Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2018 Nov 27;247(1):9–20. doi: 10.1002/path.5170

Bone morphogenetic protein receptor signal transduction in human disease

Maria Catalina Gomez‐Puerto 1, Prasanna Vasudevan Iyengar 1, Amaya García de Vinuesa 1, Peter ten Dijke 1,, Gonzalo Sanchez‐Duffhues 1
PMCID: PMC6587955  PMID: 30246251

Abstract

Bone morphogenetic proteins (BMPs) are secreted cytokines that were initially discovered on the basis of their ability to induce bone. Several decades of research have now established that these proteins function in a large variety of physiopathological processes. There are about 15 BMP family members, which signal via three transmembrane type II receptors and four transmembrane type I receptors. Mechanistically, BMP binding leads to phosphorylation of the type I receptor by the type II receptor. This activated heteromeric complex triggers intracellular signaling that is initiated by phosphorylation of receptor‐regulated SMAD1, 5, and 8 (also termed R‐SMADs). Activated R‐SMADs form heteromeric complexes with SMAD4, which engage in specific transcriptional responses. There is convergence along the signaling pathway and, besides the canonical SMAD pathway, BMP‐receptor activation can also induce non‐SMAD signaling. Each step in the pathway is fine‐tuned by positive and negative regulation and crosstalk with other signaling pathways. For example, ligand bioavailability for the receptor can be regulated by ligand‐binding proteins that sequester the ligand from interacting with receptors. Accessory co‐receptors, also known as BMP type III receptors, lack intrinsic enzymatic activity but enhance BMP signaling by presenting ligands to receptors. In this review, we discuss the role of BMP receptor signaling and how corruption of this pathway contributes to cardiovascular and musculoskeletal diseases and cancer. We describe pharmacological tools to interrogate the function of BMP receptor signaling in specific biological processes and focus on how these agents can be used as drugs to inhibit or activate the function of the receptor, thereby normalizing dysregulated BMP signaling. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Keywords: activin, BMP, TGF‐β, bone, cancer, cardiovascular, hematopoiesis, heterotopic ossification, acromesomelic dysplasia, hemorrhagic telangiectasia, pulmonary arterial hypertension, brachydactyly, fibrodysplasia ossificans progressiva, juvenile polyposis syndrome

Introduction

Bone morphogenetic proteins (BMPs) were initially isolated from demineralized bone matrix through their ability to induce bone at ectopic sites in rodents 1. In the late 1980s, the BMPs were purified to homogeneity and PCR primers were designed from the partial amino acid sequences that led to the cloning of cDNAs encoding several different BMPs 2. BMPs were found to be members of the transforming growth factor‐β (TGF‐β) family, which besides the TGF‐βs, also includes the activins. From the 33 genes encoding members of the TGF‐β family, about 15 structurally related BMPs have been identified 3. The most extensively investigated are BMP2, 4, 6, 7, 9, and 10. Like TGF‐β cytokines, BMPs are multifunctional proteins whose activities are highly dependent on cellular context 4. BMPs have pivotal roles in early and late embryonic development and in maintaining homeostasis in a plethora of tissues and organs.

In common with the TGF‐β cytokines, BMPs induce specific cellular responses through their interaction with type I and type II cell surface receptors, which activates the serine/threonine kinase activity of the receptors 5. Three distinct BMP type II receptors (BMPRIIs) and four BMP type I receptors (BMPRIs), also termed activin receptor‐like kinases (ALKs), have been characterized. Upon BMP‐induced heteromeric complex formation of specific sets of type I and type II receptors, the intracellular domains interact and the type II receptor kinase trans‐phosphorylates the type I receptor, leading to its activation. The extracellular signal is thereby transduced across the membrane and intracellular signaling initiated by phosphorylation of specific intracellular signaling proteins, including the SMAD proteins that are key transcription effectors (Figure 1). Each step of the BMP signaling pathway is controlled by positive and negative modulators allowing for signal integration with other signaling pathways 6.

Figure 1.

PATH-5170-FIG-0001-c

BMPR signaling. BMPs signal via complexes of type I (BMPRIs) and type II (BMPRIIs) transmembrane kinase receptors. The type II receptor, a constitutively active kinase (shown in the figure as phosphorylated), phosphorylates (specified as P) and activates the type I receptor, upon which intracellular canonical signaling is initiated by phosphorylation of receptor‐regulated R‐SMAD1, 5, and 8. Activated R‐SMADs partner with SMAD4 to transcriptionally regulate expression of specific target genes. BMPR activation can also induce non‐SMAD signaling, e.g. by activating p38 and JNK MAP kinases and small GTPases such as Rho and Rac. Each step in the pathway is fine‐tuned by positive and negative regulation. For example, ligand bioavailability for the receptor can be regulated by ligand‐binding proteins, also called antagonists, which sequester the ligand from interacting with receptors. BMP type III receptors or accessory co‐receptors can enhance BMP signaling by presenting ligands to type I and type II receptors.

Here we review the role of BMPR signaling in human diseases. Since the BMPRs share extracellular and intracellular domains that are druggable, we describe specific pharmacological agents that are available to elucidate BMPR functions in specific biological processes, and how they are/may be used to target BMP signaling in a therapeutic context. In particular, we highlight those genetic alterations of BMPRs that have been linked with diseases. We refer the reader to recent reviews for the roles of BMPRs in hematopoiesis and hematological diseases 7, reproductive tract and gynecological pathologies 8, immune regulation, autoimmunity and infection 9, and nervous system and neurodegenerative diseases 10.

BMPR signaling

BMPs are synthesized as large pre‐pro‐precursor proteins. Each unprocessed BMP contains an N‐terminal signal peptide connected to a pro‐domain and a mature bioactive region at the carboxy terminus 3. The pro‐BMP is cleaved by a furin type protease, liberating the mature BMP containing a noncovalently attached pro‐domain. In contrast to TGF‐β (where these pro‐domains confer latency), the pro‐domains of BMPs can usually be displaced easily by receptor binding. BMPs are secreted as homo‐ and heterodimers 3. Recent studies have revealed that heterodimers can have more potent or novel functions compared to their homodimer counterparts 11, 12. In particular, BMP9 and BMP10 can heterodimerize and this heterodimer is responsible for most of the biological BMP activity found in plasma 13. BMPs mostly act in an autocrine or paracrine manner, however, some BMPs, such as BMP6, BMP9, and BMP10 13, 14 circulate in the blood and can thus act at a distance. Many BMP binding factors that interfere with BMPR binding have been identified. Examples are Noggin and Chordin, and DAN and Gremlin, which control local BMP bioavailability 15 (Figure 1).

BMPs bind to selective combinations of BMPRIs and BMPRIIs 3. BMPs are dimers and can interact with heteromeric complexes of two BMPRIIs and two BMPRIs. Whereas BMPR‐II is selective for BMPs, ActRIIA, and ActRIIB can interact with BMPs and activins. There are four BMPRIs: ALK1, ALK2 (also termed ActRI), ALK3 (also termed BMPRIA), and ALK6 (also termed BMPRIB). BMP2 and BMP4 bind more strongly to ALK3 and ALK6, BMP6, and BMP7 bind most strongly to ALK2, and BMP9 and BMP10 are high affinity ligands for ALK1 but can also bind to ALK2 3. Both receptor types share high structural and sequence similarity; they are both single pass transmembrane proteins composed of extracellular domains that are rich in cysteines and intracellular regions which harbor the kinase domain. Type II receptors, and in particular BMPR‐II, have a longer carboxy tail when compared to type I receptors. Type I receptors have a juxtamembrane region that is rich in glycine and serine residues; termed GS‐domain, these are important for activation (see below). Unlike TGF‐β and activin, that do not interact directly with their corresponding type I receptors, BMPs can bind to type I and type II alone, although the binding affinity is greatly enhanced when BMPs interact with both receptors 3. In addition, BMP accessory co‐receptors (also termed BMP type III or auxiliary receptors) have been identified, which lack an intracellular kinase activity but can regulate access of ligands to signaling receptors and/or regulate signaling specificity. Examples include the transmembrane proteins endoglin and betaglycan and GPI‐linked dragon family members (also known as repulsion guidance molecules, RGMs). Co‐receptors can share multiple different ligands, and thereby increase the possibilities for signal integration. Moreover, co‐receptors can be shed from the plasma membrane and thereby regulate BMP bioavailability systemically or exert BMP‐independent functions 16 (Figure 1).

