Next Article in Journal
Metabolic, Physiological, and Transcriptomics Analysis of Batch Cultures of the Green Microalga Chlamydomonas Grown on Different Acetate Concentrations
Next Article in Special Issue
TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis—Updated 2019
Previous Article in Journal
Germinal Centre B Cell Functions and Lymphomagenesis: Circuits Involving MYC and MicroRNAs
Previous Article in Special Issue
Is NO the Answer? The Nitric Oxide Pathway Can Support Bone Morphogenetic Protein 2 Mediated Signaling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

ACVR1 Function in Health and Disease

by
José Antonio Valer
,
Cristina Sánchez-de-Diego
,
Carolina Pimenta-Lopes
,
Jose Luis Rosa
and
Francesc Ventura
*
Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, 08907 Barcelona, Spain
*
Author to whom correspondence should be addressed.
Cells 2019, 8(11), 1366; https://doi.org/10.3390/cells8111366
Submission received: 1 October 2019 / Revised: 28 October 2019 / Accepted: 30 October 2019 / Published: 31 October 2019
(This article belongs to the Special Issue TGF-beta/BMP Signaling Pathway)

Abstract

:
Activin A receptor type I (ACVR1) encodes for a bone morphogenetic protein type I receptor of the TGFβ receptor superfamily. It is involved in a wide variety of biological processes, including bone, heart, cartilage, nervous, and reproductive system development and regulation. Moreover, ACVR1 has been extensively studied for its causal role in fibrodysplasia ossificans progressiva (FOP), a rare genetic disorder characterised by progressive heterotopic ossification. ACVR1 is linked to different pathologies, including cardiac malformations and alterations in the reproductive system. More recently, ACVR1 has been experimentally validated as a cancer driver gene in diffuse intrinsic pontine glioma (DIPG), a malignant childhood brainstem glioma, and its function is being studied in other cancer types. Here, we review ACVR1 receptor function and signalling in physiological and pathological processes and its regulation according to cell type and mutational status. Learning from different functions and alterations linked to ACVR1 is a key step in the development of interdisciplinary research towards the identification of novel treatments for these pathologies.

Graphical Abstract

1. ACVR1 Gene and Signalling

The human activin A receptor type I (ACVR1) gene (Ensembl: ENSG00000115170), also known as ALK2, is located in chromosome 2q23-q24 [1] and encodes for the 509 amino acid protein (UniProtKB: Q04771). The ACVR1 protein product was initially described as an activin type I receptor [2], and it was found to be expressed in several tissues and different human cell lines [3]. Recent analysis using RNA-sequencing (RNA-Seq) showed that the ACVR1 gene is ubiquitously expressed in healthy human tissues, with varying expression levels (GTEx Portal) [4].
As a member of the BMP/TGFβ receptor family, the ACVR1 protein contains an extracellular N-terminal ligand-binding domain, a transmembrane (TM) domain, an intracellular glycine–serine-rich (GS) domain, and a protein kinase (PK) domain [5,6]. The loop positioned in the helix–loop–helix of the GS domain contains the key residues responsible for ACVR1 activation upon phosphorylation [5]. As a type I receptor, ACVR1 forms heterotetrameric receptor complexes with the type II receptors BMPR2, ACVR2A, and ACVR2B [7]. Such complexes consist of two type I and two type II receptors [8,9]. Upon binding of ligands to the heteromeric complexes, type II receptors transphosphorylate the GS domain of type I receptors. As a result, the kinase domain of type I receptors is activated and subsequently phosphorylates SMAD1/5/8 proteins that transduce the signal [9]. ACVR1 was first described to bind to activin A, a member of the BMP/TGFβ family that usually triggers phosphorylation and activation of SMAD2/3 upon complex formation with type II receptors [2]. Later, ACVR1 was also found to bind several BMPs with distinct affinities, triggering SMAD1/5/8 signalling [10]. Besides canonical SMAD signalling, ACVR1 can activate non-canonical signalling pathways [7,11]. Additional interactions and transduction mechanisms are detailed in each specific section, including activin A signalling via ACVR1(R206H)-ACVR2A/B (detailed in the Fibrodysplasia ossificans progressiva section) and anti-Müllerian hormone (AMH) signalling via ACVR1-AMHR2 (detailed in the Reproductive system section).

2. Fibrodysplasia Ossificans Progressiva

2.1. FOP Clinical Information

Fibrodysplasia ossificans progressiva (FOP) (OMIM#135100) is a rare genetic autosomal dominant musculoskeletal disease. It is characterised by episodic formation of endochondral heterotopic ossifications (HO) within soft tissues, including fascia, ligaments, tendons, and skeletal muscles [12,13]. The incidence of this rare condition is approximately one patient in 1.6 million, irrespective of sex or ethnical groups [14,15,16].
Episodes of heterotopic ossification can be induced by trauma or they can arise spontaneously. The most common initial symptoms of flare-ups include swelling, pain, decreased movement, stiffness, warmth, and redness. FOP flare-ups usually follow specific anatomic patterns, starting in the neck, jaw, shoulders, and back, and advancing to the trunk and limbs. These flare-ups end up in formation of heterotopic endochondral bone. Cumulative ossifications, which usually start at a median age of five years, result in progressive immobilisation, spinal fusion, scoliosis, ankylosed joints, and a reduced life span [17,18,19,20]. The most common cause of death in FOP patients is cardiorespiratory failure from thoracic insufficiency syndrome at a median age of 42 years [21]. Surgery to remove heterotopic ossifications is avoided since it leads to additional inflammation and further bone formation at the surgical site [19].
Additionally, some FOP patients present congenital alterations or associated features [13,18]. Congenital malformation of great toes, including hypoplasia or aplasia, is present in most FOP patients [13,18,22]. Congenital malformation of thumbs is also observed in approximately 50% of FOP patients [13,22]. Other variable FOP features include anomalies in the cervical spine (~80% prevalence) [23,24], proximal medial tibial benign osteochondromas (~90% prevalence) [24,25], short broad femoral necks (~70% prevalence) [24], and conductive hearing impairment (~50% prevalence) [22,24,26].

2.2. ACVR1 Genetic Mutations in FOP

In 2006, ACVR1 was identified as the gene responsible for FOP and the mutation c.617G>A, corresponding to Arg206His (R206H) of the ACVR1 gene, was found commonly linked to this disease [27]. This mutation is localised in the intracellular GS-rich domain of the ACVR1 receptor, and it is present in the majority of FOP patients, usually showing the typical clinical features detailed above [20,24]. The R206H mutation has been extensively analysed in multiple studies, different populations, and geographical groups worldwide, and it has been invariably linked to FOP [24,28,29,30,31,32,33,34,35,36,37,38,39,40]. To date, up to 14 different mutations have been identified to cause FOP, and all of them are localised in the ACVR1 gene (Figure 1). It has been estimated that the R206H mutation contributes to more than 95% of FOP patients [20,24]. The remaining cases can be attributed to atypical ACVR1 mutations, including L196P [41,42], P197_F198delinsL [24], R202I [43,44], Q207E [24], F246Y [45], R258S [28,31,33,35,46], R258G [16], G325A [47], G328E [24,34,43,48], G328R [24], G328W [24], G356D [24,33,49], and R375P [24].
While multiple case studies and reports have focused on the clinical aspects of R206H carriers, specific clinical features of atypical ACVR1 mutations have also been identified. These include differences among mutations in the age of ossification onset, association with traumatic flare-ups, anatomic patterns of ossification, malformation of toes, alopecia, facial features, hearing impairment, primary amenorrhea, cognitive impairment, severity of the disease, and life expectancy of FOP patients [18,24,52].

2.3. ACVR1 Mutants and Altered BMP Signalling

ACVR1 R206H presented mild, ligand-independent BMP signalling, as well as increased responsiveness to BMP stimulation in different cell lines, FOP patient-derived cells, and zebrafish embryonic models. Both alterations contribute to dysregulated BMP signalling [53,54,55,56,57]. In addition to the R206H mutation, almost all ACVR1 mutations detected in FOP and diffuse intrinsic pontine glioma (DIPG) also presented enhanced responsiveness to BMP, resulting in higher SMAD1/5/8 signalling upon BMP stimulation [50,58,59,60,61]. Interestingly, ACVR1 constructs with a deleted ligand-binding domain and intracellular activating mutations partially retained increased BMP signalling [59]. To explain these observations, it was proposed that changes in signalling arise from alterations in the ACVR1 interaction with FKBP12. FKBP12, a cytoplasmatic FK506-binding protein 1A (FKBP1A/FKBP12), was known as a negative regulator of type I receptors. In the absence of the ligand, FKBP12 binds to the GS domain of type I BMP receptors, suppressing their kinase activity and preventing leaky activation of type I BMP receptors. In response to complex formation, phosphorylation of the GS domain by BMP type II receptors releases FKBP12, allowing for complete type I receptor activation [5,6,62].
From crystallographic analysis, Chaikuad and colleagues found that multiple ACVR1 mutations observed in FOP patients could destabilise the inactive state of the receptor, and mutations in the GS domain could impair inhibition by FKBP12 [50]. Moreover, ACVR1 R206H showed impaired binding to FKBP12, resulting in loss of autoinhibition and leaky activation of BMP signalling, potentially contributing to the dysregulated ACVR1 BMP signalling observed [53,54,55,63]. Overexpression of FKBP12 was able to rescue, with different efficacies, the effect of almost all ACVR1 mutations detected in FOP and DIPG [60]. Remarkably, F246Y mutants showed activation levels equivalent to wild-type ACVR1, while P197_F198delinsL alteration was almost completely resistant to FKBP12 regulation due to site interaction loss [50,60]. Therefore, while most ACVR1 mutants, including R206H, present impaired binding to FKBP12, some residual interaction is still observed in almost all mutated ACVR1 receptors [60].

2.4. Activin A Signalling via ACVR1 Gain of Function Mutants

Activin A, also a member of the TGFβ superfamily, usually signals via the type I receptor ACVR1B and the type II receptor ACVR2A/B through the SMAD2/3 pathway [2,7,64]. Moreover, it was observed that activin A could bind to ACVR1 in the presence of ACVR2A/B. Interestingly, this activin A–ACVR1 binding was not observed in the presence of high levels of BMPR2 [2,61,64]. Given that activin A shares the type II receptor ACVR2A/B with BMP ligands, activin A is able to competitively antagonise SMAD1/5/8 activation upon BMP6/9 stimulation through ACVR2A/B-ACVR1 complexes [65]. Recent breakthrough studies have identified that the ACVR1 R206H mutation confers a neofunction. It was demonstrated that activin A was capable of inducing SMAD1/5/8 activation via ACVR1-R206H [61,66]. Additionally, activin A induced SMAD1/5/8 activation through ACVR1 in other atypical FOP ACVR1 mutations [59,61]. In response to activin A, this ACVR1-R206H neofunction was able to trigger endochondral ossification in vivo and enhance chondrogenesis of induced mesenchymal stromal cells derived from FOP-iPSCs in vitro [61].
It has been demonstrated that activin A signalling in fibro/adipogenic precursors (FAPs) can promote heterotopic ossification in ACVR1-R206H mice models. In these models, activin A is able to induce osteogenic differentiation and promote heterotopic ossification. Moreover, activin A blockade using ActA-mAb prevented HO of transplanted FAPs. Spontaneous and trauma-induced HO was also inhibited or partly reduced in most ActA-mAb treated mice [67]. Interestingly, while heterozygous ACVR1-R206H mutations cause FOP, complete loss of the wild-type ACVR1 allele in ACVR1R206H/+ mice results in a substantial increase in HO volume [67]. Altogether, this evidence highlights the importance of competition between wild type and mutant ACVR1 receptors in vivo. This competition for ligand binding and shared type II receptors could affect BMP signalling, which highlights the importance of the balance between type I and type II BMP receptor expression levels in the signalling outcome.
Further emphasising the importance of the relative balance between receptors, several studies have reported that activin A signalling through wild-type ACVR1 is cell type-dependent. Previously, it was assumed that activin A only signals via ACVR1B and ACVR2A/B through SMAD2/3 phosphorylation, and interaction with ACVR1-ACVR2A/B did not stimulate SMAD1/5/8 [66]. However, in immortalised murine embryonic fibroblasts (MEF cells) with wild-type ACVR1, a mild increase in SMAD1/5/8 signalling was observed in response to activin A [59]. These observations have also been reproduced in different myeloma cell lines and HepG2 liver carcinoma cells, describing activin A and activin B signalling via the wild-type ACVR1 receptor through SMAD1/5/8 [64]. Moreover, it was observed that loss of BMPR2 potentiated, and BMPR2 overexpression reduced, activin A signalling via ACVR1-ACVR2A/B through SMAD1/5/8 [64]. Therefore, it was proposed that in cells where ACVR1 is a limiting factor, complex formation preference (either ACVR1-BMPR2, ACVR1-ACVR2A/B, or ACVR1B-ACVR2A/B) determines the signalling outcome, which may be altered in the presence of ACVR1 R206H [64,68]. Cell type-specific signalling was also observed in FOP patient-derived cells. Activin A responses through SMAD1/5/8 were observed in human induced pluripotent stem cells (hiPSCs) from FOP patients. On the contrary, there are reports that hiPSC-derived endothelial cells (iECs) did not show SMAD1/5/8 activation after activin A stimulation, even though both hiPSCs and iECs carried the ACVR1-R206H mutation [69].
In recent work, a model to integrate the ligand–receptor promiscuity of the BMP pathway and the multiple possible ligand–receptor and receptor–receptor combinations was proposed. All these multiple combinatorial interactions that may vary from cell to cell or within the same cell in different environments defines the final signalling outcome [70]. Therefore, activin A responses through ACVR1 are critically affected by both the mutational status of ACVR1 and the relative balance of type II, and likely type I, receptors that are able to interact with ACVR1 (Figure 2).

2.5. ACVR1 Non-Canonical BMP Signalling

In addition to ACVR1-SMAD1 activation, SMAD-independent, non-canonical ACVR1 signalling may also play a role in ACVR1-related pathologies (Figure 2). For example, it was observed that lymphocytes derived from FOP patients presented a dysregulated ACVR1-p38 MAPK pathway that could be blocked by p38 inhibitors. However, other inhibitors for non-SMAD signalling pathways, such as ERK or JNK, did not block altered BMP signalling [71]. Additionally, mice lacking Acvr1 in cartilage presented reduced SMAD responses, but also decreased p38 MAPK activation [72].
The PI3K/AKT/mTOR pathway, which is also a non-canonical BMP pathway [11], has been linked to ACVR1. Multiple studies have linked mTOR to trauma-induced HO and FOP. Different inhibitors of mTORC complexes, including rapamycin, have decreased trauma-induced HO [73] and FOP ossification [74,75]. Additionally, the PI3K/AKT/GSK3 pathway has been linked to SMAD1 levels in murine osteoblasts, affecting bone homeostasis and regulation [76]. Recently, PI3K signalling was also linked to HO and FOP since inhibitors of PI3Kα prevented heterotopic ossification in vivo [77]. The Rho/ROCK pathway, a regulator of mechanosensing, cell motility, and cytoskeleton organisation, has also been related to ACVR1. Mutant Acvr1R206H/+ mice showed increased activation of RhoA, altered cell morphology, and misinterpretation of the tissue microenvironment [11,78].

