 Dupuytren’s

Not only does this book discuss the impact of BMP signal modulation on osteoblast differentiation (Part I), it also focuses on BMPs as potential inducers of signaling drifts in

In order to compare the susceptibility of BMP-6 and BMP-7 to inhibition by exogenous Noggin, both BMPs were tested, along with BMP-2 and BMP-4, for their ability to pro- mote

(A) CM from Saos-2 cells expressing either a non-targeting control shRNA or SOST targeting shRNA were transfected with BMP-7 (0.3 μg ) and/or SOST (0.3-1.2 μg ) expression plasmids

Accordingly, we suggest that both Wnt/ β- catenin and BMP-7/Smad signaling can act as pro-survival pathways for os- teoblasts and osteocytes which are both regulated by Sclerostin

Our analysis shows that the ARs constitute a family of GPCRs with an exceptional knowledge base of structural, biochemical, and pharmacological data, which con figures a useful

In line with this hypothesis, we found signi ficantly enhanced ALK1 expression in fibrotic human samples as derived from publically available array data with human liver samples