Similarly to other TGF‐β family members, BMPRI is a substrate for BMPRII. BMPRII phosphorylates BMPRI on specific serine and threonine residues in the GS‐domain 3. The binding site of the negative regulator FKBP12 is close to this GS domain and upon binding, FKBP12 shields the serine and threonine residues from being phosphorylated by the type II kinase. Therefore, FKBP12 binding to BMPRI creates a threshold for activation, so that when type I and type II meet at the cell surface in the absence of ligand, an intracellular signal is not immediately initiated 17 (Figure 1). Upon BMPRI activation, this receptor initiates intracellular signaling by activating receptor‐regulated (R) – SMADs, i.e. SMAD1, SMAD5, and SMAD8. This is different from TGF‐β cytokines and activins, which induce the phosphorylation of R‐SMAD2 and SMAD3 in most cells 3. Phospho‐Smad specific antibodies that recognize phospho (p)‐SMAD1/5/8 and p‐SMAD2 have been generated and are often used to measure the degree of receptor activation in cells and tissues 18. Activated R‐SMADs form heteromeric complexes with the common mediator (Co)‐SMAD4 to regulate specific gene transcriptional responses through cooperation with other transcription factors, transcriptional co‐activators and repressors. R‐SMADs and SMAD4 bind to GC‐rich and CGTA containing DNA sequences, respectively. An established target gene for BMPs is ID1. BMP‐SMAD responsive elements were identified in this gene promotor and subsequently cloned in tandem to generate a powerful BMP‐SMAD‐responsive transcription reporter, the so‐called BRE‐luc reporter 19. Besides the canonical SMAD pathway, BMPR activation can also induce non‐SMAD signaling. Non‐SMAD signaling is characterized by intracellular activation of p38 and JNK MAP kinases and small GTPases such as Rho and Rac 20 (Figure 1).

A number of pharmacological agents have been developed to modulate BMPR signaling (Figures 2 and 3). For example, deletion mutants and folding variants of mature BMP4 have been used to prevent the binding of wild type BMP to the receptor while inhibiting ossification 21. BMPs that cannot interact with secreted antagonists have been also generated. This is the case of engineered BMP2 and BMP7 that are resistant to Noggin inhibition 22. Furthermore, neutralizing antibodies that affect ligand–receptor interactions by binding to ligands have been produced 23 (Figure 3). Another strategy has been the development of antibodies that bind to secreted ligand‐binding proteins thereby promoting BMP receptor signaling 24 (Figure 2). Moreover, ecto‐domains of receptors have been used to sequester BMPs thus preventing their binding to their receptors 25. In this way, they function similarly to natural secreted antagonists such as Noggin (Figure 3). Antibodies against the secreted antagonists were found to stimulate BMPR signaling 24 (Figure 2). Furthermore, BMPRs have intrinsic serine/threonine kinase activity, which has allowed the development of small molecular kinase inhibitors that selectively interfere with BMPRI kinase activity 26 (Figure 3). An alternative approach is the use of statins 27 and low intensity pulsed ultrasound 28 that can activate BMPR signaling by promoting BMP expression. Utilization of FK506 to inhibit the interaction of the negative regulator FKBP12 with BMPRI is another strategy 29 (Figure 2). Such modalities, in combination with animal models, have been used to determine the role of BMPR signaling in disease and have been explored as potential therapies for a number of pathologies, including cardiovascular and skeletal diseases, and cancer.

Figure 2.

PATH-5170-FIG-0002-c

BMPR agonists. Several pharmacological agents have been developed to enhance BMPR signaling. Among them, small molecule activators of BMP signaling or BMP mimetics have been used. Furthermore, mutant BMPs that have super agonistic activity by being defective in interacting with secreted antagonists have also been engineered. In addition, neutralizing antibodies have been developed, interfering with secreted ligand‐binding proteins such as Gremlin and thereby promoting BMPR signaling. Utilization of FK506 to inhibit the interaction of the negative regulator FKBP12 with BMPRI is another approach to activate BMPR signaling.

Figure 3.

PATH-5170-FIG-0003-c

BMPR inhibitors. BMPR signaling can be inhibited through various pharmacological agents. One example are mutant BMPs, engineered to bind to but lacking the ability to activate the receptor, thereby blocking the binding of wild type BMPs to the receptor. Neutralizing antibodies that interfere with ligand–receptor interactions by recognizing either ligands or receptors have been also developed. Moreover, ecto‐domains of receptors (ligand traps) are used to sequester BMPs from binding to their receptors; functioning in a way similar to the natural secreted antagonists such as Noggin. Furthermore, small molecular kinase inhibitors have been developed that selectively interfere with BMPRI kinase activity. In addition, miRNAs, siRNAs or exon skipping have also been used as therapeutic tools to inhibit BMPR signaling.

BMPRs in cardiovascular disease

BMPRs are critical regulators of normal cardiovascular structure and function 30. Defects in cardiac development due to BMPR signaling disruption may lead to several congenital heart abnormalities. The cardiac precursor zone in the developing vertebrate embryo is shaped by an interplay between BMP, fibroblast growth factor (FGF) and Wnt signaling 31. This involves the differentiation of pluripotent and embryonic stem cells into the cardiac lineage, where BMPs play an important role. For an extensive review of BMPs and cardiomyocyte differentiation please refer to 32, 33. Septation of the outflow tract and valve maturation are also regulated by BMPs 34. In particular, BMP2 signaling has been identified as an inducer of epithelial‐to‐mesenchymal transition (EMT) in the endocardial cushions (a subset of cells found in the developing heart tube that are indispensable for valve and septa formation) 35, 36. Furthermore, conditional deletion of Bmpr1a (encoding ALK3) in the myocardium results in abnormal atrioventricular (AV) cushion and septal morphogenesis 37. In the developing mouse heart, ALK2 is found to mediate atrioventricular cushion transformation 38. This receptor has been associated with congenital heart defects 39, 40 and abnormal ALK2 signaling leads to the development of a bicuspid aortic valve phenotype 41.

BMPRs are key factors maintaining adult cardiovascular homeostasis and function. Here we first discuss hereditary hemorrhagic telangiectasia (HHT) and pulmonary arterial hypertension (PAH, MIM: 178600), which are two cardiovascular diseases caused by (epi)genetic alterations of BMPRs 30. HHT is a rare autosomal dominant genetic disorder leading to vascular malformations that result in direct connections between arteries and veins. The disease is characterized by mucocutaneous telangiectases and arteriovenous malformations of the gastrointestinal tract, liver, lung, and brain 42, 43. The most prevalent symptom of HHT is nosebleeds, which radically affects the quality of life of the patient. There are five subtypes of HHT: HHT type 1, 2, 3, and 4, and Juvenile Polyposis/HHT. Mutations in ENG (encoding Endoglin), ACVRL1 (encoding ALK1) and SMAD4 cause HHT1 (MIM: 187300), HHT2 (MIM: 600376), and the combined Juvenile Polyposis/HHT (MIM: 175050) syndrome, respectively 44. While mutations in ENG and ACVRL1 represent 80–85% of HHT cases, only 2% of HHT is caused by mutations in SMAD4 42. To date, no causative mutations have been identified in patients with HHT3 (MIM: 601101) and HHT4 (MIM: 610655). GDF2 (encoding BMP9) mutations that negatively affect protein processing and/or function have been associated with a vascular syndrome that phenotypically resembles HHT and is now referred to as HHT5 (MIM: 615506) 45. Homozygous null Eng or Acvrl1 mutations compromise angiogenesis and heart development leading to embryonic lethality in mice 46. Mice with heterozygous inactivating mutations of these genes exhibit particular vascular phenotypes which do not recapitulate the human disease completely. Therefore, conditional knockout mice, which recapitulate the specific arteriovenous malformations observed in patients, represent the most accepted animal models to study HHT 42, 46. Current therapies for HHT rely on inhibiting angiogenesis by means of Bevacizumab (VEGF inhibitor) 47 or by increasing clotting through the use of Thalidomide or antifibrinolytics 48. However, present treatments cause numerous side effects and only provide temporary symptomatic relief. To alleviate these problems, future therapies for HHT may focus on enhancing endoglin and ALK1 stability/expression or function (Table 1).

Table 1.