3. Bone Regulation and Skeletal Development

As a type I BMP receptor, the role of ACVR1 in skeletal development and homeostasis has been extensively studied [79]. In addition, the relevance of ACVR1 mutations in FOP has triggered multiple studies on ACVR1 function in HO. Endochondral development of HO in FOP suggests the involvement of ACVR1 not only in osteogenesis but also in inflammation and chondrogenesis. Indeed, different mutant Acvr1 models have shown enhanced chondrogenesis. It was observed that in vitro overexpression of ACVR1-R206H increased BMP/SMAD signalling and sensitised cells to BMP, increasing osteogenic and chondrogenic differentiation and boosting mineralisation in response to BMP treatment [54]. In addition to BMP dependent responses, BMP independent signalling was also observed for ACVR1-R206H, promoting chondrogenesis [53]. Additionally, ACVR1-Q207D viral overexpression in chick forelimb buds promoted cartilage expansion and joint fusion [80].
In contrast, Acvr1 knock-out mice developed cranial and axial skeletal defects and exhibited decreased levels of phosphorylated SMAD1/5 and p38. Therefore, it was proposed that ACVR1 plays coordinated functions with BMPR1A and BMPR1B during endochondral ossification and is required for chondrocyte proliferation and differentiation [72]. Furthermore, inhibition of ACVR1 reduced heterotopic ossification [81]. Knock-down of Acvr1 also impaired BMP signalling, repressed BMP6-induced osteoblast differentiation, and enhanced myogenic differentiation [82,83]. Surprisingly, conditional disruption of Acvr1 and Bmpr1a in Osterix-expressing progenitors after weaning resulted in increased bone mass and altered bone composition. Yet, certain differences were observed regarding the age of induced deletion, and further studies are required to elucidate the functional mechanisms in relation to BMP receptors and bone regulation [84,85]. Furthermore, ACVR1 is required for periodontium ligament and alveolar bone development, regulation, and remodelling [86]. Loss of Acvr1 in the dental mesenchyme promoted dental defects. Additionally, Acvr1 was found to regulate dentin formation, odontoblast cell fate determination in incisors, and odontoblast differentiation in molars [87]. Altogether, these results highlight the essential role of ACVR1 as a regulator of osteogenic and chondrogenic differentiation and its importance in skeletal development.

4. Cardiovascular Development and Cardiac Diseases

BMP signalling has been related to heart morphogenesis and cardiac valve development, and ACVR1 has been studied as an essential receptor in these processes [88,89]. Throughout development, it has been shown that Acvr1 is required for proper induction of endothelial-to-mesenchymal transdifferentiation (EndMT) during mouse atrioventricular cushion formation [90]. Interestingly, ACVR1 and BMPR1A present non-redundant functions, which are essential in the appropriate arterial pole morphogenesis [91]. Therefore, the deletion of Acvr1 leads to altered expression of crucial genes for cardiac differentiation and regional identity [91], defects in aortic valve development [92], and promotion of cardiovascular pathologies, including the bicuspid aortic valve [93]. It has recently been described that BMP signalling via ACVR1 is also required for angiotensin II-induced cardiac hypertrophy and fibrosis [94].
ACVR1 has also been related to several cases of congenital heart defects (CHDs). In a screening of human patients with atrioventricular septum (AVS) defects, ACVR1-L343P and R307L alleles were found in AVS patients when compared to healthy controls. The R307L mutation, localised in the solvent-exposed region, was not predicted to affect ACVR1 protein structure, and its activity was not significantly affected after experimental validation. On the contrary, ACVR1-L343P, localised in a β-sheet in the PK domain, affected protein structure, displaying impaired kinase activity. These observations were validated in zebrafish embryos, where ACVR1-L343P overexpression disrupted atrioventricular canal formation. Altogether, this study highlighted ACVR1 as a potential causative of AVS defects [95].
The mutation H286N, localised in the kinase domain of ACVR1, close to the nucleotide-binding site, was reported in a CHD patient with Down syndrome. This mutation showed lower activity compared to the wild-type receptor [96]. Interestingly, while ACVR1 mutations present in FOP and DIPG usually increase signalling, ACVR1 mutations in cardiac pathologies, including L343P and H286N, decrease ACVR1 responsiveness (Figure 1). Additionally, ACVR1 was proposed as a candidate for congenital cardiac malformations in a study analysing whole exome sequence data from 342 patients with left-sided lesions [97]. Another proposed candidate was SMURF1 (SMAD specific E3 ubiquitin protein ligase 1), a ubiquitin ligase that targets BMP pathway SMADs for ubiquitination and degradation [97,98]. Moreover, several cases of ventricular dysfunction have been reported in adult [99] and young [100] patients with FOP, which carry the typical FOP mutation ACVR1-R206H [100].

5. Reproductive System

Many members and receptors of the TGFβ superfamily have been proposed as important regulators of reproductive system development and homeostasis. TGFβs, BMPs, GDFs, AMH/MIS (anti-Müllerian hormone/Müllerian inhibiting substance), activins, and inhibins display different patterns of expression, depending on cellular localisation, stage of maturation, or the developmental stage of the sexual organs. For example, patterns of expression of these ligands or receptors differ between oocyte, granulosa, or theca cells. Patterns of expression and function can also vary during different stages of follicle development [89,101,102].
ACVR1 has been substantially associated with reproductive regulation through BMP and AMH signalling. The anti-Müllerian hormone receptor type 2/MIS type II receptor (AMHR2/MISRII) has been previously identified as a type II receptor for AMH/MIS. Different studies have demonstrated that ACVR1 is a type I receptor for AMH/MIS, activating a BMP-like pathway [103,104,105]. BMPR-1A [106] and BMPR-1B [105] have also been reported to be type I receptors for AMH. AMH has been proposed as an important regulator and marker of ovarian function and folliculogenesis. AMH is mainly expressed by follicles between initial follicle recruitment and cyclic recruitment, and its expression decreases after follicle-stimulating hormone (FSH)-dependent follicle growth and selection [107]. AMH inhibits initial follicle recruitment, and it also decreases FSH sensitivity [108]. It has been suggested that the high AMH levels present in polycystic ovary syndrome (PCOS) patients decreases FSH sensitivity, thus disturbing normal follicle selection [107]. Additionally, both BMP6 [109] and BMP7 [110] decrease FSH sensitivity. Considering the previous observations, it has been suggested that alterations in ACVR1 may include modification of follicle selection, growth, and accumulation in PCOS patients [111].
In a cohort of PCOS patients, seven different polymorphisms within non-coding regions of the ACVR1 gene were studied. Interestingly, some polymorphisms (rs1220134, rs10497189, and rs2033962) were associated with the total follicle number and higher AMH levels, highlighting the importance of ACVR1 in folliculogenesis in humans [111]. Interestingly, the ACVR1 polymorphism rs1220134 was also associated with lower breast cancer risk [112]. Furthermore, a possible regulation of ACVR1 levels by cell-secreted exosomes during follicle maturation has been suggested [113].
AMH is also responsible for the regression of the müllerian ducts, the primordial anlage of the female reproductive tract, during male sexual differentiation [106]. A FOP patient with the R258G mutation in the ACVR1 gene that presented gonadal dysgenesis with sex reversal (karyotype 46, XY female) has been reported. It was suggested that the important role of ACVR1 in the mediation of AMH signalling during embryogenesis could be related to the observed sexual alteration [16]. ACVR1, in addition to BMPR1A and BMPR2/ACVR2A, through BMP2 signalling via SMAD1/5/8, contributes to the regulation of pentraxin 3 (PTX3) in human granulosa-lutein cells. PTX3 plays an essential role in fertility, regulating the formation and expansion of the cumulus-oophorus complex (COC), essential for ovulation [114]. Similar downregulation, mediated by ACVR1/BMPR1A of connexin 43 (Cx43) expression, a gap junction protein involved in cell–cell communication and corpus luteum development, was demonstrated in this model. Decreased Cx43 expression by BMP2 reduced cell–cell communication in human granulosa-lutein cells [115].
During embryonic implantation, uterine stromal cells react to embryonic invasion, triggering decidualisation, a process defined by the proliferation and differentiation of these cells. Since BMP2 is essential for embryonic development and ablation of BMP2 in the uterine stroma precludes decidualisation and embryonic development [116], ACVR1 was studied during uterine peri-implantation [117] and embryonic development [118]. In the absence of Acvr1, embryonic invasion and implantation were delayed, and uterine stromal cells failed to undergo the decidualisation process, resulting in sterility. Microarray analysis comparing control and knock-out Acvr1 murine uteri showed that the ablation of Acvr1 suppressed CCAAT/enhancer-binding protein b (Cebpb) expression during decidualisation, which in turn regulates the expression of the progesterone receptor (Pgr). This regulation occurred through direct binding between a regulatory sequence in the 3′ UTR of the Cebpb gene and SMAD1/5 [117]. Additionally, it was demonstrated that Acvr1 was essential for proper embryonic development, and embryos lacking Acvr1 presented defects in primordial germ cell formation [119] and were arrested at late gastrulation, provoking embryonic lethality [118].

6. ACVR1 in the Central Nervous System

Several reports have highlighted the potential importance of ACVR1 in the development and regulation of the central nervous system (CNS). Due to the relevance of ACVR1 in FOP and, more recently, DIPG pathologies, additional evidence has been observed. FOP patients present atypical CNS alterations, including mild cognitive impairment, cerebral cavernous malformations, cerebellar abnormalities, craniopharyngioma, hypoplasia of the brainstem (including pons and cerebellum), and diffuse cerebral dysfunction [16,18,24]. Additionally, neurological symptoms and sensory abnormalities are common in FOP patients [120].
BMP signalling plays an important role in CNS development and regulation. For instance, it has been shown that spinal cord development and proliferation, as well as cerebellar granule neuron differentiation, rely on BMP signalling [121]. Postnatal differentiation of cerebellar cells is also modulated by BMP and SMAD1 [122]. Furthermore, BMPs inhibit oligodendroglial differentiation and promote astroglial lineage commitment by neural progenitor cells during the late embryonic and postnatal periods and regulate the balance of adult neurogenesis and differentiation [121,123,124,125,126]. Strikingly, increased BMP signalling diminishes hippocampal neurogenesis and impaired cognition, while decreased BMP responsiveness enhances hippocampal cognition and neurogenesis in mouse models [127].
Recent MRI studies have identified FOP patients with T2-hyperintensity of the dentate nucleus regions with dentate nucleus lesions [128,129,130,131]. T2-hyperintense lesions were also observed in frontal periventricular white matter, the spinal cord, and the dorsal pons. Interestingly, BMP signalling increased in response to induced demyelination in the subventricular zone. It was suggested that enhanced BMP signalling in response to demyelination could disrupt reparation since increased BMP responses reduce oligodendrocyte production and increase the number of astrocytes [132]. Additionally, some FOP patients presented either abnormal soft tissue mass surrounding the brainstem or the ventral portion of the pons or lesions in the fourth ventricle, causing hydrocephalus [128,129]. In mouse models with dysregulated BMP signalling, either by BMP4 overexpression or ACVR1R206H mutation, demyelinated lesions and focal inflammatory changes of the CNS were observed. Remarkably, demyelinated areas seemed to correlate with the previously detected hyperintense lesions [131]. In a study involving 13 FOP patients, 11 carrying the R206H mutation and two carrying the R258S mutation all displayed thin T2-hyperintense bands at the ventral pons. These brainstem lesions resembled the hamartomatous tissue. All patients showed brainstem dysmorphism, with bulging of the dorsal pons, a thickened pontomedullary junction, and enlarged medulla [129]. Additionally, dentate nuclei abnormalities were observed in all patients. The size of the brainstem lesions did not correlate with age, onset of ossification, or severity of disability. Moreover, brainstem lesions were observed in two FOP patients in the first month of life [129]. These findings possibly suggest either a congenital malformation resulting from impaired regulation of brainstem progenitors during development or early, impaired regulation of the CNS, potentially affected by inflammation and dysregulated BMP signalling due to the ACVR1 mutations [129,131].
All this data correlate mutated ACVR1 with CNS alterations, suggesting a possible explanation to CNS alterations in FOP patients and a possible link to DIPG pathology. Nevertheless, additional studies should be performed to further explain the alterations exposed in this section. Specifically, we should improve the histological description of the phenotype and confirm the demyelination origin associated to the observed lesions. Additional studies should also demonstrate the relevance of altered BMP signalling in CNS lesions, in the balance of neural populations in FOP and DIPG patients, and the potential transfer of therapeutic strategies between FOP, DIPG, and CNS pathologies.

7. BMP Signalling and Pluripotency

BMPs have been shown to play a role in pluripotent stem cell (PSC) regulation. Initial investigations reported that BMP signalling supported self-renewal of mouse embryonic stem cells (mESCs) [133,134], although BMP signalling, in combination with basic fibroblast growth factor, inhibited human ESC self-renewal [135]. In mouse PSCs, BMP signalling induced mesenchymal-to-epithelial transition during the initial stages of reprogramming, promoting early stages of reprogramming of induced PSCs (iPSCs) [136].
Together with the development of human iPSC (hiPSC) protocols, several groups have generated hiPCSs from FOP patients carrying the ACVR1-R206H mutation. This increased the array of in vitro models to elucidate the molecular mechanisms and alterations underlying FOP and the study of ACVR1 functions [61,69,137,138,139,140,141]. Originally, it was reported that activation of BMP-SMAD signalling impaired hiPSC self-renewal in FOP patient cells [137]. However, it was also observed that BMP-SMAD signalling promotes reprogramming to pluripotency and hiPSC generation in the early reprogramming phase. Inhibition of BMP-SMAD responses reduced the efficiency of iPSC generation from FOP patients carrying the ACVR1-R206H mutation. In contrast, activation of the BMP-SMAD pathway in the early reprogramming phase increased the efficiency of hiPSC generation, while enhancement of BMP signalling in the late reprogramming phase promotes differentiation [141].
It is known that ID genes, as direct targets of the BMP-SMAD pathway [142], regulate cellular senescence through transcriptional repression of p16/INK4A (CDKN2A) [143]. p16/INK4A has important roles in iPSC generation since enhanced cell senescence impairs iPSC reprogramming [144]. Therefore, it was found that increased IDs expression, induced by BMP-SMAD activation in response to the ACVR1-R206H mutation, promoted reprogramming into iPSCs. Enhanced expression of ID genes decreased p16/INK4A protein levels, inhibiting p16/INK4A-mediated cell senescence and bypassing the cell senescence barrier to iPSC reprogramming [141]. Therefore, ACVR1 and the BMP-SMAD-ID pathway play essential roles during reprogramming processes.