Examples of diseases related to BMPRs and/or pharmacological agents based on BMPRs

Disease Receptor* (Experimental) Therapy References
Cardiovascular
HHT1, HHT2 ENDOnitric oxide GLIN, ALK1 Tranexamic acid (NCT01031992) 49, 50, 51, 52
PAH BMPRII, ALK1, ALK6 Tacrolimus /FK506 (NCT01647945), BMP9, Ataluren/PTC124, ActRIIA‐IgG1Fc (NCT03496207), Gremlin‐1 ab 24, 29, 53, 54
Aortic valve development ALK2 41
Left–right axis malformations ActRIIB 55
Vascular calcification LDN‐193189, ActRIIA‐Fc, ALK3‐Fc, Tacrolimus/FK506(NCT01612299), Dipyridamole (NCT00767663) 56, 57
Musculoskeletal
Sarcopenia, cachexia, Duchenne muscular dystrophy ActRII‐Fc, ActRII ab, Follistatin (NCT01519349)
Fracture healing OP‐1/BMP7(NCT00679328)
Osteoarthritis OP‐1/BMP7 (NCT01111045)
Muscular dystrophy ActRIIA‐Fc, ActrRIIB‐Fc 58
Fibrodysplasia ossificans progressiva ALK2 LDN‐193189, LDN‐212854/ALK2 kinase inhibitor, activin ab, ActRIIA‐Fc, Dipyridamole 59, 60, 61, 62
Type 2 brachydactyly ALK6
Acromesomelic dysplasia ALK6
Osteoporosis ActRIIA‐Fc, ALK3‐Fc, 63, 64, 65
Cancer
Diffuse intrinsic pontine glioma ALK2 LDN‐212854/ALK2 inhibitor, LDN‐214117, LDN‐193189,TP‐0184, TP‐0184 (NCT03429218, Phase 1 solid tumors) 66, 67, 68
Juvenile polyposis ALK3 69, 70
Pancreatic cancer ActRIIB, ALK6 71
Colon cancer BMPRII, ActRII 72
Tumor angiogenesis ALK1, endoglin PF‐03446962/anti‐hALK1 ab, (NCT01620970, NCT00557856)
ALK1‐Fc/Dalantercept/ACE‐041, Tracon/ TRC105 /endoglin ab (NCT02979899, NCT01381861, NCT02664961, NCT02560779, NCT01564914, NCT01332721, NCT01975519, NCT01806064, NCT02429843)
73, 74
Breast cancer BMPR1A, BMPR2 LDN‐193189 75
Other diseases
Alzheimer disease OP‐1/BMP7(NCT02547818)
Pierre Robin ALK3 76
Juvenile hemochromatosis RGMc/Hemojuvilin (co‐receptor for bone morphogenetic proteins) 77, 78
Anemia, defective erythropoiesis, beta thalassemia ActRIIA‐IgG1Fc (NCT00931606), LDN‐212854 (ALK2 inhibitor), or HJV‐Fc 79, 80
Myelodysplastic syndrome ActRIIA‐IgG1Fc (NCT01736683) 81
Myelofibrosis ActRIIA‐IgG1Fc (NCT01712308) 82
*

Specific BMPR genes that have been found mutated in particular diseases are indicated. For some diseases no BMPR mutations are found.

Therapies or experimental therapies that modulate the receptor expression or BMP signaling are indicated. The ClinicalTrials.gov identifier is also specified.

ActRIIA‐IgG1Fc is also known as sotatercept or ACE‐011.

PAH is a chronic disease characterized by an increase in mean pulmonary arterial pressure (greater than 25 mmHg at rest), pulmonary capillary wedge pressure (15 mmHg) and pulmonary vascular resistance (greater than 3 Wood Units) 83. It is further characterized by increased endothelial cell proliferation and vascular smooth muscle cell hypertrophy resulting in progressive occlusion of the artery lumen. As a consequence, patients develop right ventricular dysfunction resulting in shortness of breath. In the absence of lung transplantation, PAH leads to right heart failure and death. More than 70% of patients with familial PAH and 20% of patients with idiopathic PAH have heterozygous mutations that compromise BMPR2 function. These mutations target sequences that encode the ligand‐binding and kinase domain and the long cytoplasmic tail 84. Patients with PAH and BMPR2 mutations exhibit a more severe disease and are at increased risk of death compared to those without BMPR2 mutations 85. However, the incomplete penetrance of BMPR2 mutations (20–30%) suggests that other genetic and environmental factors contribute to the disease. Infections, toxic exposure, inflammation 86, or alterations in estrogen metabolism 87, 88, have all been described and some were reported to downregulate BMPRII expression 89. Besides mutations in the BMPR2 gene, mutations in genes encoding for other BMP signaling components (such as GDF‐2 90, 91, ACVRL1 92, ENDOGLIN 93, and SMAD8 53, 94) are also associated with PAH development. The BMP/SMAD signaling axis is perturbed in monocrotaline (MCT)‐treated rats 95, one of the most broadly used models to study PAH. The authors showed a decrease in BMPR2, ALK6, and SMADs 4, 5, 6, and 8 expression in the lungs but not the kidneys of MCT‐treated rats 95. Furthermore, Bmpr2 deficient rats generated to study PAH revealed increased endothelial‐to‐mesenchymal transition (EndMT), which was implicated in occlusive vascular remodeling 96. Current PAH therapies target prostacyclin, endothelin, and nitric oxide (NO) pathways, which are involved in vasodilation 97. High‐dose calcium channel blockers, anti‐inflammatory and anti‐proliferative drugs are also being used. Novel strategies to treat PAH patients focus on increasing BMPRII transcription or expression and on blocking BMPRII degradation. Finally, activating BMPRII/SMAD signaling by FK506 98 or enhancing BMP signaling by exogenous recombinant BMP9 54, 99 are interesting approaches that have shown promising results in PAH animal models, but could prove difficult to translate into treatments for patients (Table 1).

Lastly, in addition to the aforementioned vascular diseases that have been linked to genetic mutations in BMPRs, aberrant expression of BMPRs has also been implicated in atherosclerosis 100, vascular calcification, and anemia 101.

BMPRs in musculoskeletal disease

BMPs tightly modulate bone homeostasis by targeting cells with osteogenic and chondrogenic potential, as well as osteoclasts and vascular cells (reviewed in 102). Due to their pleiotropic roles, complete ablation of BMPRs often leads to embryonic lethality. To dissect the specific in vivo roles of these receptors, conditional knock‐outs in defined cell types have been generated, showing that disturbed BMPR expression usually leads to bone and cartilage disorders in animal models, including low 103 and high bone mass 104, 105, heterotopic ossification (HO) 106 and defective cartilage formation 107, 108, 109, 110, 111.

A number of genetic bone disorders in which BMPRs are mutated have been described. One example is Acromesomelic dysplasia, a particular form of dwarfism that affects the bones of the hands and feet (acromelia) as well as the forearms and lower legs (mesomelia). There are several forms of this condition. Acromesomelic dysplasia Grebe type (AMDG, MIM: 200700) 112 and Demirhan type (AMDD, MIM: 609441) 113 are associated with biallelic loss of activity mutations in BMPR1B (encoding the BMP type I receptor ALK6). Brachydactyly type B (BDB1, MIM: 113000) is a very rare skeletal condition characterized by shortening of the middle phalanges and absent or rudimentary terminal phalanges. BDB1 has been associated to inactivating mutations in the ROR2 gene 114, 115. Interestingly, the receptor tyrosine kinase ROR2 has been shown to physically interact with ALK6 in a very specific manner 116, 117. BDB1 shares most of its clinical features with Brachydactyly type A2 (BDA2, MIM: 112600), characterized by shortening of the middle phalanx of the index finger and abnormal second toe development. Additionally, inactivating mutations in the GDF5 and BMP2 genes have been described 118, 119. BMPR1B was found to be mutated in either the kinase domain (I200K) or the GS domain (R486W) 120. Using micromass cell cultures and chick limbs assays, overexpression of either I200K or R486W ALK6 caused reduced cartilage differentiation and bone formation. Interestingly, although both mutant BMPRs retained their usual localization at the membrane, only the mutation in the kinase domain resulted in decreased kinase activity 120.