8. ACVR1 in Cancer

8.1. Diffuse Intrinsic Pontine Glioma

DIPG, a high-grade paediatric brainstem glioma, is a lethal tumour with a mean age of diagnosis of 6–7 years and a median survival of 10 months after detection [145,146,147]. Surgical resection is inadequate due to tumour location within the brain, and radiotherapy is the only temporary treatment available to date [147,148,149]. Molecular knowledge and biological understanding of DIPG has been limited due to intrinsic difficulties regarding biopsy acquisition. As a result, attempts to find an effective therapy have been mainly based on research in glioblastomas from adults, which present highly different molecular alterations compared to DIPG [150]. In 2014, four different groups reported independent genomic analyses of DIPG, which increased our knowledge regarding the genomic landscape of this paediatric glioma. Interestingly, all four groups identified recurrent activating somatic mutations in the ACVR1 gene [149,151,152,153].
Among the molecular markers for DIPG, recurrent mutations in histone variants H3.3 (H3F3A) or H3.1 (HIST1H3B/C/I) with the K27M substitution are the most prevalent alterations present in 70–84% of DIPG cases. TP53 is the second most common mutation in DIPG patients (42–71%). The third most commonly mutated gene in DIPG is ACVR1, with a frequency of 20–32% among DIPG patients [147,149,151,152]. ACVR1 mutations in DIPG are strongly associated with the presence of the HIST1H3B mutation encoding K27M or with activation of the PI3K pathway [149,151,152,153]. Clinically, ACVR1 mutations are associated with younger age and slightly longer patient survival [149,151]. Surprisingly, Taylor and colleagues observed a strong predominance of females in the DIPG ACVR1 mutant group compared to the sex distribution in DIPG cases carrying wild-type ACVR1 [149].
Recurrent somatic mutations in the ACVR1 gene are clustered in the GS-rich domain or in the kinase domain, encoding R206H, Q207E, R258G, G328V, G328E, G328W, and G356D substitutions [149,151,152,153]. Surprisingly, these mutations that had not previously been reported in cancer are the same mutations found in FOP patients, excluding mutation G328V, which is, to date, exclusive of DIPG (Figure 1). These mutations lead to a gain of function of ACVR1, increasing BMP signalling, which increases ID proteins. Consistent with this, DIPG samples showed increased levels of signalling through ACVR1 [149,151,152,153].
Even though most ACVR1 mutations are shared between FOP and DIPG, the frequencies of the ACVR1 mutations are different. In fact, while the ACVR1-R206H mutation is found in more than 95% of FOP patients [20,24], ACVR1 mutations in DIPG are more diversely distributed, comprising R206H (20%), Q207E (2%), R258G (13%), G328V (28%), G328E (24%), G328W (4%), and G356D (9%) (Figure 1) [154]. The high ACVR1 mutational status in patients with DIPG suggests that ACVR1 directly contributes to DIPG oncogenesis. However, FOP patients with identical ACVR1 mutations do not present increased oncogenic predisposition to cancer or DIPG.
Brainstem progenitor cells expressing mutant ACVR1 showed enhanced proliferation and cell survival, and ACVR1-R206H was the most potent mutation [150,152]. Additionally, brainstem progenitors expressing ACVR1-R206H, with or without H3.1-K27M, showed increased markers for EMT and STAT3 signalling, which has been involved in mesenchymal transformation into glioblastomas [150,155]. This information strongly supports the hypothesis of both ACVR1 and H3.1 cooperating in DIPG oncogenesis. Mutant forms of both ACVR1 and histone H3.3 K27M increased the levels of phosphorylated SMAD1 and ID protein expression. The co-expression of mutant ACVR1 and H3.3 K27M created an additive effect, suggesting that several mechanisms may alter BMP signalling in DIPG [149,152].
Very interestingly, mutant Acvr1 (R206H, G328V, G328E) alone did not cause tumours in mouse models. In combination with H3.1-K27M, p53 loss, and Pten loss, mutant Acvr1 was only able to develop glioma-like lesions. The addition of the PDGFA ligand in the presence of Acvr1-R206H and H3.1-K27M increased the tumour occurrence. This is consistent with the PDGFRA amplifications detected in human DIPG [151,152]. In addition, a recent study on human DIPG patients to assess spatial and temporal driver mutations pointed to H3K27M as the initial oncogenic event in DIPG, which needed a partner driver, such as ACVR1 (or TP53, PPMID, or PIK3R1) to induce oncogenesis [156]. Altogether, this suggested that mutated ACVR1 is an oncogenic driver in a context where other driver mutations have occurred. Alterations in PI3K, which has a central role in cancer biology [157], may affect ACVR1-related DIPG oncogenesis. As introduced above, activating mutations in PI3K-MAPK pathways have been detected in human DIPGs, including nonsense mutations or deletions in PTEN and missense mutations in PIK3CA and PIK3R1 [149,152,156].
In addition to the described effects of mutant ACVR1 in DIPG pathology, further non-confirmed hypotheses have been proposed to explain the link between ACVR1 and DIPG. These include cell migration and apoptosis regulation, altered astroglial cell differentiation, regulation of cancer stem cell self-renewal, or the potential importance of hedgehog proteins and HOX genes, which are also affected by BMP signalling [20,158]. All these data have strongly validated ACVR1 as a cancer driver in DIPG. However, the functional mechanisms of ACVR1 in the development of DIPG or CNS alterations have not been fully elucidated.

8.2. Adult Glioblastoma

ACVR1 was studied using primary grade IV adult glioblastoma cells to elucidate the importance of BMPs in glioblastoma progression. Overexpression of constitutively active ACVR1 induced apoptosis in glioma-initiating cells (GICs), while inhibition of ACVR1 reduced apoptosis of GICs [159]. Additionally, mRNA and protein levels of DLX2, a transcription factor involved in development, cell proliferation, and differentiation [160,161], were upregulated in response to either BMP4 or BMP7. Overexpression of ACVR1 or DLX2 in primary grade IV glioblastoma cells orthotopically inoculated in nude mice resulted in impeded tumour growth and led to longer survival in vivo [159].

8.3. Ovarian Cancer

The essential functions of ACVR1 in the reproductive system have already been described in this review. Given that AMH, BMPs, and ACVR1 modulate female fertility and ovarian function, ACVR1 has also been studied in ovarian cancer. Wang and colleagues found that stress-induced phosphoprotein 1 (STIP1), a protein adaptor and modulator of heat shock proteins HSP70 and HSP90, was secreted by ovarian cancer cells and found in the blood of ovarian cancer patients [162]. STIP1 was observed to directly interact with ACVR1, activating it and increasing SMAD1/5 phosphorylation downstream of ACVR1. This signal was found to induce proliferation of ovarian cancer cells through this ACVR1-SMAD signalling pathway [163]. Altered BMP signalling in ovarian cancer was also observed in epithelial ovarian cancer cells since autocrine BMP9, which usually signals through ACVRL1, also signals through ACVR1, increasing the phosphorylation of SMAD1/5 and promoting ovarian cancer cell proliferation [164].
As explained in the reproductive system section, ACVR1 is one of the BMP type I receptors that binds AMH, which has been associated with ovarian cancer [165]. In granulosa cell tumours (GCTs), sex cord stromal ovarian tumours, it was observed that AMH responses through BMP type I receptors inhibited the growth of GCTs, possibly including the induction of cellular apoptosis [166]. In epithelial ovarian cancer (EOC), the most common type of ovarian cancer, it was suggested that AMH was also able to inhibit proliferation and induce apoptosis of EOC cells, inducing G1/S cell cycle arrest [167]. AMH was also observed to inhibit proliferation and growth of human ovarian cancer cell lines in vitro [168] and in vivo [169].
Surprisingly, even though the main signalling pathway SMAD1/5 is shared, BMP or AMH can either promote or inhibit ovarian cancer cell proliferation. AMH inhibits proliferation, while BMP or other ACVR1 activation mechanisms enhance proliferation. These differences may result from different expression abundances of BMP type I receptors in each cellular type [102], different BMP type I receptor affinities to AMH [103], or different environmental conditions altering ACVR1 and/or SMAD downstream signalling. In conclusion, while the main players in ovarian cancer are already known, further in-depth studies are needed to refine the relevance of ACVR1 in ovarian cancer.

8.4. Endometrial Cancer

A recent bioinformatic analysis using the mutational data generated by The Cancer Genome Atlas (TCGA) consortium revealed that 3.3% of the endometrial cancer cohort carried ACVR1 mutations. Interestingly, some of these mutations were observed in FOP and DIPG patients. As in DIPG, ACVR1 mutations have a tendency to co-occur associated with those on the PI3K-mTOR pathway [170].
When analysing mutations annotated in The Catalogue of Somatic Mutations In Cancer (COSMIC) [171], one of the largest curated databases of somatic mutations, which includes additional endometrial cancer datasets [172], ACVR1 mutations are found in 2.69% (25 mutated samples out of 931 samples tested) of endometrial cancers. Strikingly, 10 out of 25 ACVR1 mutations found in patients are localised in positions also important in FOP and DIPG. Mutations R202I (identical mutation in FOP) and R375C (R375P is found in FOP) are present in endometrial cancer patients. A small cluster of two patients with G356D (identical mutation in FOP and DIPG) is also observed. The R206H mutation, observed in DIPG, and the most prevalent mutation in FOP, was present in six different patients [171,173].

8.5. Prostate Cancer

Prostate cancer has been associated with ACVR1 through endoglin, a TGFβ superfamily auxiliary receptor identified as a marker of tumour angiogenesis and neovascularisation [174]. The canonical signalling mechanism comprises endoglin modulating TGFβ responses (TGF-β1 and TGF-β3) by association to TGF-βR2. This interaction activates TGF-βR1 (either TGFBR1 or ACVRL1) intracellular kinase, phosphorylating SMAD proteins (SMAD2/3 or SMAD1/5, respectively) [174]. However, it has been reported that endoglin also interacts with and activates ACVR1, inhibiting prostate cancer cell migration [175]. On the contrary, ACVR1 could also phosphorylate endoglin, promoting prostate cancer cell migration [176]. This dual role of ACVR1 in prostate cancer cell migration should be further confirmed to ascertain whether this mechanism is also present in different cancer types and ACVR1-related pathologies. It could suggest a similar signalling balance depending on the receptor association with endoglin.

8.6. Multiple Myeloma

ACVR1 has been associated with multiple myeloma, a hematological cancer that arises from hematopoietic cells. It was described that BMP9 displayed tumour suppressor activity through ACVR1 signalling, inducing apoptosis and growth arrest in myeloma cells. It was suggested that high levels of endoglin in myeloma cancer cells may scavenge BMP9, impairing the growth inhibitory effects of BMP9 [177]. This hypothesis adds information to the endoglin roles observed in prostate cancer, yet the mechanistic process regulating endoglin-ACVR1 functions should still be defined.
In recent work, it was observed that activin A and B could signal through wild-type ACVR1 in myeloma cells, activating the SMAD1/5/8 signalling pathway and inducing myeloma cell death. Loss of BMPR2 enhanced activin and BMP induced SMAD1/5/8 signalling, and expression of BMPR2 reduced activin-induced SMAD1/5/8 activity. Therefore, it was proposed that a reduction in BMPR2 levels could increase ACVR1 complex formation with ACVR2A or ACVR2B, thus enhancing ACVR1-mediated SMAD1/5/8 signalling and myeloma cell death [64].

8.7. Eye Lens Tumour

ACVR1 has also been proposed as a regulator of mouse eye fibre cell proliferation and apoptosis [178]. It was suggested that ACVR1 signalling suppresses proliferation in the lens. P53 knock-out formed epithelioid plaques similar to subcapsular cataracts, while double knock-out ACVR1/P53 promoted large proliferating tumour-like masses in the lens. Therefore, ACVR1 was proposed as a potential tumour suppressor for eye lens tumours [179].

8.8. Erythroleukemia

Overexpression of ACVR1 in the human cell line TF-1, a model of erythroleukemia, resulted in enhanced SMAD1/5 signalling in response to BMP9 and increased cell proliferation. Inhibition of ACVR1 reversed the observed effect, reducing cell proliferation [180].

8.9. Head and Neck Squamous Cell Carcinomas

Head and neck squamous cell carcinomas (HNSCCs) present several risk factors, highlighting smoking and alcohol consumption. Furthermore, several genes with frequent somatic mutations have been associated with HNSCCs [181,182]. Copy number gains of the ACVR1 gene were reported in HNSCCs in different anatomical regions, including the oral cavity, pharynx, and larynx. Carcinomas with copy number gains of ACVR1 had increased expression of ACVR1 and were associated with longer survival [181]. Further research is needed to decipher the molecular mechanism underlying these observations and to define ACVR1 as a potential HNSCC prognosis marker.

8.10. Dual Role of ACVR1 and BMPs in Cancer Biology

Multiple reports have highlighted the apparent dual function of BMPs in cancer [183,184,185]. Different BMPs, or the same BMP ligand in different cancer cell types, can promote or inhibit several cancer hallmarks, including cell proliferation, survival, stemness, and migration. For example, BMP7 was shown to promote, inhibit, or not affect cell growth, depending on the breast cancer cell line [185,186]. Similarly, diverse BMP7 cell type-dependent functions affecting cell growth, invasiveness, and cell survival were observed in different prostate tumour cell lines [187]. BMP2 promoted cell proliferation in ovarian cancer cells [188] and invasiveness in pancreatic cancer cells [189], induced EMT and stemness in breast cancer cells [190], and enhanced proliferation and invasion in nasopharyngeal cancer cell lines [191]. Conversely, BMP2 inhibited growth of human osteosarcoma cells [192]; inhibited colon cancer cell proliferation, migration, and invasiveness [193,194]; suppressed tumour growth of human renal cell carcinoma cell lines [195], and inhibited growth and migration of hepatocellular carcinoma cell lines [196].
Multiple additional examples of dual function have been observed and reviewed, raising the question of the role of BMPs as tumour suppressors or oncogenes [183,184]. In light of this evidence, we suggest that the tumour and cell type, together with the tumour microenvironment, are key factors in the regulation of signalling by BMPs. As detailed above, ACVR1 is a well-known cancer driver due to its essential role in diffuse intrinsic pontine glioma initiation and progression. However, similar to the dual role of BMPs, ACVR1 could act as a tumour suppressor or oncogene, depending on the cancer type, cell type, or ligand involved (Table 1). Therefore, ACVR1 should be considered a complex regulator of cancer biology.

9. Conclusions and Perspectives

ACVR1 and its signalling were initially studied as a type I BMP receptor. Following the discovery of ACVR1 as the gene responsible for FOP, prior knowledge of ACVR1 was combined with new findings regarding its function and regulation in order to advance towards a potential treatment for this pathology. Similarly, after the discovery of ACVR1 as a cancer driver gene in DIPG, the research on this type of cancer was highly benefited from prior accumulated knowledge. Even though ACVR1 is now mainly known for its relevance in FOP and DIPG, this receptor is also involved in other pathologies. Moreover, due to its relevance in DIPG, ACVR1 is also being studied in multiple cancers. However, further in-depth studies are needed to refine the molecular basis of ACVR1 function in normal physiology and related pathologies. Therefore, we consider that open, multidisciplinary collaboration and information exchange across research from basic molecular and cellular biology and research from ACVR1-linked pathologies, including FOP, DIPG, and other pathologies, will help to promote robust knowledge and to identify novel therapeutic opportunities.

Author Contributions

J.A.V., C.S.-d.-D., C.P.-L., J.L.R. and F.V. conceived and discussed the manuscript. J.A.V. and F.V. wrote the manuscript and drew the figures.

Funding

This research was supported by grants from the Ministry of Education of Spain (M.E.C.) (BFU2017-8 2421-P) and from ACT for FOP Research Grant (IFOPA).

Acknowledgments

We would like to thank Claudia Arnedo-Pac for her help with figure preparation. For Figure 1B, molecular graphics and analyses were performed with UCSF Chimera, developed by the Resource for Biocomputing, Visualization, and Informatics at the University of California, San Francisco, with support from NIH P41-GM103311. Cristina Sánchez de Diego and Carolina Pimenta Costa were the recipients of a F.P.U. fellowship from the Spanish Ministry of Education.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Röijer, E.; Miyazono, K.; Åström, A.K.; Van Kessel, A.G.; Ten Dijke, P.; Stenman, G. Chromosomal localization of three human genes encoding members of the TGF-β superfamily of type I serine/threonine kinase receptors. Mamm. Genome 1998, 9, 266–268. [Google Scholar] [CrossRef] [PubMed]
  2. Attisano, L.; Cárcamo, J.; Ventura, F.; Weis, F.M.B.; Massagué, J.; Wrana, J.L. Identification of human activin and TGFβ type I receptors that form heteromeric kinase complexes with type II receptors. Cell 1993, 75, 671–680. [Google Scholar] [CrossRef]
  3. Matsuzaki, K.; Xu, J.; Wang, F.; McKeehan, W.L.; Krummen, L.; Kan, M. A widely expressed transmembrane serine/threonine kinase that does not bind activin, inhibin, transforming growth factor β, or bone morphogenic factor. J. Biol. Chem. 1993, 268, 12719–12723. [Google Scholar]
  4. Melé, M.; Ferreira, P.G.; Reverter, F.; DeLuca, D.S.; Monlong, J.; Sammeth, M.; Young, T.R.; Goldmann, J.M.; Pervouchine, D.D.; Sullivan, T.J.; et al. The human transcriptome across tissues and individuals. Science 2015, 348, 660–665. [Google Scholar] [CrossRef] [Green Version]
  5. Huse, M.; Muir, T.W.; Xu, L.; Chen, Y.G.; Kuriyan, J.; Massagué, J. The TGFβ receptor activation process: An inhibitor- to substrate-binding switch. Mol. Cell 2001, 8, 671–682. [Google Scholar] [CrossRef]
  6. Huse, M.; Chen, Y.G.; Massagué, J.; Kuriyan, J. Crystal structure of the cytoplasmic domain of the type I TGFβ receptor in complex with FKBP12. Cell 1999, 96, 425–436. [Google Scholar] [CrossRef]
  7. Yadin, D.; Knaus, P.; Mueller, T.D. Structural insights into BMP receptors: Specificity, activation and inhibition. Cytokine Growth Factor Rev. 2016, 27, 13–34. [Google Scholar] [CrossRef]
  8. Wrana, J.L.; Attisano, L.; Cárcamo, J.; Zentella, A.; Doody, J.; Laiho, M.; Wang, X.F.; Massague, J. TGFβ signals through a heteromeric protein kinase receptor complex. Cell 1992, 71, 1003–1014. [Google Scholar] [CrossRef]
  9. Derynck, R.; Zhang, Y.E. Smad-dependent and Smad-independent pathways in TGF-β family signalling. Nature 2003, 425, 577–584. [Google Scholar] [CrossRef]
  10. Chen, Y.G.; Massagué, J. Smad1 recognition and activation by the ALK1 group of transforming growth factor-β family receptors. J. Biol. Chem. 1999, 274, 3672–3677. [Google Scholar] [CrossRef]
  11. Zhang, Y.E. Non-Smad signaling pathways of the TGF-β family. Cold Spring Harb. Perspect. Biol. 2017, 9. [Google Scholar] [CrossRef] [PubMed]
  12. Shore, E.M.; Kaplan, F.S. Inherited human diseases of heterotopic bone formation. Nat. Rev. Rheumatol. 2010, 6, 518–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kaplan, F.S.; Chakkalakal, S.A.; Shore, E.M. Fibrodysplasia ossificans progressiva: mechanisms and models of skeletal metamorphosis. Dis. Model. Mech. 2012, 5, 756–762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Connor, J.M.; Evans, D.A. Genetic aspects of fibrodysplasia ossificans progressiva. J. Med. Genet. 1982, 19, 35–39. [Google Scholar] [CrossRef]
  15. Shore, E.M.; Feldman, G.J.; Xu, M.; Kaplan, F.S. The genetics of fibrodysplasia ossificans progressiva. Clin. Rev. Bone Miner. Metab. 2005, 3, 201–204. [Google Scholar] [CrossRef]
  16. Kaplan, F.S.; Kobori, J.A.; Orellana, C.; Calvo, I.; Rosello, M.; Martinez, F.; Lopez, B.; Xu, M.; Pignolo, R.J.; Shore, E.M.; et al. Multi-system involvement in a severe variant of fibrodysplasia ossificans progressiva (ACVR1 c.772G>A; R258G): A report of two patients. Am. J. Med. Genet. Part A 2015, 167, 2265–2271. [Google Scholar] [CrossRef]
  17. Pignolo, R.J.; Suda, R.K.; Kaplan, F.S. The fibrodysplasia ossificans progressiva lesion. Clin. Rev. Bone Miner. Metab. 2005, 3, 195–200. [Google Scholar] [CrossRef]
  18. Hüning, I.; Gillessen-Kaesbach, G. Fibrodysplasia ossificans progressiva: Clinical course, genetic mutations and genotype-phenotype correlation. Mol. Syndromol. 2014, 5, 201–211. [Google Scholar] [CrossRef]
  19. Pignolo, R.J.; Bedford-Gay, C.; Liljesthröm, M.; Durbin-Johnson, B.P.; Shore, E.M.; Rocke, D.M.; Kaplan, F.S. The Natural History of Flare-Ups in Fibrodysplasia Ossificans Progressiva (FOP): A Comprehensive Global Assessment. J. Bone Miner. Res. 2016, 31, 650–656. [Google Scholar] [CrossRef]
  20. Pacifici, M.; Shore, E.M. Common mutations in ALK2/ACVR1, a multi-faceted receptor, have roles in distinct pediatric musculoskeletal and neural orphan disorders. Cytokine Growth Factor Rev. 2016, 27, 93–104. [Google Scholar] [CrossRef]
  21. Kaplan, F.S.; Zasloff, M.A.; Kitterman, J.A.; Shore, E.M.; Hong, C.C.; Rocke, D.M. Early mortality and cardiorespiratory failure in patients with fibrodysplasia ossificans progressiva. J. Bone Jt. Surg. Ser. A 2010, 92, 686–691. [Google Scholar] [CrossRef] [PubMed]
  22. Kaplan, F.S.; Glaser, D.L.; Shore, E.M.; Deirmengian, G.K.; Gupta, R.; Delai, P.; Morhart, R.; Smith, R.; Le Merrer, M.; Rogers, J.G.; et al. The phenotype of fibrodysplasia ossificans progressiva. Clin. Rev. Bone Miner. Metab. 2005, 3, 183–188. [Google Scholar] [CrossRef]
  23. Schaffer, A.A.; Kaplan, F.S.; Tracy, M.R.; O’Brien, M.L.; Dormans, J.P.; Shore, E.M.; Harland, R.M.; Kusumi, K. Developmental anomalies of the cervical spine in patients with fibrodysplasia ossificans progressiva are distinctly different from those in patients with Klippel-Feil syndrome: Clues from the BMP signaling pathway. Spine (Phila. Pa. 1976) 2005, 30, 1379–1385. [Google Scholar] [CrossRef] [PubMed]
  24. Kaplan, F.S.; Xu, M.; Seemann, P.; Connor, J.M.; Glaser, D.L.; Carroll, L.; Delai, P.; Fastnacht-Urban, E.; Forman, S.J.; Gillessen-Kaesbach, G.; et al. Classic and atypical fibrodysplasia ossificans progressiva (FOP) phenotypes are caused by mutations in the bone morphogenetic protein (BMP) type I receptor ACVR1. Hum. Mutat. 2009, 30, 379–390. [Google Scholar] [CrossRef]
  25. Deirmengian, G.K.; Hebela, N.M.; O’Connell, M.; Glaser, D.L.; Shore, E.M.; Kaplan, F.S. Proximal tibial osteochondromas in patients with fibrodysplasia ossificans progressiva. J. Bone Jt. Surg. Ser. A 2008, 90, 366–374. [Google Scholar] [CrossRef]
  26. Levy, C.E.; Lash, A.T.; Janoff, H.B.; Kaplan, F.S. Conductive Hearing Loss in Individuals with Fibrodysplasia Ossificans Progressiva. Am. J. Audiol. 1999, 8, 29–33. [Google Scholar] [CrossRef]
  27. Shore, E.M.; Xu, M.; Feldman, G.J.; Fenstermacher, D.A.; Brown, M.A.; Kaplan, F.S. A recurrent mutation in the BMP type I receptor ACVR1 causes inherited and sporadic fibrodysplasia ossificans progressiva. Nat. Genet. 2006, 38, 525–527. [Google Scholar] [CrossRef]
  28. Bocciardi, R.; Bordo, D.; Di Duca, M.; Di Rocco, M.; Ravazzolo, R. Mutational analysis of the ACVR1 gene in Italian patients affected with fibrodysplasia ossificans progressiva: Confirmations and advancements. Eur. J. Hum. Genet. 2009, 17, 311–318. [Google Scholar] [CrossRef]
  29. Guo, H.; Peng, D.; Xu, M.; Xue, J.; Lu, L.; Xu, X.; Liu, Y.; Xiong, Z.; Pan, Q.; Hu, Z.; et al. Report of two FOP cases with 617G > A mutation in the ACVR1 gene from Chinese population. Clin. Dysmorphol. 2010, 19, 206–208. [Google Scholar] [CrossRef]
  30. Dandara, C.; Scott, C.; Urban, M.; Fieggen, K.; Arendse, R.; Beighton, P. Confirmation of the recurrent ACVR1 617G>A mutation in South Africans with fibrodysplasia ossificans progressiva. South African Med. J. 2012, 102, 631–633. [Google Scholar] [CrossRef]
  31. Morales-Piga, A.; Bachiller-Corral, J.; Trujillo-Tiebas, M.J.; Villaverde-Hueso, A.; Gamir-Gamir, M.L.; Alonso-Ferreira, V.; Vázquez-Díaz, M.; Posada de la Paz, M.; Ayuso-García, C. Fibrodysplasia ossificans progressiva in Spain: Epidemiological, clinical, and genetic aspects. Bone 2012, 51, 748–755. [Google Scholar] [CrossRef] [PubMed]
  32. Nakajima, M.; Haga, N.; Takikawa, K.; Manabe, N.; Nishimura, G.; Ikegawa, S. The ACVR1 617G>A mutation is also recurrent in three Japanese patients with fibrodysplasia ossificans progressiva. J. Hum. Genet. 2007, 52, 473–475. [Google Scholar] [CrossRef]
  33. Zhang, W.; Zhang, K.; Song, L.; Pang, J.; Ma, H.; Shore, E.M.; Kaplan, F.S.; Wang, P. The phenotype and genotype of fibrodysplasia ossificans progressiva in China: A report of 72 cases. Bone 2013, 57, 386–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Carvalho, D.R.; Navarro, M.M.M.; Martins, B.J.A.F.; Coelho, K.E.F.A.; Mello, W.D.; Takata, R.I.; Speck-Martins, C.E. Mutational screening of ACVR1 gene in Brazilian fibrodysplasia ossificans progressiva patients. Clin. Genet. 2010, 77, 171–176. [Google Scholar] [CrossRef] [PubMed]
  35. Eresen Yazicioǧlu, Ç.; Karatosun, V.; Kizildaǧ, S.; Özsoylu, D.; Kavukçu, S. ACVR1 gene mutations in four Turkish patients diagnosed as fibrodysplasia ossificans progressiva. Gene 2013, 515, 444–446. [Google Scholar] [CrossRef]
  36. Shukla, A.; Taywade, O.; Stephen, J.; Gupta, D.; Phadke, S.R. Fibrodysplasia ossificans progressiva: Three Indian patients with mutation in the ACVR1 gene. Indian J. Pediatr. 2014, 81, 617–619. [Google Scholar] [CrossRef]
  37. Tian, S.; Zhu, J.; Lu, Y. Difficult diagnosis and genetic analysis of fibrodysplasia ossificans progressiva: A case report. BMC Med. Genet. 2018, 19. [Google Scholar] [CrossRef]
  38. Lin, G.T.; Chang, H.W.; Liu, C.S.; Huang, P.J.; Wang, H.C.; Cheng, Y.M. De novo 617G-A nucleotide mutation in the ACVR1 gene in a Taiwanese patient with fibrodysplasia ossificans progressiva. J. Hum. Genet. 2006, 51, 1083–1086. [Google Scholar] [CrossRef]
  39. Lee, D.Y.; Cho, T.J.; Lee, H.R.; Park, M.S.; Yoo, W.J.; Chung, C.Y.; Choi, I.H. ACVR1 gene mutation in sporadic Korean patients with fibrodysplasia ossificans progressiva. J. Korean Med. Sci. 2009, 24, 433–437. [Google Scholar] [CrossRef]
  40. Sun, Y.; Xia, W.; Jiang, Y.; Xing, X.; Li, M.; Wang, O.; Zhang, H.; Hu, Y.; Liu, H.; Meng, X.; et al. A recurrent mutation c.617G>A in the ACVR1 gene causes fibrodysplasia ossificans progressiva in two chinese patients. Calcif. Tissue Int. 2009, 84, 361–365. [Google Scholar] [CrossRef]
  41. Gregson, C.L.; Hollingworth, P.; Williams, M.; Petrie, K.A.; Bullock, A.N.; Brown, M.A.; Tobias, J.H.; Triffitt, J.T. A novel ACVR1 mutation in the glycine/serine-rich domain found in the most benign case of a fibrodysplasia ossificans progressiva variant reported to date. Bone 2011, 48, 654–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Nakahara, Y.; Katagiri, T.; Ogata, N.; Haga, N. ACVR1 (587T>C) mutation in a variant form of fibrodysplasia ossificans progressiva: Second report. Am. J. Med. Genet. Part A 2014, 164, 220–224. [Google Scholar] [CrossRef] [PubMed]
  43. Petrie, K.A.; Lee, W.H.; Bullock, A.N.; Pointon, J.J.; Smith, R.; Russell, R.G.G.; Brown, M.A.; Wordsworth, B.P.; Triffitt, J.T. Novel mutations in ACVR1 result in atypical features in two fibrodysplasia ossificans progressiva patients. PLoS ONE 2009, 4, e5005. [Google Scholar] [CrossRef] [PubMed]
  44. Barnett, C.P.; Dugar, M.; Haan, E.A. Late-onset variant fibrodysplasia ossificans progressiva leading to misdiagnosis of ankylosing spondylitis. Am. J. Med. Genet. Part A 2011, 155, 1492–1495. [Google Scholar] [CrossRef]