Fibrodysplasia ossificans progressiva (FOP, MIM: 135100) is the most devastating congenital disorder involving HO, that is, endochondral bone formation at extra‐skeletal sites. FOP is characterized by malformation of the big toes at birth and episodic HO in tendons, fascia, ligaments, and muscle 121, which progressively results in the patients being wheelchair‐bound by the third or fourth decade of life. It is noteworthy that inflammation has been identified as a common trigger of HO in FOP 122. The ACVR1 gene encodes ALK2 and a point mutation in the ACVR1 gene (c.617G>A) leading to illicit activation of ALK2 was identified more than a decade ago in nearly 95% of all studied cases of FOP 123. The mutation, which effects the cytosolic GS domain of the receptor, causes an amino acid substitution R206H that was shown to constitutively activate the receptor by interfering with binding of the negative regulator FKBP12 124 and/or enhancing 125 the response to certain BMP ligands. This led to the development of BMPRI kinase inhibitors as a possible treatment for FOP (Table 1). A recent discovery showed that activin A is capable of inducing ALK2 downstream signaling in FOP mutant cells 59, 126. Although activins are known to interact with ALK2, they do not engage the receptor directly and instead compete with osteogenic BMPs 127 . It is noteworthy that other ACVR1 mutations found in a minority of FOP patients also induce ALK2 downstream signaling in response to activin A 126, 128. How intracellular mutations alter the activation of ALK2 upon extracellular binding of activin A remains to be unveiled. Receptor binding affinity assays showed increased binding of iodinidated activin A to ALK2 R206H in a receptor complex including ActRIIA or ActRIIB 126. This suggests that the mutant ALK2 receptor exhibits a higher affinity for activin A than the wild type receptor. Accordingly, sequestration of circulating activins by an ActRIIa‐Fc ligand trap or a specific activin A antibody prevented HO in an animal model of FOP. This mechanism might specifically target bone progenitor cells in FOP. In this regard, a recent publication has identified one tendon‐derived Scx+ cell population and one muscle resident Mx1+ cell population, which could specifically drive the formation of heterotopic bone either in tendons or in muscle 129.

Myostatin, which like TGF‐β induces R‐SMAD2/3 phosphorylation, is best known for its ability to negatively regulate muscle mass and muscle fiber size 130. This process is counterposed by SMAD1/5 signaling, which induces protein synthesis in muscle (reviewed in 131). Accordingly, intramuscular AAV‐mediated overexpression of a constitutive form of ALK3 resulted in increased fiber size, maximal force and muscle mass 131, whereas overexpression of the BMP inhibitor Noggin, or injection of short hairpin RNAs (shRNAs) targeting SMAD1 or SMAD5, led to reduced myofiber size and muscle atrophy. Another group independently showed that over‐activation of BMP signaling in muscle by ectopic expression of either BMP7 or a constitutively active ALK3 prevented muscular atrophy. Furthermore, the authors showed that BMP‐induced signaling counteracts the histone deacetylase (HDAC)4‐myogenin axis that normally contributes to muscle atrophy in denervated muscles 132. Mice in which Bmpr1a (encoding ALK3) was selectively ablated in Myf5+ or MyoD+ quiescent satellite cells revealed more fat accumulation in muscles when compared to controls 133. The authors demonstrated that ALK3 expression in myo‐endothelial Myf5+ cells is necessary for these cells to support the activity of adipogenic progenitors within the muscle. Taken together, accumulating knowledge suggests that bone and muscle homeostasis is tightly controlled by BMP signaling and aberrant BMPR function may lead to bone and skeletal muscle disorders.

BMPRs in cancer

Several studies have demonstrated a strong link between mutations of certain BMPRs and the progression of specific cancers. Moreover, aberrant expression of these receptors has been correlated with a poorer prognosis for cancer patients. For instance, in Juvenile polyposis syndrome (JPS, MIM: 174900), an autosomal dominant inherited disorder, mutations in BMPR1A have been associated with the development of gastrointestinal cancers 134. A study of several families with JPS identified nonsense inactivating mutations giving rise to ALK3 receptors which lack the intracellular kinase domain 135. Other studies confirmed the overall prevalence of BMPR1A mutations in JPS patients, revealing point mutations as well as large deletions of the gene in approximately 23% of the JPS patients studied 136.

Mutations in ACVR1 have been described in pediatric cases of diffuse intrinsic pontine gliomas (DIPGs) 137, 138. These gain of function mutations are similar to those found in FOP (R206H, R258G/S, G328V/E/R/W, and G356D) 128. DIPG is a highly infiltrative and fatal form of cancer that is usually inoperable due to the location of the tumor. Frequent somatic mutations in histone H3 have been well‐documented in these patients. Moreover, co‐occurrence of ACVR1 mutations have been shown to cause an overall increase in ID1 and ID2 levels. Also, through in vitro experiments, ALK2 mutations have been shown to enhance cell proliferation 139. Surprisingly, although benign osteochondromas have been reported in the majority of FOP patients 140, lack of oncogenic predisposition in FOP patients with identical mutations in ACVR1 suggests that the mutant ACVR1 favors tumor development initiated by Histone H3 disruptions. In sporadic colorectal cancer with a microsatellite unstable phenotype, the BMPR2 3′ untranslated region is frequently mutated 72. This often coincides with mutations in ActRII and TGFBR2 leading to a loss of TGF‐β receptor signaling. In pancreatic cancer, reduced expression of ALK3 was found to be associated with a poor prognosis 141. In such cases, changes in receptor expression could be used as prognostic biomarkers.

Besides these genetic changes, misexpression of BMPRs can also contribute to cancer progression. For example, interference with the function of endoglin and ALK1 co‐receptors, which are expressed on proliferating endothelial cells, has been shown to inhibit tumor angiogenesis. The latter is needed for the growth of tumors beyond a few cm3 in size and also for efficient metastasis. Targeting endoglin with a neutralizing antibody is currently being tested in clinical trials, which are showing variable outcomes (Table 1) 142, 143, 144. The use of Tracon/TRC105 has been tested either alone or in combination with other anti‐angiogenic drugs to inhibit the formation of new blood vessels in solid tumors. Inhibition of ALK1 function using a neutralizing antibody or an ALK1‐Fc ligand trap (dalatracept) resulted in a minor therapeutic response as single agent in phase 2 clinical trials in cancer patients (Table 1) 145, 146, 147.

Conclusions and perspectives

BMPs stimulate multiple signaling pathways in a large variety of cell types via three type II and four type I receptors. The inactivation or over‐activation of these receptors by genetic alterations or misexpression can lead to specific cardiovascular, musculo‐skeletal diseases and cancer: (1) HHT is linked to mutations in the co‐receptor endoglin and ALK1; (2) primary PAH is associated with mutations in BMPR2 or its reduced expression; (3) gain of function mutations in ALK2 result in FOP; and (4) loss of function mutations in ALK3 are associated with JPS.

BMPR agonists or antagonists have been used to investigate the role of BMPR function in various pathophysiological processes. New functions for BMPRs continue to be discovered beyond the role as an inducer of bone. Moreover, BMPR agonists or antagonists can be used as pharmacological agents to treat diseases characterized by changes in BMPR activity. Nevertheless, because of the pleiotropic roles of BMPRs in different cell types and diseases, it is important to consider possible off‐target effects including undesirable actions of receptor complexes or aberrant immune responses. However, it is likely that the advantages of targeting these pathways will outweigh the problems and such strategies might be used to inhibit tumor angiogenesis by antagonizing endoglin or ALK1 function and also to increase muscle mass and alleviate anemia using ActRII‐Fc ligand traps. While potent effects have been reported in animal disease models, translation towards human patients has been disappointing thus far, with no significant effects reported to date 99. An obvious reason for this failure is that animal models do not recapitulate the human disease completely, or that these BMPR targeting agents are only relevant for a particular subset of patients. Another explanation is that the drugs are tested as single agents on patients that have not been carefully screened to determine if they might benefit from the treatment.

The use of CRISPR‐Cas9 may allow the development of better animal models that more closely mimic human diseases. Utilization of patient‐derived models using human (induced pluripotent cell‐derived) heterotypic organoid cell cultures grown in 3D within an appropriate extracellular matrix 148 may enable a more thorough preclinical assessment of the therapeutic potential of BMPR modulators either as single agents or in combination with other molecules that synergistically enhance their activities. Finally, the efficacy of treatments could be enhanced, and off‐target effects reduced, by more precise pharmacological dosing regimens, e.g. by putting patients on so‐called drug holidays 149 and/or by using precision delivery devices 150.

Author contributions statement

MCGP, PVI, PTD, and GSD performed the literature search. AGV designed and generated the figures. MCGP, PVI, PTD, and GSD wrote the manuscript. All authors were involved in the manuscript and all authors approved the final version.

Acknowledgements

We would like to thank Dr. David Baker for valuable comments on the manuscript. Research in our laboratory is supported by Netherlands Cardiovascular Research Initiative: the Dutch Heart Foundation, Dutch Federation of University Medical Centers, the Netherlands Organization for Health Research and Development, and the Royal Netherlands Academy of Sciences (CVON‐PHAEDRA) and Cancer Genomics Centre Netherlands to PTD. GSD is supported by CVON‐RECONNECT. PVI is supported by the European Union Horizon 2020 Research and Innovation Programme with a Marie Skłodowska‐Curie Individual Fellowship (786880).