  45. Rafati, M.; Mohamadhashem, F.; Hoseini, A.; Hoseininasab, F.; Ghaffari, S.R. A novel ACVR1 mutation detected by whole exome sequencing in a family with an unusual skeletal dysplasia. Eur. J. Med. Genet. 2016, 59, 330–336. [Google Scholar] [CrossRef]
  46. Ratbi, I.; Borcciadi, R.; Regragui, A.; Ravazzolo, R.; Sefiani, A. Rarely occurring mutation of ACVR1 gene in Moroccan patient with fibrodysplasia ossificans progressiva. Clin. Rheumatol. 2010, 29, 119–121. [Google Scholar] [CrossRef]
  47. Whyte, M.P.; Wenkert, D.; Demertzis, J.L.; Dicarlo, E.F.; Westenberg, E.; Mumm, S. Fibrodysplasia ossificans progressiva: Middle-age onset of heterotopic ossification from a unique missense mutation (c.974G > C, p.G325A) in ACVR1. J. Bone Miner. Res. 2012, 27, 729–737. [Google Scholar] [CrossRef]
  48. Gucev, Z.; Tasic, V.; Plaseska-Karanfilska, D.; Dimishkovska, M.; Laban, N.; Bozinovski, Z.; Kostovski, M.; Saveski, A.; Polenakovic, M.; Towler, O.W.; et al. Severe digital malformations in a rare variant of fibrodysplasia ossificans progressiva. Am. J. Med. Genet. Part A 2019, 179, 1310–1314. [Google Scholar] [CrossRef]
  49. Fukuda, T.; Kanomata, K.; Nojima, J.; Kokabu, S.; Akita, M.; Ikebuchi, K.; Jimi, E.; Komori, T.; Maruki, Y.; Matsuoka, M.; et al. A unique mutation of ALK2, G356D, found in a patient with fibrodysplasia ossificans progressiva is a moderately activated BMP type I receptor. Biochem. Biophys. Res. Commun. 2008, 377, 905–909. [Google Scholar] [CrossRef]
  50. Chaikuad, A.; Alfano, I.; Kerr, G.; Sanvitale, C.E.; Boergermann, J.H.; Triffitt, J.T.; Von Delft, F.; Knapp, S.; Knaus, P.; Bullock, A.N. Structure of the bone morphogenetic protein receptor ALK2 and implications for fibrodysplasia ossificans progressiva. J. Biol. Chem. 2012, 287, 36990–36998. [Google Scholar] [CrossRef]
  51. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimera—A visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605–1612. [Google Scholar] [CrossRef] [PubMed]
  52. Nakahara, Y.; Suzuki, R.; Katagiri, T.; Toguchida, J.; Haga, N. Phenotypic differences of patients with fibrodysplasia ossificans progressive due to p.Arg258Ser variants of ACVR1. Hum. Genome Var. 2015, 2. [Google Scholar] [CrossRef] [PubMed]
  53. Shen, Q.; Little, S.C.; Xu, M.; Haupt, J.; Ast, C.; Katagiri, T.; Mundlos, S.; Seemann, P.; Kaplan, F.S.; Mullins, M.C.; et al. The fibrodysplasia ossificans progressiva R206H ACVR1 mutation activates BMP-independent chondrogenesis and zebrafish embryo ventralization. J. Clin. Investig. 2009, 119, 3462–3471. [Google Scholar] [CrossRef] [PubMed]
  54. Van Dinther, M.; Visser, N.; De Gorter, D.J.J.; Doorn, J.; Goumans, M.J.; De Boer, J.; Ten Dijke, P. 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. [Google Scholar] [CrossRef] [PubMed]
  55. Song, G.A.; Kim, H.J.; Woo, K.M.; Baek, J.H.; Kim, G.S.; Choi, J.Y.; Ryoo, H.M. Molecular consequences of the ACVR1 R206H mutation of fibrodysplasia ossificans progressiva. J. Biol. Chem. 2010, 285, 22542–22553. [Google Scholar] [CrossRef] [PubMed]
  56. Billings, P.C.; Fiori, J.L.; Bentwood, J.L.; O’Connell, M.P.; Jiao, X.; Nussbaum, B.; Caron, R.J.; Shore, E.M.; Kaplan, F.S. 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. [Google Scholar] [CrossRef] [PubMed]
  57. Fukuda, T.; Kohda, M.; Kanomata, K.; Nojima, J.; Nakamura, A.; Kamizono, J.; Noguchi, Y.; Iwakiri, K.; Kondo, T.; Kurose, J.; et al. Constitutively activated ALK2 and increased SMAD1/5 cooperatively induce bone morphogenetic protein signaling in fibrodysplasia ossificans progressiva. J. Biol. Chem. 2009, 284, 7149–7156. [Google Scholar] [CrossRef] [PubMed]
  58. Haupt, J.; Deichsel, A.; Stange, K.; Ast, C.; Bocciardi, R.; Ravazzolo, R.; Di Rocco, M.; Ferrari, P.; Landi, A.; Kaplan, F.S.; et al. ACVR1 p.Q207E causes classic fibrodysplasia ossificans progressiva and is functionally distinct from the engineered constitutively active ACVR1 p.Q207D variant. Hum. Mol. Genet. 2014, 23, 5364–5377. [Google Scholar] [CrossRef]
  59. Haupt, J.; Xu, M.; Shore, E.M. Variable signaling activity by FOP ACVR1 mutations. Bone 2018, 109, 232–240. [Google Scholar] [CrossRef]
  60. Machiya, A.; Tsukamoto, S.; Ohte, S.; Kuratani, M.; Fujimoto, M.; Kumagai, K.; Osawa, K.; Suda, N.; Bullock, A.N.; Katagiri, T. Effects of FKBP12 and type II BMP receptors on signal transduction by ALK2 activating mutations associated with genetic disorders. Bone 2018, 111, 101–108. [Google Scholar] [CrossRef]
  61. Hino, K.; Ikeya, M.; Horigome, K.; Matsumoto, Y.; Ebise, H.; Nishio, M.; Sekiguchi, K.; Shibata, M.; Nagata, S.; Matsuda, S.; et al. Neofunction of ACVR1 in fibrodysplasia ossificans progressiva. Proc. Natl. Acad. Sci. USA 2015, 112, 15438–15443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Wang, T.; Li, B.Y.; Danielson, P.D.; Shah, P.C.; Rockwell, S.; Lechleider, R.J.; Martin, J.; Manganaro, T.; Donahoe, P.K. The immunophilin FKBP12 functions as a common inhibitor of the TGFβ family type I receptors. Cell 1996, 86, 435–444. [Google Scholar] [CrossRef]
  63. Groppe, J.C.; Wu, J.; Shore, E.M.; Kaplan, F.S. In vitro analyses of the dysregulated R206H ALK2 kinase-FKBP12 interaction associated with heterotopic ossification in FOP. Cells Tissues Organs 2011, 194, 291–295. [Google Scholar]
  64. Olsen, O.E.; Sankar, M.; Elsaadi, S.; Hella, H.; Buene, G.; Darvekar, S.R.; Misund, K.; Katagiri, T.; Knaus, P.; Holien, T. BMPR2 inhibits activin and BMP signaling via wild-type ALK2. J. Cell Sci. 2018, 131, jcs213512. [Google Scholar] [CrossRef] [PubMed]
  65. Olsen, O.E.; Wader, K.F.; Hella, H.; Mylin, A.K.; Turesson, I.; Nesthus, I.; Waage, A.; Sundan, A.; Holien, T. Activin A inhibits BMP-signaling by binding ACVR2A and ACVR2B. Cell Commun. Signal. 2015, 13. [Google Scholar] [CrossRef] [PubMed]
  66. Hatsell, S.J.; Idone, V.; Wolken, D.M.A.; Huang, L.; Kim, H.J.; Wang, L.; Wen, X.; Nannuru, K.C.; Jimenez, J.; Xie, L.; et al. ACVR1R206H receptor mutation causes fibrodysplasia ossificans progressiva by imparting responsiveness to activin A. Sci. Transl. Med. 2015, 7. [Google Scholar] [CrossRef]
  67. Lees-Shepard, J.B.; Yamamoto, M.; Biswas, A.A.; Stoessel, S.J.; Nicholas, S.A.E.; Cogswell, C.A.; Devarakonda, P.M.; Schneider, M.J.; Cummins, S.M.; Legendre, N.P.; et al. Activin-dependent signaling in fibro/adipogenic progenitors causes fibrodysplasia ossificans progressiva. Nat. Commun. 2018, 9. [Google Scholar] [CrossRef]
  68. Hildebrand, L.; Stange, K.; Deichsel, A.; Gossen, M.; Seemann, P. The Fibrodysplasia Ossificans Progressiva (FOP) mutation p.R206H in ACVR1 confers an altered ligand response. Cell. Signal. 2017, 29, 23–30. [Google Scholar] [CrossRef] [Green Version]
  69. Barruet, E.; Morales, B.M.; Lwin, W.; White, M.P.; Theodoris, C.V.; Kim, H.; Urrutia, A.; Wong, S.A.; Srivastava, D.; Hsiao, E.C. The ACVR1 R206H mutation found in fibrodysplasia ossificans progressiva increases human induced pluripotent stem cell-derived endothelial cell formation and collagen production through BMP-mediated SMAD1/5/8 signaling. Stem Cell Res. Ther. 2016, 7. [Google Scholar] [CrossRef]
  70. Antebi, Y.E.; Linton, J.M.; Klumpe, H.; Bintu, B.; Gong, M.; Su, C.; McCardell, R.; Elowitz, M.B. Combinatorial Signal Perception in the BMP Pathway. Cell 2017, 170, 1184–1196.e24. [Google Scholar] [CrossRef] [Green Version]
  71. Fiori, J.L.; Billings, P.C.; Serrano De La Peña, L.; Kaplan, F.S.; Shore, E.M. Dysregulation of the BMP-p38 MAPK signaling pathway in cells from patients with fibrodysplasia ossificans progressiva (FOP). J. Bone Miner. Res. 2006, 21, 902–909. [Google Scholar]
  72. Rigueur, D.; Brugger, S.; Anbarchian, T.; Kim, J.K.; Lee, Y.; Lyons, K.M. The type i BMP receptor ACVR1/ALK2 is required for chondrogenesis during development. J. Bone Miner. Res. 2015, 30, 733–741. [Google Scholar] [CrossRef]
  73. Qureshi, A.T.; Dey, D.; Sanders, E.M.; Seavey, J.G.; Tomasino, A.M.; Moss, K.; Wheatley, B.; Cholok, D.; Loder, S.; Li, J.; et al. Inhibition of Mammalian Target of Rapamycin Signaling with Rapamycin Prevents Trauma-Induced Heterotopic Ossification. Am. J. Pathol. 2017, 187, 2536–2545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hino, K.; Horigome, K.; Nishio, M.; Komura, S.; Nagata, S.; Zhao, C.; Jin, Y.; Kawakami, K.; Yamada, Y.; Ohta, A.; et al. Activin-A enhances mTOR signaling to promote aberrant chondrogenesis in fibrodysplasia ossificans progressiva. J. Clin. Investig. 2017, 127, 3339–3352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Hino, K.; Zhao, C.; Horigome, K.; Nishio, M.; Okanishi, Y.; Nagata, S.; Komura, S.; Yamada, Y.; Toguchida, J.; Ohta, A.; et al. An mTOR Signaling Modulator Suppressed Heterotopic Ossification of Fibrodysplasia Ossificans Progressiva. Stem Cell Rep. 2018, 11, 1106–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Gámez, B.; Rodríguez-Carballo, E.; Graupera, M.; Rosa, J.L.; Ventura, F. Class I PI-3-Kinase Signaling Is Critical for Bone Formation Through Regulation of SMAD1 Activity in Osteoblasts. J. Bone Miner. Res. 2016, 31, 1617–1630. [Google Scholar] [CrossRef] [Green Version]
  77. Valer, J.A.; Sánchez-de-Diego, C.; Gámez, B.; Mishina, Y.; Rosa, J.L.; Ventura, F. Inhibition of phosphatidylinositol 3-kinase α ( PI 3Kα) prevents heterotopic ossification. EMBO Mol. Med. 2019, 11. [Google Scholar] [CrossRef]
  78. Haupt, J.; Stanley, A.; McLeod, C.M.; Cosgrove, B.D.; Culbert, A.L.; Wang, L.; Mourkioti, F.; Mauck, R.L.; Shore, E.M. ACVR1 R206H FOP mutation alters mechanosensing and tissue stiffness during heterotopic ossification. Mol. Biol. Cell 2018, 30, 17–29. [Google Scholar] [CrossRef]
  79. Lin, S.; Svoboda, K.K.H.; Feng, J.Q.; Jiang, X. The biological function of type i receptors of bone morphogenetic protein in bone. Bone Res. 2016, 4. [Google Scholar] [CrossRef]
  80. Zhang, D.; Schwarz, E.M.; Rosier, R.N.; Zuscik, M.J.; Puzas, J.E.; O’Keefe, R.J. ALK2 functions as a BMP type I receptor and induces Indian hedgehog in chondrocytes during skeletal development. J. Bone Miner. Res. 2003, 18, 1593–1604. [Google Scholar] [CrossRef]
  81. Yu, P.B.; Deng, D.Y.; Lai, C.S.; Hong, C.C.; Cuny, G.D.; Bouxsein, M.L.; Hong, D.W.; McManus, P.M.; Katagiri, T.; Sachidanandan, C.; et al. BMP type I receptor inhibition reduces heterotopic ossification. Nat. Med. 2008, 14, 1363–1369. [Google Scholar] [CrossRef]
  82. Kaplan, J.; Kaplan, F.S.; Shore, E.M. Restoration of normal BMP signaling levels and osteogenic differentiation in FOP mesenchymal progenitor cells by mutant allele-specific targeting. Gene Ther. 2012, 19, 786–790. [Google Scholar] [CrossRef]
  83. Shi, S.T.; Cai, J.; de Gorter, D.J.J.; Sanchez-Duffhues, G.; Kemaladewi, D.U.; Hoogaars, W.M.H.; Aartsma-Rus, A.; ’t Hoen, P.A.C.; ten Dijke, P. Antisense-Oligonucleotide Mediated Exon Skipping in Activin-Receptor-Like Kinase 2: Inhibiting the Receptor That Is Overactive in Fibrodysplasia Ossificans Progressiva. PLoS ONE 2013, 8, e69096. [Google Scholar] [CrossRef] [PubMed]
  84. Shi, C.; Mandair, G.S.; Zhang, H.; Vanrenterghem, G.G.; Ridella, R.; Takahashi, A.; Zhang, Y.; Kohn, D.H.; Morris, M.D.; Mishina, Y.; et al. Bone morphogenetic protein signaling through ACVR1 and BMPR1A negatively regulates bone mass along with alterations in bone composition. J. Struct. Biol. 2018, 201, 237–246. [Google Scholar] [CrossRef] [PubMed]
  85. Kamiya, N.; Kaartinen, V.M.; Mishina, Y. Loss-of-function of ACVR1 in osteoblasts increases bone mass and activates canonical Wnt signaling through suppression of Wnt inhibitors SOST and DKK1. Biochem. Biophys. Res. Commun. 2011, 414, 326–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Zhang, X.; Liu, Q.; Zhao, H.; Hu, Y.; Liu, C.; Yan, G.; Li, D.; Mishina, Y.; Shi, C.; Sun, H. ACVR1 is essential for periodontium development and promotes alveolar bone formation. Arch. Oral Biol. 2018, 95, 108–117. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, X.; Shi, C.; Zhao, H.; Zhou, Y.; Hu, Y.; Yan, G.; Liu, C.; Li, D.; Hao, X.; Mishina, Y.; et al. Distinctive role of ACVR1 in dentin formation: requirement for dentin thickness in molars and prevention of osteodentin formation in incisors of mice. J. Mol. Histol. 2019, 50, 43–61. [Google Scholar] [CrossRef] [PubMed]
  88. Gomez-Puerto, M.C.; Iyengar, P.V.; García de Vinuesa, A.; ten Dijke, P.; Sanchez-Duffhues, G. Bone morphogenetic protein receptor signal transduction in human disease. J. Pathol. 2019, 247, 9–20. [Google Scholar] [CrossRef]
  89. Wang, R.N.; Green, J.; Wang, Z.; Deng, Y.; Qiao, M.; Peabody, M.; Zhang, Q.; Ye, J.; Yan, Z.; Denduluri, S.; et al. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis. 2014, 1, 87–105. [Google Scholar] [CrossRef] [Green Version]
  90. Wang, J.; Sridurongrit, S.; Dudas, M.; Thomas, P.; Nagy, A.; Schneider, M.D.; Epstein, J.A.; Kaartinen, V. Atrioventricular cushion transformation is mediated by ALK2 in the developing mouse heart. Dev. Biol. 2005, 286, 299–310. [Google Scholar] [CrossRef] [Green Version]
  91. Thomas, P.S.; Rajderkar, S.; Lane, J.; Mishina, Y.; Kaartinen, V. AcvR1-mediated BMP signaling in second heart field is required for arterial pole development: Implications for myocardial differentiation and regional identity. Dev. Biol. 2014, 390, 191–207. [Google Scholar] [CrossRef] [Green Version]
  92. Kaartinen, V.; Dudas, M.; Nagy, A.; Sridurongrit, S.; Lu, M.M.; Epstein, J.A. Cardiac outflow tract defects in mice lacking ALK2 in neural crestcells. Development 2004, 131, 3481–3490. [Google Scholar] [CrossRef]
  93. Thomas, P.S.; Sridurongrit, S.; Ruiz-Lozano, P.; Kaartinen, V. Deficient signaling via Alk2 (Acvr1) leads to Bicuspid aortic valve development. PLoS ONE 2012, 7, e35539. [Google Scholar] [CrossRef] [PubMed]
  94. Shahid, M.; Spagnolli, E.; Ernande, L.; Thoonen, R.; Kolodziej, S.A.; Leyton, P.A.; Cheng, J.; Tainsh, R.E.T.; Mayeur, C.; Rhee, D.K.; et al. BMP type I receptor ALK2 is required for angiotensin II-induced cardiac hypertrophy. Am. J. Physiol. Circ. Physiol. 2016, 310, H984–H994. [Google Scholar] [CrossRef] [PubMed]
  95. Smith, K.A.; Joziasse, I.C.; Chocron, S.; Van Dinther, M.; Guryev, V.; Verhoeven, M.C.; Rehmann, H.; Van Der Smagt, J.J.; Doevendans, P.A.; Cuppen, E.; et al. Dominant-negative alk2 allele associates with congenital heart defects. Circulation 2009, 119, 3062–3069. [Google Scholar] [CrossRef] [PubMed]
  96. Joziasse, I.C.; Smith, K.A.; Chocron, S.; Van Dinther, M.; Guryev, V.; Van De Smagt, J.J.; Cuppen, E.; Ten Dijke, P.; Mulder, B.J.M.; Maslen, C.L.; et al. ALK2 mutation in a patient with Down’s syndrome and a congenital heart defect. Eur. J. Hum. Genet. 2011, 19, 389–393. [Google Scholar] [CrossRef] [PubMed]
  97. Li, A.H.; Hanchard, N.A.; Furthner, D.; Fernbach, S.; Azamian, M.; Nicosia, A.; Rosenfeld, J.; Muzny, D.; D’Alessandro, L.C.A.; Morris, S.; et al. Whole exome sequencing in 342 congenital cardiac left sided lesion cases reveals extensive genetic heterogeneity and complex inheritance patterns. Genome Med. 2017, 9. [Google Scholar] [CrossRef] [PubMed]
  98. Zhu, H.; Kavsak, P.; Abdollah, S.; Wrana, J.L.; Thomsen, G.H. A SMAD ubiquitin ligase targets the BMP pathway and affects embryonic pattern formation. Nature 1999, 400, 687–693. [Google Scholar] [CrossRef]
  99. Kussmaul, W.G.; Esmail, A.N.; Sagar, Y.; Ross, J.; Gregory, S.; Kaplan, F.S. Pulmonary and Cardiac Function in Advanced Fibrodysplasia Ossificans Progressiva. Clin. Orthop. Relat. Res. 1998, 346, 104–109. [Google Scholar] [CrossRef]
  100. Marseglia, L.; D’Angelo, G.; Manti, S.; Manganaro, A.; Calabrò, M.P.; Salpietro, C.; Gitto, E. Fibrodysplasia ossificans progressiva in a newborn with cardiac involvement. Pediatr. Int. 2015, 57, 719–721. [Google Scholar] [CrossRef]
  101. Knight, P.G.; Glister, C. TGF-β superfamily members and ovarian follicle development. Reproduction 2006, 132, 191–206. [Google Scholar] [CrossRef]
  102. Shimasaki, S.; Moore, R.K.; Otsuka, F.; Erickson, G.F. The Bone Morphogenetic Protein System in Mammalian Reproduction. Endocr. Rev. 2004, 25, 72–101. [Google Scholar] [CrossRef]
  103. Visser, J.A.; Olaso, R.; Verhoef-Post, M.; Kramer, P.; Themmen, A.P.N.; Ingraham, H.A. The Serine/Threonine Transmembrane Receptor ALK2 Mediates Müllerian Inhibiting Substance Signaling. Mol. Endocrinol. 2014, 15, 936–945. [Google Scholar]
  104. Clarke, T.R.; Hoshiya, Y.; Yi, S.E.; Liu, X.; Lyons, K.M.; Donahoe, P.K. Müllerian Inhibiting Substance Signaling Uses a Bone Morphogenetic Protein (BMP)-Like Pathway Mediated by ALK2 and Induces Smad6 Expression. Mol. Endocrinol. 2014, 15, 946–959. [Google Scholar] [CrossRef]
  105. Gouédard, L.; Chen, Y.G.; Thevenet, L.; Racine, C.; Borie, S.; Lamarre, I.; Josso, N.; Massaguè, J.; Di Clemente, N. Engagement of bone morphogenetic protein type IB receptor and Smad1 signaling by anti-Mullerian hormone and its type II receptor. J. Biol. Chem. 2000, 275, 27973–27978. [Google Scholar] [CrossRef] [PubMed]
  106. Jamin, S.P.; Arango, N.A.; Mishina, Y.; Hanks, M.C.; Behringer, R.R. Genetic studies of the AMH/MIS signaling pathway for Müllerian duct regression. Mol. Cell. Endocrinol. 2003, 211, 15–19. [Google Scholar]
  107. Van Houten, E.L.A.F.; Themmen, A.P.N.; Visser, J.A. Hormone anti-müllérienne (AMH): Régulateur et marqueur de la fonction ovarienne. Ann. Endocrinol. (Paris) 2010, 71, 191–197. [Google Scholar] [CrossRef] [PubMed]
  108. Durlinger, A.L.L.; Gruijters, M.J.G.; Kramer, P.; Karels, B.; Kumar, T.R.; Matzuk, M.M.; Rose, U.M.; De Jong, F.H.; Uilenbroek, J.T.J.; Grootegoed, J.A.; et al. Anti-Müllerian hormone attenuates the effects of FSH on follicle development in the mouse ovary. Endocrinology 2001, 142, 4891–4899. [Google Scholar] [CrossRef] [PubMed]
  109. Otsuka, F.; Moore, R.K.; Shimasaki, S. Biological Function and Cellular Mechanism of Bone Morphogenetic Protein-6 in the Ovary. J. Biol. Chem. 2001, 276, 32889–32895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Lee, W.S.; Yoon, S.J.; Yoon, T.K.; Cha, K.Y.; Lee, S.H.; Shimasaki, S.; Lee, S.; Lee, K.A. Effects of bone morphogenetic protein-7 (BMP-7) on primordial follicular growth in the mouse ovary. Mol. Reprod. Dev. 2004, 69, 159–163. [Google Scholar] [CrossRef]
  111. Kevenaar, M.E.; Themmen, A.P.N.; Van Kerkwijk, A.J.; Valkenburg, O.; Uitterlinden, A.G.; De Jong, F.H.; Laven, J.S.E.; Visser, J.A. Variants in the ACVR1 gene are associated with AMH levels in women with polycystic ovary syndrome. Hum. Reprod. 2009, 24, 241–249. [Google Scholar] [CrossRef]
  112. Slattery, M.L.; John, E.M.; Torres-Mejia, G.; Herrick, J.S.; Giuliano, A.R.; Baumgartner, K.B.; Hines, L.M.; Wolff, R.K. Genetic variation in bone morphogenetic proteins and breast cancer risk in hispanic and non-hispanic white women: The breast cancer health disparities study. Int. J. Cancer 2013, 132, 2928–2939. [Google Scholar] [CrossRef]
  113. Da Silveira, J.C.; Carnevale, E.M.; Winger, Q.A.; Bouma, G.J. Regulation of ACVR1 and ID2 by cell-secreted exosomes during follicle maturation in the mare. Reprod. Biol. Endocrinol. 2014, 12. [Google Scholar] [CrossRef]
  114. Bai, L.; Chang, H.M.; Cheng, J.C.; Chu, G.; Leung, P.C.K.; Yang, G. ALK2/ALK3-BMPR2/ACVR2A Mediate BMP2-induced downregulation of pentraxin 3 expression in human granulosa-lutein cells. Endocrinology 2017, 158, 3501–3511. [Google Scholar] [CrossRef] [PubMed]
  115. Wu, Y.T.; Chang, H.M.; Huang, H.F.; Sheng, J.Z.; Leung, P.C.K. Bone morphogenetic protein 2 regulates cell-cell communication by down-regulating connexin43 expression in luteinized human granulosa cells. Mol. Hum. Reprod. 2017, 23, 155–165. [Google Scholar] [CrossRef] [PubMed]
  116. Lee, K.Y.; Jeong, J.-W.; Wang, J.; Ma, L.; Martin, J.F.; Tsai, S.Y.; Lydon, J.P.; DeMayo, F.J. Bmp2 Is Critical for the Murine Uterine Decidual Response. Mol. Cell. Biol. 2007, 27, 5468–5478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Clementi, C.; Tripurani, S.K.; Large, M.J.; Edson, M.A.; Creighton, C.J.; Hawkins, S.M.; Kovanci, E.; Kaartinen, V.; Lydon, J.P.; Pangas, S.A.; et al. Activin-Like Kinase 2 Functions in Peri-implantation Uterine Signaling in Mice and Humans. PLoS Genet. 2013, 9. [Google Scholar] [CrossRef] [PubMed]
  118. Komatsu, Y.; Scott, G.; Nagy, A.; Kaartinen, V.; Mishina, Y. BMP type I receptor ALK2 is essential for proper patterning at late gastrulation during mouse embryogenesis. Dev. Dyn. 2007, 236, 512–517. [Google Scholar] [CrossRef] [PubMed]
  119. Chuva De Sousa Lopes, S.M.; Roelen, B.A.J.; Monteiro, R.M.; Emmens, R.; Lin, H.Y.; Li, E.; Lawson, K.A.; Mummery, C.L. BMP signaling mediated by ALK2 in the visceral endoderm is necessary for the generation of primordial germ cells in the mouse embryo. Genes Dev. 2004, 18, 1838–1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Kitterman, J.A.; Strober, J.B.; Kan, L.; Rocke, D.M.; Cali, A.; Peeper, J.; Snow, J.; Delai, P.L.R.; Morhart, R.; Pignolo, R.J.; et al. Neurological symptoms in individuals with fibrodysplasia ossificans progressiva. J. Neurol. 2012, 259, 2636–2643. [Google Scholar] [CrossRef] [Green Version]
  121. Bond, A.M.; Bhalala, O.G.; Kessler, J.A. The dynamic role of bone morphogenetic proteins in neural stem cell fate and maturation. Dev. Neurobiol. 2012, 72, 1068–1084. [Google Scholar] [CrossRef] [Green Version]
  122. Angley, C.; Kumar, M.; Dinsio, K.J.; Hall, A.K.; Siegel, R.E. Signaling by Bone Morphogenetic Proteins and Smad1 Modulates the Postnatal Differentiation of Cerebellar Cells. J. Neurosci. 2003, 23, 260–268. [Google Scholar] [CrossRef] [Green Version]
  123. Gomes, W.A.; Mehler, M.F.; Kessler, J.A. Transgenic overexpression of BMP4 increases astroglial and decreases oligodendroglial lineage commitment. Dev. Biol. 2003, 255, 164–177. [Google Scholar] [CrossRef] [Green Version]
  124. Samanta, J. Interactions between ID and OLIG proteins mediate the inhibitory effects of BMP4 on oligodendroglial differentiation. Development 2004, 131, 4131–4142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Gross, R.E.; Mehler, M.F.; Mabie, P.C.; Zang, Z.; Santschi, L.; Kessler, J.A. Bone morphogenetic proteins promote astroglial lineage commitment by mammalian subventricular zone progenitor cells. Neuron 1996, 17, 595–606. [Google Scholar] [CrossRef]
  126. Mehler, M.F.; Mabie, P.C.; Zhu, G.; Gokhan, S.; Kessler, J.A. Developmental changes in progenitor cell responsiveness to gone morphogenetic; Proteins differentially modulate progressive CNS lineage fate. Dev. Neurosci. 2000, 22, 74–85. [Google Scholar] [CrossRef] [PubMed]
  127. Gobeske, K.T.; Das, S.; Bonaguidi, M.A.; Weiss, C.; Radulovic, J.; Disterhoft, J.F.; Kessler, J.A. BMP signaling mediates effects of exercise on hippocampal neurogenesis and cognition in mice. PLoS ONE 2009, 4, e7506. [Google Scholar] [CrossRef]
  128. Bertamino, M.; Severino, M.; Schiaffino, M.C.; Garrè, M.L.; Bocciardi, R.; Ravazzolo, R.; Rossi, A.; Di Rocco, M. New insights into central nervous system involvement in FOP: Case report and review of the literature. Am. J. Med. Genet. Part A 2015, 167, 2817–2821. [Google Scholar] [CrossRef]
  129. Severino, M.; Bertamino, M.; Tortora, D.; Morana, G.; Uccella, S.; Bocciardi, R.; Ravazzolo, R.; Rossi, A.; Di Rocco, M. Novel asymptomatic CNS findings in patients with ACVR1/ALK2 mutations causing fibrodysplasia ossificans progressiva. J. Med. Genet. 2016, 53, 859–864. [Google Scholar] [CrossRef]
  130. Shiva Kumar, R.; Keerthiraj, B.; Kesavadas, C. Teaching NeuroImages: MRI in fibrodysplasia ossificans progressiva. Neurology 2010, 74. [Google Scholar] [CrossRef]