No conflicts of interest were declared.

References

  • 1. Urist MR. Bone: formation by autoinduction. Science 1965; 150: 893–899. [DOI] [PubMed] [Google Scholar]
  • 2. Wozney JM, Rosen V, Celeste AJ, et al Novel regulators of bone formation: molecular clones and activities. Science 1988; 242: 1528–1534. [DOI] [PubMed] [Google Scholar]
  • 3. Katagiri T, Watabe T. Bone morphogenetic proteins. Cold Spring Harb Perspect Biol 2016; 8: a021899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Morikawa M, Derynck R, Miyazono K. TGF‐β and the TGF‐β family: context‐dependent roles in cell and tissue physiology. Cold Spring Harb Perspect Biol 2016; 8: a021873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Heldin C‐H, Miyazono K, ten Dijke P. TGF‐β signalling from cell membrane to nucleus through SMAD proteins. Nature 1997; 390: 465–471. [DOI] [PubMed] [Google Scholar]
  • 6. Luo K. Signaling cross talk between TGF‐β/Smad and other signaling pathways. Cold Spring Harb Perspect Biol 2017; 9: a022137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Naka K, Hirao A. Regulation of hematopoiesis and hematological disease by TGF‐β family signaling molecules. Cold Spring Harb Perspect Biol 2017; 9: a027987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Monsivais D, Matzuk MM, Pangas SA. The TGF‐β family in the reproductive tract. Cold Spring Harb Perspect Biol 2017; 9: a022251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Sanjabi S, Oh SA, Li MO. Regulation of the immune response by TGF‐β: from conception to autoimmunity and infection. Cold Spring Harb Perspect Biol 2017; 9: a022236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Meyers EA, Kessler JA. TGF‐β family signaling in neural and neuronal differentiation, development, and function. Cold Spring Harb Perspect Biol 2017; 9: a022244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Kaito T, Morimoto T, Mori Y, et al BMP‐2/7 heterodimer strongly induces bone regeneration in the absence of increased soft tissue inflammation. Spine J 2018; 18: 139–146. [DOI] [PubMed] [Google Scholar]
  • 12. Israel DI, Nove J, Kerns KM, et al Heterodimeric bone morphogenetic proteins show enhanced activity in vitro and in vivo. Growth Factors 1996; 13: 291–300. [DOI] [PubMed] [Google Scholar]
  • 13. Tillet E, Ouarné M, Desroches‐Castan A, et al A heterodimer formed by bone morphogenetic protein 9 (BMP9) and BMP10 provides most BMP biological activity in plasma. J Biol Chem 2018; 293: 10963–10974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Jiang H, Salmon RM, Upton PD, et al The prodomain‐bound form of bone morphogenetic protein 10 is biologically active on endothelial cells. J Biol Chem 2016; 291: 2954–2966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Brazil DP, Church RH, Surae S, et al BMP signalling: agony and antagony in the family. Trends Cell Biol 2015; 25: 249–264. [DOI] [PubMed] [Google Scholar]
  • 16. Nickel J, ten Dijke P, Mueller TD. TGF‐β family co‐receptor function and signaling. Acta Biochim Biophys Sin (Shanghai) 2018; 50: 12–36. [DOI] [PubMed] [Google Scholar]
  • 17. Chen YG, Liu F, Massague J. Mechanism of TGF‐β receptor inhibition by FKBP12. EMBO J 1997; 16: 3866–3876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Persson U, Izumi H, Souchelnytskyi S, et al The L45 loop in type I receptors for TGF‐β family members is a critical determinant in specifying Smad isoform activation. FEBS Lett 1998; 434: 83–87. [DOI] [PubMed] [Google Scholar]
  • 19. Korchynskyi O, ten Dijke P. Identification and functional characterization of distinct critically important bone morphogenetic protein‐specific response elements in the Id1 promoter. J Biol Chem 2002; 277: 4883–4891. [DOI] [PubMed] [Google Scholar]
  • 20. Zhang YE. Non‐Smad signaling pathways of the TGF‐β family. Cold Spring Harb Perspect Biol 2017; 9: a022129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Weber FE, Schmökel H, Oelgeschläger M, et al Deletion mutants of BMP folding variants act as BMP antagonists and are efficient inhibitors for heterotopic ossification. J Bone Miner Res 2003; 18: 2142–2151. [DOI] [PubMed] [Google Scholar]
  • 22. Song K, Krause C, Shi S, et al 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 2010; 285: 12169–12180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Calpe S, Correia ACP, Sancho‐Serra MDC, et al Comparison of newly developed anti‐bone morphogenetic protein 4 llama‐derived antibodies with commercially available BMP4 inhibitors. MAbs 2016; 8: 678–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Ciuclan L, Sheppard K, Dong L, et al Treatment with anti‐gremlin 1 antibody ameliorates chronic hypoxia/SU5416‐induced pulmonary arterial hypertension in mice. Am J Pathol 2013; 183: 1461–1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Cunha SI, Pardali E, Thorikay M, et al Genetic and pharmacological targeting of activin receptor‐like kinase 1 impairs tumor growth and angiogenesis. J Exp Med 2010; 207: 85–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Williams E, Bullock AN. Structural basis for the potent and selective binding of LDN‐212854 to the BMP receptor kinase ALK2. Bone 2018; 109: 251–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Kodach LL, Bleuming SA, Peppelenbosch MP, et al The effect of statins in colorectal cancer is mediated through the bone morphogenetic protein pathway. Gastroenterology 2007; 133: 1272–1281. [DOI] [PubMed] [Google Scholar]
  • 28. Huang W, Hasegawa T, Imai Y, et al Low‐intensity pulsed ultrasound enhances bone morphogenetic protein expression of human mandibular fracture haematoma‐derived cells. Int J Oral Maxillofac Surg 2015; 44: 929–935. [DOI] [PubMed] [Google Scholar]
  • 29. Merklinger S, Jones P, Martinez E, et al FK506 activates BMPR2, rescues endothelial dysfunction, and reverses pulmonary hypertension. Circulation 2013; 112: 423–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Goumans M‐J, Zwijsen A, ten Dijke P, et al Bone morphogenetic proteins in vascular homeostasis and disease. Cold Spring Harb Perspect Biol 2018; 10: a031989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Harvey RP. Organogenesis: patterning the vertebrate heart. Nat Rev Genet 2002; 3: 544–556. [DOI] [PubMed] [Google Scholar]
  • 32. Monzen K, Nagai R, Komuro I. A role for bone morphogenetic protein signaling in cardiomyocyte differentiation. Trends Cardiovasc Med 2002; 12: 263–269. [DOI] [PubMed] [Google Scholar]
  • 33. van Wijk B, Moorman AFM, van den Hoff MJB. Role of bone morphogenetic proteins in cardiac differentiation. Cardiovasc Res 2007; 74: 244–255. [DOI] [PubMed] [Google Scholar]
  • 34. Délot EC. Control of endocardial cushion and cardiac valve maturation by BMP signaling pathways. Mol Genet Metab 2003; 80: 27–35. [DOI] [PubMed] [Google Scholar]
  • 35. Sugi Y, Yamamura H, Okagawa H, et al Bone morphogenetic protein‐2 can mediate myocardial regulation of atrioventricular cushion mesenchymal cell formation in mice. Dev Biol 2004; 269: 505–518. [DOI] [PubMed] [Google Scholar]
  • 36. Ma L. Bmp2 is essential for cardiac cushion epithelial‐mesenchymal transition and myocardial patterning. Development 2005; 132: 5601–5611. [DOI] [PubMed] [Google Scholar]
  • 37. Gaussin V, Van de Putte T, Mishina Y, et al Endocardial cushion and myocardial defects after cardiac myocyte‐specific conditional deletion of the bone morphogenetic protein receptor ALK3. Proc Natl Acad Sci U S A 2002; 99: 2878–2883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Wang J, Sridurongrit S, Dudas M, et al Atrioventricular cushion transformation is mediated by ALK2 in the developing mouse heart. Dev Biol 2005; 286: 299–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Smith KA, Joziasse IC, Chocron S, et al Dominant‐negative alk2 allele associates with congenital heart defects. Circulation 2009; 119: 3062–3069. [DOI] [PubMed] [Google Scholar]