  131. Kan, L.; Kitterman, J.A.; Procissi, D.; Chakkalakal, S.; Peng, C.Y.; McGuire, T.L.; Goldsby, R.E.; Pignolo, R.J.; Shore, E.M.; Kaplan, F.S.; et al. CNS demyelination in fibrodysplasia ossificans progressiva. J. Neurol. 2012, 259, 2644–2655. [Google Scholar] [CrossRef] [Green Version]
  132. Cate, H.S.; Sabo, J.K.; Merlo, D.; Kemper, D.; Aumann, T.D.; Robinson, J.; Merson, T.D.; Emery, B.; Perreau, V.M.; Kilpatrick, T.J. Modulation of bone morphogenic protein signalling alters numbers of astrocytes and oligodendroglia in the subventricular zone during cuprizone-induced demyelination. J. Neurochem. 2010, 115, 11–22. [Google Scholar] [CrossRef]
  133. Qi, X.; Li, T.-G.; Hao, J.; Hu, J.; Wang, J.; Simmons, H.; Miura, S.; Mishina, Y.; Zhao, G.-Q. BMP4 supports self-renewal of embryonic stem cells by inhibiting mitogen-activated protein kinase pathways. Proc. Natl. Acad. Sci. USA 2004, 101, 6027–6032. [Google Scholar] [CrossRef] [Green Version]
  134. Ying, Q.L.; Nichols, J.; Chambers, I.; Smith, A. BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell 2003, 115, 281–292. [Google Scholar] [CrossRef]
  135. Xu, R.H.; Peck, R.M.; Li, D.S.; Feng, X.; Ludwig, T.; Thomson, J.A. Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells. Nat. Methods 2005, 2, 185–190. [Google Scholar] [CrossRef]
  136. Samavarchi-Tehrani, P.; Golipour, A.; David, L.; Sung, H.K.; Beyer, T.A.; Datti, A.; Woltjen, K.; Nagy, A.; Wrana, J.L. Functional genomics reveals a BMP-Driven mesenchymal-to-Epithelial transition in the initiation of somatic cell reprogramming. Cell Stem Cell 2010, 7, 64–77. [Google Scholar] [CrossRef] [PubMed]
  137. Hamasaki, M.; Hashizume, Y.; Yamada, Y.; Katayama, T.; Hohjoh, H.; Fusaki, N.; Nakashima, Y.; Furuya, H.; Haga, N.; Takami, Y.; et al. Pathogenic mutation of ALK2 inhibits induced pluripotent stem cell reprogramming and maintenance: Mechanisms of reprogramming and strategy for drug identification. Stem Cells 2012, 30, 2437–2449. [Google Scholar] [CrossRef] [PubMed]
  138. Matsumoto, Y.; Ikeya, M.; Hino, K.; Horigome, K.; Fukuta, M.; Watanabe, M.; Nagata, S.; Yamamoto, T.; Otsuka, T.; Toguchida, J. New protocol to optimize iPS cells for genome analysis of fibrodysplasia ossificans progressiva. Stem Cells 2015, 33, 1730–1742. [Google Scholar] [CrossRef]
  139. Cai, J.; Orlova, V.V.; Cai, X.; Eekhoff, E.M.W.; Zhang, K.; Pei, D.; Pan, G.; Mummery, C.L.; Ten Dijke, P. Induced Pluripotent Stem Cells to Model Human Fibrodysplasia Ossificans Progressiva. Stem Cell Rep. 2015, 5, 963–970. [Google Scholar] [CrossRef] [Green Version]
  140. Kim, B.Y.; Jeong, S.K.; Lee, S.Y.; Lee, S.M.; Gweon, E.J.; Ahn, H.; Kim, J.; Chung, S.K. Concurrent progress of reprogramming and gene correction to overcome therapeutic limitation of mutant ALK2-iPSC. Exp. Mol. Med. 2016, 48. [Google Scholar] [CrossRef]
  141. Hayashi, Y.; Hsiao, E.C.; Sami, S.; Lancero, M.; Schlieve, C.R.; Nguyen, T.; Yano, K.; Nagahashi, A.; Ikeya, M.; Matsumoto, Y.; et al. BMP-SMAD-ID promotes reprogramming to pluripotency by inhibiting p16/INK4A-dependent senescence. Proc. Natl. Acad. Sci. USA 2016, 113, 13057–13062. [Google Scholar] [CrossRef] [Green Version]
  142. López-Rovira, T.; Chalaux, E.; Massagué, J.; Rosa, J.L.; Ventura, F. Direct binding of Smad1 and Smad4 to two distinct motifs mediates bone morphogenetic protein-specific transcriptional activation of Id1 gene. J. Biol. Chem. 2002, 277, 3176–3185. [Google Scholar] [CrossRef]
  143. Alani, R.M.; Young, A.Z.; Shifflett, C.B. Id1 regulation of cellular senescence through transcriptional repression of p16/Ink4a. Proc. Natl. Acad. Sci. USA 2001, 98, 7812–7816. [Google Scholar] [CrossRef] [Green Version]
  144. Banito, A.; Rashid, S.T.; Acosta, J.C.; Li, S.D.; Pereira, C.F.; Geti, I.; Pinho, S.; Silva, J.C.; Azuara, V.; Walsh, M.; et al. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 2009, 23, 2134–2139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Zadeh, G.; Aldape, K. ACVR1 mutations and the genomic landscape of pediatric diffuse glioma. Nat. Genet. 2014, 46, 421–422. [Google Scholar] [CrossRef] [PubMed]
  146. Buczkowicz, P.; Bartels, U.; Bouffet, E.; Becher, O.; Hawkins, C. Histopathological spectrum of paediatric diffuse intrinsic pontine glioma: diagnostic and therapeutic implications. Acta Neuropathol. 2014, 128, 573–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Buczkowicz, P.; Hawkins, C. Pathology, Molecular Genetics, and Epigenetics of Diffuse Intrinsic Pontine Glioma. Front. Oncol. 2015, 5. [Google Scholar] [CrossRef] [PubMed]
  148. Villanueva, M.T. Genetics: ACVR1 mutations-A key piece in paediatric diffuse glioma. Nat. Rev. Clin. Oncol. 2014, 11, 300. [Google Scholar] [CrossRef]
  149. Taylor, K.R.; Mackay, A.; Truffaux, N.; Butterfield, Y.S.; Morozova, O.; Philippe, C.; Castel, D.; Grasso, C.S.; Vinci, M.; Carvalho, D.; et al. Recurrent activating ACVR1 mutations in diffuse intrinsic pontine glioma. Nat. Genet. 2014, 46, 457–461. [Google Scholar] [CrossRef] [PubMed]
  150. Hoeman, C.M.; Cordero, F.J.; Hu, G.; Misuraca, K.; Romero, M.M.; Cardona, H.J.; Nazarian, J.; Hashizume, R.; McLendon, R.; Yu, P.; et al. ACVR1 R206H cooperates with H3.1K27M in promoting diffuse intrinsic pontine glioma pathogenesis. Nat. Commun. 2019, 10. [Google Scholar] [CrossRef]
  151. Wu, G.; Diaz, A.K.; Paugh, B.S.; Rankin, S.L.; Ju, B.; Li, Y.; Zhu, X.; Qu, C.; Chen, X.; Zhang, J.; et al. The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma. Nat. Genet. 2014, 46, 444–450. [Google Scholar]
  152. Buczkowicz, P.; Hoeman, C.; Rakopoulos, P.; Pajovic, S.; Letourneau, L.; Dzamba, M.; Morrison, A.; Lewis, P.; Bouffet, E.; Bartels, U.; et al. Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nat. Genet. 2014, 46, 451–456. [Google Scholar] [CrossRef] [Green Version]
  153. Fontebasso, A.M.; Papillon-Cavanagh, S.; Schwartzentruber, J.; Nikbakht, H.; Gerges, N.; Fiset, P.O.; Bechet, D.; Faury, D.; De Jay, N.; Ramkissoon, L.A.; et al. Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma. Nat. Genet. 2014, 46, 462–466. [Google Scholar] [CrossRef]
  154. Taylor, K.R.; Vinci, M.; Bullock, A.N.; Jones, C. ACVR1 mutations in DIPG: Lessons learned from FOP. Cancer Res. 2014, 74, 4565–4570. [Google Scholar] [CrossRef] [PubMed]
  155. Carro, M.S.; Lim, W.K.; Alvarez, M.J.; Bollo, R.J.; Zhao, X.; Snyder, E.Y.; Sulman, E.P.; Anne, S.L.; Doetsch, F.; Colman, H.; et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature 2010, 463, 318–325. [Google Scholar] [CrossRef] [PubMed]
  156. Nikbakht, H.; Panditharatna, E.; Mikael, L.G.; Li, R.; Gayden, T.; Osmond, M.; Ho, C.Y.; Kambhampati, M.; Hwang, E.I.; Faury, D.; et al. Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma. Nat. Commun. 2016, 7. [Google Scholar] [CrossRef] [PubMed]
  157. Fruman, D.A.; Rommel, C. PI3K and cancer: Lessons, challenges and opportunities. Nat. Rev. Drug Discov. 2014, 13, 140–156. [Google Scholar] [CrossRef] [PubMed]
  158. Han, H.J.; Jain, P.; Resnick, A.C. Shared ACVR1 mutations in FOP and DIPG: Opportunities and challenges in extending biological and clinical implications across rare diseases. Bone 2018, 109, 91–100. [Google Scholar] [CrossRef] [PubMed]
  159. Raja, E.; Komuro, A.; Tanabe, R.; Sakai, S.; Ino, Y.; Saito, N.; Todo, T.; Morikawa, M.; Aburatani, H.; Koinuma, D.; et al. Bone morphogenetic protein signaling mediated by ALK-2 and DLX2 regulates apoptosis in glioma-initiating cells. Oncogene 2017, 36, 4963–4974. [Google Scholar] [CrossRef]
  160. Ghanem, N.; Andrusiak, M.G.; Svoboda, D.; Al Lafi, S.M.; Julian, L.M.; McClellan, K.A.; De Repentigny, Y.; Kothary, R.; Ekker, M.; Blais, A.; et al. The Rb/E2F Pathway Modulates Neurogenesis through Direct Regulation of the Dlx1/Dlx2 Bigene Cluster. J. Neurosci. 2012, 32, 8219–8230. [Google Scholar] [CrossRef] [Green Version]
  161. Panganiban, G.; Rubenstein, J.L.R. Developmental functions of the Distal-less/Dlx homeobox genes. Development 2002, 129, 4371–4386. [Google Scholar]
  162. Wang, T.-H.; Chao, A.; Tsai, C.-L.; Chang, C.-L.; Chen, S.-H.; Lee, Y.-S.; Chen, J.-K.; Lin, Y.-J.; Chang, P.-Y.; Wang, C.-J.; et al. Stress-induced Phosphoprotein 1 as a Secreted Biomarker for Human Ovarian Cancer Promotes Cancer Cell Proliferation. Mol. Cell. Proteomics 2010, 9, 1873–1884. [Google Scholar] [CrossRef] [Green Version]
  163. Tsai, C.L.; Tsai, C.N.; Lin, C.Y.; Chen, H.W.; Lee, Y.S.; Chao, A.; Wang, T.H.; Wang, H.S.; Lai, C.H. Secreted Stress-Induced Phosphoprotein 1 Activates the ALK2-SMAD Signaling Pathways and Promotes Cell Proliferation of Ovarian Cancer Cells. Cell Rep. 2012, 2, 283–293. [Google Scholar] [CrossRef] [Green Version]
  164. Herrera, B.; Van Dinther, M.; Ten Dijke, P.; Inman, G.J. Autocrine bone morphogenetic protein-9 signals through activin receptor-like kinase-2/Smad1/Smad4 to promote ovarian cancer cell proliferation. Cancer Res. 2009, 69, 9254–9262. [Google Scholar] [CrossRef] [PubMed]
  165. La Marca, A.; Volpe, A. The Anti-Mullerian hormone and ovarian cancer. Hum. Reprod. Update 2007, 13, 265–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Anttonen, M.; Färkkilä, A.; Tauriala, H.; Kauppinen, M.; MacLaughlin, D.T.; Unkila-Kallio, L.; Bützow, R.; Heikinheimo, M. Anti-Müllerian hormone inhibits growth of AMH type II receptor-positive human ovarian granulosa cell tumor cells by activating apoptosis. Lab. Investig. 2011, 91, 1605–1614. [Google Scholar] [CrossRef] [PubMed]
  167. Zhang, T.; Deng, L.; Xiong, Q.; Su, S.; Gu, J. Anti-müllerian hormone inhibits proliferation and induces apoptosis in epithelial ovarian cancer cells by regulating the cell cycle and decreasing the secretion of stem cell factor. Oncol. Lett. 2018, 16, 3260–3266. [Google Scholar]
  168. Masiakos, P.T.; MacLaughlin, D.T.; Maheswaran, S.; Teixeira, J.; Shah, P.C.; Kehas, D.J.; Kenneally, M.K.; Ha, T.U.; Fuller, A.F., Jr.; Dombkowski, D.M.; et al. Human ovarian cancer, cell lines, and primary ascites cells express the human Mullerian inhibiting substance (MIS) type II receptor, bind, and are responsive to MIS. Clin. Cancer Res. 1999, 5, 3488–3499. [Google Scholar]
  169. Stephen, A.E.; Pearsall, L.A.; Christian, B.P.; Donahoe, P.K.; Vacanti, J.P.; MacLaughlin, D.T. Highly purified Müllerian inhibiting substance inhibits human ovarian cancer in vivo. Clin. Cancer Res. 2002, 8, 2640–2646. [Google Scholar]
  170. Zhao, B.; Pritchard, J.R. Inherited Disease Genetics Improves the Identification of Cancer-Associated Genes. PLoS Genet. 2016, 12. [Google Scholar] [CrossRef]
  171. Tate, J.G.; Bamford, S.; Jubb, H.C.; Sondka, Z.; Beare, D.M.; Bindal, N.; Boutselakis, H.; Cole, C.G.; Creatore, C.; Dawson, E.; et al. COSMIC: The Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019, 47, D941–D947. [Google Scholar] [CrossRef]
  172. Zehir, A.; Benayed, R.; Shah, R.H.; Syed, A.; Middha, S.; Kim, H.R.; Srinivasan, P.; Gao, J.; Chakravarty, D.; Devlin, S.M.; et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 2017, 23, 703–713. [Google Scholar] [CrossRef]
  173. COSMIC The Catalogue Of Somatic Mutations In Cancer. Available online: https://cancer.sanger.ac.uk/cosmic (accessed on 26 September 2019).
  174. Nassiri, F.; Cusimano, M.D.; Scheithauer, B.W.; Rotondo, F.; Fazio, A.; Yousef, G.M.; Syro, L.V.; Kovacs, K.; Lloyd, R.V. Endoglin (CD105): A review of its role in angiogenesis and tumor diagnosis, progression and therapy. Anticancer Res. 2011, 31, 2283–2290. [Google Scholar]
  175. Craft, C.S.; Romero, D.; Vary, C.P.H.; Bergan, R.C. Endoglin inhibits prostate cancer motility via activation of the ALK2-Smad1 pathway. Oncogene 2007, 26, 7240–7250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Romero, D.; Terzic, A.; Conley, B.A.; Craft, C.S.; Jovanovic, B.; Bergan, R.C.; Vary, C.P.H. Endoglin phosphorylation by ALK2 contributes to the regulation of prostate cancer cell migration. Carcinogenesis 2010, 31, 359–366. [Google Scholar] [CrossRef] [PubMed]