  • 40. Joziasse IC, Smith KA, Chocron S, et al ALK2 mutation in a patient with Down's syndrome and a congenital heart defect. Eur J Hum Genet 2011; 19: 389–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Thomas PS, Sridurongrit S, Ruiz‐Lozano P, et al Deficient signaling via Alk2 (Acvr1) leads to bicuspid aortic valve development. PLoS One 2012; 7: e35539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Roman BL, Finegold DN. Genetic and molecular basis for hereditary hemorrhagic telangiectasia. Curr Genet Med Rep 2015; 3: 35–47. [Google Scholar]
  • 43. Ruiz‐Llorente L, Gallardo‐Vara E, Rossi E, et al Endoglin and alk1 as therapeutic targets for hereditary hemorrhagic telangiectasia. Expert Opin Ther Targets 2017; 21: 933–947. [DOI] [PubMed] [Google Scholar]
  • 44. Morrell NW, Bloch DB, ten Dijke P, et al Targeting BMP signalling in cardiovascular disease and anaemia. Nat Rev Cardiol 2015; 13: 106–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Wooderchak‐Donahue WL, McDonald J, O'Fallon B, et al BMP9 mutations cause a vascular‐anomaly syndrome with phenotypic overlap with hereditary hemorrhagic telangiectasia. Am J Hum Genet 2013; 93: 530–537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Tual‐Chalot S, Oh SP, Arthur HM. Mouse models of hereditary hemorrhagic telangiectasia: recent advances and future challenges. Front Genet 2015; 6: 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Grigg C, Anderson D, Earnshaw J. Diagnosis and treatment of hereditary hemorrhagic telangiectasia. Ochsner J 2017; 17: 157–161. [PMC free article] [PubMed] [Google Scholar]
  • 48. Kumar N, Garg N, Khunger M, et al Optimal management of hereditary hemorrhagic telangiectasia. J Blood Med 2014; 5: 191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Fernandez‐L A, Garrido‐Martin EM, Sanz‐Rodriguez F, et al Therapeutic action of tranexamic acid in hereditary haemorrhagic telangiectasia (HHT): regulation of ALK‐1/endoglin pathway in endothelial cells. Thromb Haemost 2007; 97: 254–262. [PubMed] [Google Scholar]
  • 50. Geisthoff UW, Seyfert UT, Kübler M, et al Treatment of epistaxis in hereditary hemorrhagic telangiectasia with tranexamic acid – a double‐blind placebo‐controlled cross‐over phase IIIB study. Thromb Res 2014; 134: 565–571. [DOI] [PubMed] [Google Scholar]
  • 51. Lebrin F, Srun S, Raymond K, et al Thalidomide stimulates vessel maturation and reduces epistaxis in individuals with hereditary hemorrhagic telangiectasia. Nat Med 2010; 16: 420–428. [DOI] [PubMed] [Google Scholar]
  • 52. Dupuis‐Girod S, Ambrun A, Decullier E, et al Effect of bevacizumab nasal spray on epistaxis duration in hereditary hemorrhagic telangectasia. JAMA 2016; 316: 934–942. [DOI] [PubMed] [Google Scholar]
  • 53. Drake KM, Dunmore BJ, McNelly LN, et al Correction of nonsense BMPR2 and SMAD9 mutations by ataluren in pulmonary arterial hypertension. Am J Respir Cell Mol Biol 2013; 49: 403–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Long L, Ormiston ML, Yang X, et al Selective enhancement of endothelial BMPR‐II with BMP9 reverses pulmonary arterial hypertension. Nat Med 2015; 21: 777–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Kosaki R, Gebbia M, Kosaki K, et al Left‐right axis malformations associated with mutations in ACVR2B, the gene for human activin receptor type IIB. Am J Med Genet 1999; 82: 70–76. [DOI] [PubMed] [Google Scholar]
  • 56. Agapova OA, Fang Y, Sugatani T, et al Ligand trap for the activin type IIA receptor protects against vascular disease and renal fibrosis in mice with chronic kidney disease. Kidney Int 2016; 89: 1231–1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Derwall M, Malhotra R, Lai CS, et al Inhibition of bone morphogenetic protein signaling reduces vascular calcification and atherosclerosis. Arterioscler Thromb Vasc Biol 2012; 32: 613–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Morvan F, Rondeau J‐M, Zou C, et al Blockade of activin type II receptors with a dual anti‐ActRIIA/IIB antibody is critical to promote maximal skeletal muscle hypertrophy. Proc Natl Acad Sci U S A 2017; 114: 12448–12453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Hatsell SJ, Idone V, Wolken DMA, et al ACVR1R206H receptor mutation causes fibrodysplasia ossificans progressiva by imparting responsiveness to activin A. Sci Transl Med 2015; 7: 303ra137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Cappato S, Tonachini L, Giacopelli F, et al High‐throughput screening for modulators of ACVR1 transcription: discovery of potential therapeutics for fibrodysplasia ossificans progressiva. Dis Model Mech 2016; 9: 685–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Mohedas AH, Xing X, Armstrong KA, et al Development of an ALK2‐biased BMP type I receptor kinase inhibitor. ACS Chem Biol 2013; 8: 1291–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Yu PB, Deng DY, Lai CS, et al BMP type I receptor inhibition reduces heterotopic [corrected] ossification. Nat Med 2008; 14: 1363–1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Fajardo RJ, Manoharan RK, Pearsall RS, et al Treatment with a soluble receptor for activin improves bone mass and structure in the axial and appendicular skeleton of female cynomolgus macaques (Macaca fascicularis). Bone 2010; 46: 64–71. [DOI] [PubMed] [Google Scholar]
  • 64. Lotinun S, Pearsall RS, Horne WC, et al Activin receptor signaling: a potential therapeutic target for osteoporosis. Curr Mol Pharmacol 2012; 5: 195–204. [DOI] [PubMed] [Google Scholar]
  • 65. Ruckle J, Jacobs M, Kramer W, et al Single‐dose, randomized, double‐blind, placebo‐controlled study of ACE‐011 (ActRIIA‐IgGl) in postmenopausal women. J Bone Miner Res 2009; 24: 744–752. [DOI] [PubMed] [Google Scholar]
  • 66. Taylor KR, Vinci M, Bullock AN, et al ACVR1 mutations in DIPG: lessons learned from FOP. Cancer Res 2014; 74: 4565–4570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Mohedas AH, Wang Y, Sanvitale CE, et al Structure–activity relationship of 3,5‐diaryl‐2‐aminopyridine ALK2 inhibitors reveals unaltered binding affinity for fibrodysplasia ossificans progressiva causing mutants. J Med Chem 2014; 57: 7900–7915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Carvalho D, Taylor K, Olaciregui NG, et al Preclinical efficacy of alk2 inhibitors in ACVR1 mutant DIPG. Neuro Oncol 2016; 18(Suppl 3): hg‐67. [Google Scholar]
  • 69. Howe JR, Sayed MG, Ahmed AF, et al The prevalence of MADH4 and BMPR1A mutations in juvenile polyposis and absence of BMPR2, BMPR1B, and ACVR1 mutations. J Med Genet 2004; 41: 484–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Blatter RHE, Plasilova M, Wenzel F, et al Somatic alterations in juvenile polyps from BMPR1A and SMAD4 mutation carriers. Genes Chromosomes Cancer 2015; 54: 575–582. [DOI] [PubMed] [Google Scholar]
  • 71. Togashi Y, Sakamoto H, Hayashi H, et al Homozygous deletion of the activin a receptor, type IB gene is associated with an aggressive cancer phenotype in pancreatic cancer. Mol Cancer 2014; 13: 126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Kodach LL, Wiercinska E, de Miranda NFCC, et al The bone morphogenetic protein pathway is inactivated in the majority of sporadic colorectal cancers. Gastroenterology 2008; 134: 1332–1341. [DOI] [PubMed] [Google Scholar]
  • 73. de Vinuesa AG, Bocci M, Pietras K, et al Targeting tumour vasculature by inhibiting activin receptor‐like kinase (ALK)1 function. Biochem Soc Trans 2016; 44: 1142–1149. [DOI] [PubMed] [Google Scholar]
  • 74. Hu‐Lowe DD, Chen E, Zhang L, et al Targeting activin receptor‐like kinase 1 inhibits angiogenesis and tumorigenesis through a mechanism of action complementary to anti‐VEGF therapies. Cancer Res 2011; 71: 1362–1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Owens P, Pickup MW, Novitskiy SV, et al Inhibition of BMP signaling suppresses metastasis in mammary cancer. Oncogene 2015; 34: 2437–2449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Yang Y, Yuan J, Yao X, et al BMPR1B mutation causes Pierre Robin sequence. Oncotarget 2017; 8: 25864–25871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Lanzara C, Roetto A, Daraio F, et al Spectrum of hemojuvelin gene mutations in 1q‐linked juvenile hemochromatosis. Blood 2004; 103: 4317–4321. [DOI] [PubMed] [Google Scholar]
  • 78. Andriopoulos B Jr, Corradini E, Xia Y, et al BMP6 is a key endogenous regulator of hepcidin expression and iron metabolism. Nat Genet 2009; 41: 482–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Walpurgis K, Thomas A, Vogel M, et al Testing for the erythropoiesis‐stimulating agent Sotatercept/ACE‐011 (ActRIIA‐Fc) in serum by means of Western blotting and LC‐HRMS. Drug Test Anal 2016; 8: 1152–1161. [DOI] [PubMed] [Google Scholar]
  • 80. Dussiot M, Maciel TT, Fricot A, et al An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β‐thalassemia. Nat Med 2014; 20: 398–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Komrokji R, Garcia‐Manero G, Ades L, et al Sotatercept with long‐term extension for the treatment of anaemia in patients with lower‐risk myelodysplastic syndromes: a phase 2, dose‐ranging trial. Lancet Haematol 2018; 5: e63–e72. [DOI] [PubMed] [Google Scholar]
  • 82. Bose P, Verstovsek S. Developmental therapeutics in myeloproliferative neoplasms. Clin Lymphoma Myeloma Leuk 2017; 17: S43–S52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Hoeper MM, Bogaard HJ, Condliffe R, et al Definitions and diagnosis of pulmonary hypertension. J Am Coll Cardiol 2013; 62: D42–D50. [DOI] [PubMed] [Google Scholar]
  • 84. Machado RD, Southgate L, Eichstaedt CA, et al Pulmonary arterial hypertension: a current perspective on established and emerging molecular genetic defects. Hum Mutat 2015; 36: 1113–1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Evans JDW, Girerd B, Montani D, et al BMPR2 mutations and survival in pulmonary arterial hypertension: an individual participant data meta‐analysis. Lancet Respir Med 2016; 4: 129–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Song Y, Coleman L, Shi J, et al Inflammation, endothelial injury, and persistent pulmonary hypertension in heterozygous BMPR2‐mutant mice. Am J Physiol Heart Circ Physiol 2008; 295: H677–H690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Austin ED, Cogan JD, West JD, et al Alterations in oestrogen metabolism: implications for higher penetrance of familial pulmonary arterial hypertension in females. Eur Respir J 2009; 34: 1093–1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Mair KM, Yang XD, Long L, et al Sex affects bone morphogenetic protein type II receptor signaling in pulmonary artery smooth muscle cells. Am J Respir Crit Care Med 2015; 191: 693–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Orriols M, Gomez‐Puerto MC, Ten Dijke P. BMP type II receptor as a therapeutic target in pulmonary arterial hypertension. Cell Mol Life Sci 2017; 74: 1979–2995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Wang G, Fan R, Ji R, et al Novel homozygous BMP9 nonsense mutation causes pulmonary arterial hypertension: a case report. BMC Pulm Med 2016; 16: 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Gräf S, Haimel M, Bleda M, et al Identification of rare sequence variation underlying heritable pulmonary arterial hypertension. Nat Commun 2018; 9: 1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Girerd B, Montani D, Coulet F, et al Clinical outcomes of pulmonary arterial hypertension in patients carrying an ACVRL1 (ALK1) mutation. Am J Respir Crit Care Med 2010; 181: 851–861. [DOI] [PubMed] [Google Scholar]
  • 93. Pousada G, Baloira A, Fontán D, et al Mutational and clinical analysis of the ENG gene in patients with pulmonary arterial hypertension. BMC Genet 2016; 17: 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Shintani M, Yagi H, Nakayama T, et al A new nonsense mutation of SMAD8 associated with pulmonary arterial hypertension. J Med Genet 2009; 46: 331–337. [DOI] [PubMed] [Google Scholar]
  • 95. Morty RE, Nejman B, Kwapiszewska G, et al Dysregulated bone morphogenetic protein signaling in monocrotaline‐induced pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol 2007; 27: 1072–1078. [DOI] [PubMed] [Google Scholar]
  • 96. Ranchoux B, Antigny F, Rucker‐Martin C, et al Endothelial‐to‐mesenchymal transition in pulmonary hypertension. Circulation 2015; 131: 1006–1018. [DOI] [PubMed] [Google Scholar]
  • 97. Thenappan T, Ormiston ML, Ryan JJ, et al Pulmonary arterial hypertension: pathogenesis and clinical management. BMJ 2018; 360: j5492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Spiekerkoetter E, Tian X, Cai J, et al FK506 activates BMPR2, rescues endothelial dysfunction, and reverses pulmonary hypertension. J Clin Invest 2013; 123: 3600–3613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Spiekerkoetter E, Sung YK, Sudheendra D, et al Randomised placebo‐controlled safety and tolerability trial of FK506 (tacrolimus) for pulmonary arterial hypertension. Eur Respir J 2017; 50: 1602449. [DOI] [PubMed] [Google Scholar]
  • 100. Simões Sato AY, Bub GL, Campos AH. BMP‐2 and ‐4 produced by vascular smooth muscle cells from atherosclerotic lesions induce monocyte chemotaxis through direct BMPRII activation. Atherosclerosis 2014; 235: 45–55. [DOI] [PubMed] [Google Scholar]
  • 101. Pagani A, Colucci S, Bocciardi R, et al A new form of IRIDA due to combined heterozygous mutations of TMPRSS6 and ACVR1A encoding the BMP receptor ALK2. Blood 2017; 129: 3392–3395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Sánchez‐Duffhues G, Hiepen C, Knaus P, et al Bone morphogenetic protein signaling in bone homeostasis. Bone 2015; 80: 43–59. [DOI] [PubMed] [Google Scholar]
  • 103. Mishina Y, Starbuck MW, Gentile MA, et al Bone morphogenetic protein type IA receptor signaling regulates postnatal osteoblast function and bone remodeling. J Biol Chem 2004; 279: 27560–27566. [DOI] [PubMed] [Google Scholar]
  • 104. Kamiya N, Ye L, Kobayashi T, et al Disruption of BMP signaling in osteoblasts through type IA receptor (BMPRIA) increases bone mass. J Bone Miner Res 2008; 23: 2007–2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Lowery JW, Intini G, Gamer L, et al Loss of BMPR2 leads to high bone mass due to increased osteoblast activity. J Cell Sci 2015; 128: 1308–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Chakkalakal SA, Zhang D, Culbert AL, et al An Acvr1 R206H knock‐in mouse has fibrodysplasia ossificans progressiva. J Bone Miner Res 2012; 27: 1746–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Rountree RB, Schoor M, Chen H, et al BMP receptor signaling is required for postnatal maintenance of articular cartilage. PLoS Biol 2004; 2: e355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Jing J, Ren Y, Zong Z, et al BMP receptor 1A determines the cell fate of the postnatal growth plate. Int J Biol Sci 2013; 9: 895–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Baur ST, Mai JJ, Dymecki SM. Combinatorial signaling through BMP receptor IB and GDF5: shaping of the distal mouse limb and the genetics of distal limb diversity. Development 2000; 127: 605–619. [DOI] [PubMed] [Google Scholar]
  • 110. Yi SE, Daluiski A, Pederson R, et al The type I BMP receptor BMPRIB is required for chondrogenesis in the mouse limb. Development 2000; 127: 621–630. [DOI] [PubMed] [Google Scholar]
  • 111. Rigueur D, Brugger S, Anbarchian T, et al The type I BMP receptor ACVR1/ALK2 is required for chondrogenesis during development. J Bone Miner Res 2015; 30: 733–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Graul‐Neumann LM, Deichsel A, Wille U, et al Homozygous missense and nonsense mutations in BMPR1B cause acromesomelic chondrodysplasia‐type Grebe. Eur J Hum Genet 2014; 22: 726–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Demirhan O, Türkmen S, Schwabe GC, et al A homozygous BMPR1B mutation causes a new subtype of acromesomelic chondrodysplasia with genital anomalies. J Med Genet 2005; 42: 314–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Oldridge M, Fortuna A, Maringa M, et al Dominant mutations in ROR2, encoding an orphan receptor tyrosine kinase, cause brachydactyly type B. Nat Genet 2000; 24: 275–278. [DOI] [PubMed] [Google Scholar]
  • 115. Schwabe GC, Tinschert S, Buschow C, et al Distinct mutations in the receptor tyrosine kinase gene ROR2 cause brachydactyly type B. Am J Hum Genet 2000; 67: 822–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Sammar M, Stricker S, Schwabe GC, et al Modulation of GDF5/BRI‐b signalling through interaction with the tyrosine kinase receptor Ror2. Genes Cells 2004; 9: 1227–1238. [DOI] [PubMed] [Google Scholar]
  • 117. Sammar M, Sieber C, Knaus P. Biochemical and functional characterization of the Ror2/BRIb receptor complex. Biochem Biophys Res Commun 2009; 381: 1–6. [DOI] [PubMed] [Google Scholar]
  • 118. Seemann P, Schwappacher R, Kjaer KW, et al Activating and deactivating mutations in the receptor interaction site of GDF5 cause symphalangism or brachydactyly type A2. J Clin Invest 2005; 115: 2373–2381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Dathe K, Kjaer KW, Brehm A, et al Duplications involving a conserved regulatory element downstream of BMP2 are associated with brachydactyly type A2. Am J Hum Genet 2009; 84: 483–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Lehmann K, Seemann P, Stricker S, et al Mutations in bone morphogenetic protein receptor 1B cause brachydactyly type A2. Proc Natl Acad Sci U S A 2003; 100: 12277–12282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Shore EM, Kaplan FS. Insights from a rare genetic disorder of extra‐skeletal bone formation, fibrodysplasia ossificans progressiva (FOP). Bone 2008; 43: 427–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Glaser DL, Kaplan FS. Treatment considerations for the management of fibrodysplasia ossificans progressiva. Clin Rev Bone Miner Metab 2005; 3: 243–250. [Google Scholar]
  • 123. Shore EM, Xu M, Feldman GJ, et al A recurrent mutation in the BMP type I receptor ACVR1 causes inherited and sporadic fibrodysplasia ossificans progressiva. Nat Genet 2006; 38: 525–527. [DOI] [PubMed] [Google Scholar]
  • 124. van Dinther M, Visser N, de Gorter DJJ, et al ALK2 R206H mutation linked to fibrodysplasia ossificans progressiva confers constitutive activity to the BMP type I receptor and sensitizes mesenchymal cells to BMP‐induced osteoblast differentiation and bone formation. J Bone Miner Res 2010; 25: 1208–1215. [DOI] [PubMed] [Google Scholar]
  • 125. Billings PC, Fiori JL, Bentwood JL, et al Dysregulated BMP signaling and enhanced osteogenic differentiation of connective tissue progenitor cells from patients with fibrodysplasia ossificans progressiva (FOP). J Bone Miner Res 2008; 23: 305–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Hino K, Ikeya M, Horigome K, et al Neofunction of ACVR1 in fibrodysplasia ossificans progressiva. Proc Natl Acad Sci U S A 2015; 112: 15438–15443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. ten Dijke P, Yamashita H, Ichijo H, et al Characterization of type I receptors for transforming growth factor‐β and activin. Science 1994; 264: 101–104. [DOI] [PubMed] [Google Scholar]
  • 128. Haupt J, Xu M, Shore EM. Variable signaling activity by FOP ACVR1 mutations. Bone 2018; 109: 232–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Dey D, Bagarova J, Hatsell SJ, et al Two tissue‐resident progenitor lineages drive distinct phenotypes of heterotopic ossification. Sci Transl Med 2016; 8: 366ra163–366ra163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. McPherron AC, Lawler AM, Lee S‐J. Regulation of skeletal muscle mass in mice by a new TGF‐β superfamily member. Nature 1997; 387: 83–90. [DOI] [PubMed] [Google Scholar]
  • 131. Sartori R, Gregorevic P, Sandri M. TGFβ and BMP signaling in skeletal muscle: potential significance for muscle‐related disease. Trends Endocrinol Metab 2014; 25: 464–471. [DOI] [PubMed] [Google Scholar]
  • 132. Winbanks CE, Chen JL, Qian H, et al The bone morphogenetic protein axis is a positive regulator of skeletal muscle mass. J Cell Biol 2013; 203: 345–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Huang P, Schulz TJ, Beauvais A, et al Intramuscular adipogenesis is inhibited by myo‐endothelial progenitors with functioning Bmpr1a signalling. Nat Commun 2014; 5: 4063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Brosens LAA, van Hattem A, Hylind LM, et al Risk of colorectal cancer in juvenile polyposis. Gut 2007; 56: 965–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Howe JR, Bair JL, Sayed MG, et al Germline mutations of the gene encoding bone morphogenetic protein receptor 1A in juvenile polyposis. Nat Genet 2001; 28: 184–187. [DOI] [PubMed] [Google Scholar]
  • 136. Calva‐Cerqueira D, Chinnathambi S, Pechman B, et al The rate of germline mutations and large deletions of SMAD4 and BMPR1A in juvenile polyposis. Clin Genet 2009; 75: 79–85. [DOI] [PubMed] [Google Scholar]
  • 137. Zadeh G, Aldape K. ACVR1 mutations and the genomic landscape of pediatric diffuse glioma. Nat Genet 2014; 46: 421–422. [DOI] [PubMed] [Google Scholar]
  • 138. Taylor KR, Mackay A, Truffaux N, et al Recurrent activating ACVR1 mutations in diffuse intrinsic pontine glioma. Nat Genet 2014; 46: 457–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Buczkowicz P, Hoeman C, Rakopoulos P, et al Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nat Genet 2014; 46: 451–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Morales‐Piga A, Bachiller‐Corral J, González‐Herranz P, et al Osteochondromas in fibrodysplasia ossificans progressiva: a widespread trait with a streaking but overlooked appearance when arising at femoral bone end. Rheumatol Int 2015; 35: 1759–1767. [DOI] [PubMed] [Google Scholar]
  • 141. Voorneveld PW, Stache V, Jacobs RJ, et al Reduced expression of bone morphogenetic protein receptor IA in pancreatic cancer is associated with a poor prognosis. Br J Cancer 2013; 109: 1805–1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Duffy AG, Ulahannan SV, Cao L, et al A phase II study of TRC105 in patients with hepatocellular carcinoma who have progressed on sorafenib. United European Gastroenterol J 2015; 3: 453–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Apolo AB, Karzai FH, Trepel JB, et al A phase II clinical trial of TRC105 (anti‐endoglin antibody) in adults with advanced/metastatic urothelial carcinoma. Clin Genitourin Cancer 2017; 15: 77–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Dorff TB, Longmate JA, Pal SK, et al Bevacizumab alone or in combination with TRC105 for patients with refractory metastatic renal cell cancer. Cancer 2017; 123: 4566–4573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Jimeno A, Posner MR, Wirth LJ, et al A phase 2 study of dalantercept, an activin receptor‐like kinase‐1 ligand trap, in patients with recurrent or metastatic squamous cell carcinoma of the head and neck. Cancer 2016; 122: 3641–3649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Makker V, Filiaci VL, Chen LM, et al Phase II evaluation of dalantercept, a soluble recombinant activin receptor‐like kinase 1 (ALK1) receptor fusion protein, for the treatment of recurrent or persistent endometrial cancer: an NRG Oncology/Gynecologic Oncology Group Study 0229N. Gynecol Oncol 2015; 138: 24–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Necchi A, Giannatempo P, Mariani L, et al PF‐03446962, a fully‐human monoclonal antibody against transforming growth‐factor β (TGFβ) receptor ALK1, in pre‐treated patients with urothelial cancer: an open label, single‐group, phase 2 trial. Invest New Drugs 2014; 32: 555–560. [DOI] [PubMed] [Google Scholar]
  • 148. Dutta D, Heo I, Clevers H. Disease modeling in stem cell‐derived 3D organoid systems. Trends Mol Med 2017; 23: 393–410. [DOI] [PubMed] [Google Scholar]
  • 149. Kovacs RJ, Maldonado G, Azaro A, et al Cardiac safety of TGF‐β receptor I kinase inhibitor LY2157299 monohydrate in cancer patients in a first‐in‐human dose study. Cardiovasc Toxicol 2015; 15: 309–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Bobo D, Robinson KJ, Islam J, et al Nanoparticle‐based medicines: a review of FDA‐approved materials and clinical trials to date. Pharm Res 2016; 33: 2373–2387. [DOI] [PubMed] [Google Scholar]

Articles from The Journal of Pathology are provided here courtesy of Wiley

RESOURCES