  177. Olsen, O.E.; Wader, K.F.; Misund, K.; Våtsveen, T.K.; Rø, T.B.; Mylin, A.K.; Turesson, I.; Størdal, B.F.; Moen, S.H.; Standal, T.; et al. Bone morphogenetic protein-9 suppresses growth of myeloma cells by signaling through ALK2 but is inhibited by endoglin. Blood Cancer J. 2014, 4. [Google Scholar] [CrossRef] [PubMed]
  178. Rajagopal, R.; Dattilo, L.K.; Kaartinen, V.; Deng, C.X.; Umans, L.; Zwijsen, A.; Roberts, A.B.; Bottinger, E.P.; Beebe, D.C. Functions of the type 1 BMP receptor Acvr1 (Alk2) in lens development: Cell proliferation, terminal differentiation, and survival. Investig. Ophthalmol. Vis. Sci. 2008, 49, 4953–4960. [Google Scholar] [CrossRef] [PubMed]
  179. Wiley, L.A.; Rajagopal, R.; Dattilo, L.K.; Beebe, D.C. The tumor suppressor gene Trp53 protects the mouse lens against posterior subcapsular cataracts and the BMP receptor Acvr1 acts as a tumor suppressor in the lens. Dis. Model. Mech. 2011, 4, 484–495. [Google Scholar] [CrossRef] [PubMed]
  180. Kim, M.; Choi, O.; Pyo, S.; Choi, S.U.; Park, C.H. Identification of novel ALK2 inhibitors and their effect on cancer cells. Biochem. Biophys. Res. Commun. 2017, 492, 121–127. [Google Scholar] [CrossRef] [PubMed]
  181. Ambrosio, E.P.; Drigo, S.A.; Bérgamo, N.A.; Rosa, F.E.; Bertonha, F.B.; de Abreu, F.B.; Kowalski, L.P.; Rogatto, S.R. Recurrent copy number gains of ACVR1 and corresponding transcript overexpression are associated with survival in head and neck squamous cell carcinomas. Histopathology 2011, 59, 81–89. [Google Scholar] [CrossRef]
  182. Leemans, C.R.; Snijders, P.J.F.; Brakenhoff, R.H. The molecular landscape of head and neck cancer. Nat. Rev. Cancer 2018, 18, 269–282. [Google Scholar] [CrossRef]
  183. Bach, D.H.; Park, H.J.; Lee, S.K. The Dual Role of Bone Morphogenetic Proteins in Cancer. Mol. Ther. Oncolytics 2018, 8, 1–13. [Google Scholar] [CrossRef]
  184. Zhang, L.; Ye, Y.; Long, X.; Xiao, P.; Ren, X.; Yu, J. BMP signaling and its paradoxical effects in tumorigenesis and dissemination. Oncotarget 2016, 7. [Google Scholar] [CrossRef]
  185. Alarmo, E.L.; Kallioniemi, A. Bone morphogenetic proteins in breast cancer: Dual role in tumourigenesis? Endocr. Relat. Cancer 2010, 17. [Google Scholar] [CrossRef] [PubMed]
  186. Alarmo, E.L.; Pärssinen, J.; Ketolainen, J.M.; Savinainen, K.; Karhu, R.; Kallioniemi, A. BMP7 influences proliferation, migration, and invasion of breast cancer cells. Cancer Lett. 2009, 275, 35–43. [Google Scholar] [CrossRef] [PubMed]
  187. Yang, S.; Zhong, C.; Frenkel, B.; Reddi, A.H.; Roy-Burman, P. Diverse biological effect and Smad signaling of bone morphogenetic protein 7 in prostate tumor cells. Cancer Res. 2005, 65, 5769–5777. [Google Scholar] [CrossRef] [PubMed]
  188. Peng, J.; Yoshioka, Y.; Mandai, M.; Matsumura, N.; Baba, T.; Yamaguchi, K.; Hamanishi, J.; Kharma, B.; Murakami, R.; Abiko, K.; et al. The BMP signaling pathway leads to enhanced proliferation in serous ovarian cancer-A potential therapeutic target. Mol. Carcinog. 2016, 55, 335–345. [Google Scholar] [CrossRef] [PubMed]
  189. Gordon, K.J.; Kirkbride, K.C.; How, T.; Blobe, G.C. Bone morphogenetic proteins induce pancreatic cancer cell invasiveness through a Smad1-dependent mechanism that involves matrix metalloproteinase-2. Carcinogenesis 2009, 30, 238–248. [Google Scholar] [CrossRef] [PubMed]
  190. Huang, P.; Chen, A.; He, W.; Li, Z.; Zhang, G.; Liu, Z.; Liu, G.; Liu, X.; He, S.; Xiao, G.; et al. BMP-2 induces EMT and breast cancer stemness through Rb and CD44. Cell Death Discov. 2017, 3. [Google Scholar] [CrossRef] [PubMed]
  191. Wang, M.H.; Zhou, X.M.; Zhang, M.Y.; Shi, L.; Xiao, R.W.; Zeng, L.S.; Yang, X.Z.; Zheng, X.F.S.; Wang, H.Y.; Mai, S.J. BMP2 promotes proliferation and invasion of nasopharyngeal carcinoma cells via mTORC1 pathway. Aging (Albany. NY) 2017, 9, 1326–1340. [Google Scholar] [CrossRef] [Green Version]
  192. Wang, L.; Park, P.; Zhang, H.; La Marca, F.; Claeson, A.; Valdivia, J.; Lin, C.Y. BMP-2 inhibits the tumorigenicity of cancer stem cells in human osteosarcoma OS99-1 cell line. Cancer Biol. Ther. 2011, 11, 457–463. [Google Scholar] [CrossRef] [Green Version]
  193. Zhang, Y.; Chen, X.; Qiao, M.; Zhang, B.Q.; Wang, N.; Zhang, Z.; Liao, Z.; Zeng, L.; Deng, Y.; Deng, F.; et al. Bone morphogenetic protein 2 inhibits the proliferation and growth of human colorectal cancer cells. Oncol. Rep. 2014, 32, 1013–1020. [Google Scholar] [CrossRef] [Green Version]
  194. Beck, S.E.; Jung, B.H.; Fiorino, A.; Gomez, J.; Rosario, E.D.; Cabrera, B.L.; Huang, S.C.; Chow, J.Y.C.; Carethers, J.M. Bone morphogenetic protein signaling and growth suppression in colon cancer. Am. J. Physiol. Liver Physiol. 2006, 291, G135–G145. [Google Scholar] [CrossRef]
  195. Wang, L.; Park, P.; Zhang, H.; La Marca, F.; Claeson, A.; Than, K.; Rahman, S.; Lin, C.Y. BMP-2 inhibits tumor growth of human renal cell carcinoma and induces bone formation. Int. J. Cancer 2012, 131, 1941–1950. [Google Scholar] [CrossRef] [Green Version]
  196. Zheng, Y.; Wang, X.; Wang, H.; Yan, W.; Zhang, Q.; Chang, X. Bone morphogenetic protein 2 inhibits hepatocellular carcinoma growth and migration through downregulation of the PI3K/AKT pathway. Tumor Biol. 2014, 35, 5189–5198. [Google Scholar] [CrossRef]
Figure 1. Distribution of recurrent ACVR1 mutations in human pathologies. (A) Schematic representation of ACVR1 protein and observed mutations in fibrodysplasia ossificans progressive (FOP), diffuse intrinsic pontine glioma (DIPG), and congenital heart defects (CHD). Domains depicted from amino acids 1 to 509, including signal peptide (SP), ligand-binding domain, transmembrane domain (TM), glycine-serine-rich domain (GS), and protein kinase domain. (B) 3D crystal structure of GS and protein kinase domains of ACVR1 protein. Secondary structure elements shown as ribbon diagram. Mutations observed in both FOP and DIPG (red), mutations only observed in FOP (green), DIPG (yellow), or CHD (blue) are labelled in each mutation site. Highlighted amino acids are represented as space-filling spheres. Dorsomorphin, a type I BMP receptor inhibitor, (yellow stick) is represented in the ATP-pocket of ACVR1 kinase. 3H9R structure was obtained from PDB [50] and visualised using UCSF Chimera software [51]. (C) Proportion of FOP and DIPG cases affected by ACVR1 mutations.
Figure 1. Distribution of recurrent ACVR1 mutations in human pathologies. (A) Schematic representation of ACVR1 protein and observed mutations in fibrodysplasia ossificans progressive (FOP), diffuse intrinsic pontine glioma (DIPG), and congenital heart defects (CHD). Domains depicted from amino acids 1 to 509, including signal peptide (SP), ligand-binding domain, transmembrane domain (TM), glycine-serine-rich domain (GS), and protein kinase domain. (B) 3D crystal structure of GS and protein kinase domains of ACVR1 protein. Secondary structure elements shown as ribbon diagram. Mutations observed in both FOP and DIPG (red), mutations only observed in FOP (green), DIPG (yellow), or CHD (blue) are labelled in each mutation site. Highlighted amino acids are represented as space-filling spheres. Dorsomorphin, a type I BMP receptor inhibitor, (yellow stick) is represented in the ATP-pocket of ACVR1 kinase. 3H9R structure was obtained from PDB [50] and visualised using UCSF Chimera software [51]. (C) Proportion of FOP and DIPG cases affected by ACVR1 mutations.
Cells 08 01366 g001
Figure 2. Schematic representation of main ACVR1 signal transduction. ACVR1 is a type I receptor that can interact with type II receptors (BMPR2, ACVR2A, ACVR2B) to form transmembrane heterotetrameric receptor complexes. These complexes can bind ligands including BMPs (e.g., BMP6/7/9) and activins (e.g., activin A/B). Type II receptors can phosphorylate (P) and activate type I receptors, which can trigger canonical SMAD1/5/8 or non-SMAD signal transduction pathways. Phosphorylated SMAD1/5/8 interacts with SMAD4 to translocate into the nucleus and regulate transcription of SMAD target genes. ACVR1 signals through SMAD1/5/8 signalling pathway upon BMP ligand binding. In the presence of wild-type ACVR1 and a high BMPRII/ACVR2A-B expression ratio, activin A could antagonise SMAD1/5/8 signalling through SMAD2/3 and a possible ACVR2A-B competition for ACVR1 or ACVR1B complex formation. In the presence of mutated ACVR1R206H, or in the presence of low BMPRII/ACVR1A-B expression ratio in some cellular models, activin A could also signal through SMAD1/5/8.
Figure 2. Schematic representation of main ACVR1 signal transduction. ACVR1 is a type I receptor that can interact with type II receptors (BMPR2, ACVR2A, ACVR2B) to form transmembrane heterotetrameric receptor complexes. These complexes can bind ligands including BMPs (e.g., BMP6/7/9) and activins (e.g., activin A/B). Type II receptors can phosphorylate (P) and activate type I receptors, which can trigger canonical SMAD1/5/8 or non-SMAD signal transduction pathways. Phosphorylated SMAD1/5/8 interacts with SMAD4 to translocate into the nucleus and regulate transcription of SMAD target genes. ACVR1 signals through SMAD1/5/8 signalling pathway upon BMP ligand binding. In the presence of wild-type ACVR1 and a high BMPRII/ACVR2A-B expression ratio, activin A could antagonise SMAD1/5/8 signalling through SMAD2/3 and a possible ACVR2A-B competition for ACVR1 or ACVR1B complex formation. In the presence of mutated ACVR1R206H, or in the presence of low BMPRII/ACVR1A-B expression ratio in some cellular models, activin A could also signal through SMAD1/5/8.
Cells 08 01366 g002
Table 1. Role of ACVR1 in different cancer types.
Table 1. Role of ACVR1 in different cancer types.
Cancer TypeFunctionHallmarkRole in Cancer
Diffuse intrinsic pontine gliomaMutant ACVR1 (including R206H) increases cell proliferation in brainstem progenitors [150,152].Proliferative signallingOncogene
Mutant forms of ACVR1 lead to increased STAT3 signalling promoting mesenchymal profile and EMT [150].Epithelial-mesenchymal transition (EMT)Oncogene
GlioblastomaOverexpression of ACVR1 induced apoptosis in glioma-initiating cells and impaired tumour growth [159].Regulation of programmed cell deathTumour suppressor
Suppression of growth
Ovarian cancerACVR1-STIP1 interaction promotes human ovarian cancer cell proliferation [163].Proliferative signalling Oncogene
Autocrine BMP9 signalling through ACVR1 promotes ovarian cancer cell proliferation [164].Proliferative signalling Oncogene
AMH signalling reduces granulosa cell tumour growth, increasing apoptosis through type I ACVR1 and BMPR1A/B [166]. Regulation of programmed cell deathTumour suppressor
AMH inhibits cell proliferation of epithelial ovarian cancer cells [167].Proliferative signallingTumour suppressor
AMH inhibits human ovarian cancer cell proliferation [168,169].Proliferative signallingTumour suppressor
Prostate cancerACVR1-Smad1 phosphorylation upon endoglin interaction promotes cell migration [175].Invasion and metastasisTumour suppressor
ACVR1 phosphorylation of endoglin promotes cell migration [176].Invasion and metastasisOncogene
Multiple myelomaBMP9 signalling through ACVR1 induces apoptosis and inhibits cell growth [177].Proliferative signallingTumour suppressor
Regulation of programmed cell death
Eye lens tumourACVR1 signalling inhibits cell proliferation and impairs tumour growth [179].Proliferative signallingTumour suppressor
Suppression of growth
ErythroleukemiaOverexpression of ACVR1 increases cell proliferation in TF-1 cells [180].Proliferative signallingOncogene

Share and Cite

MDPI and ACS Style

Valer, J.A.; Sánchez-de-Diego, C.; Pimenta-Lopes, C.; Rosa, J.L.; Ventura, F. ACVR1 Function in Health and Disease. Cells 2019, 8, 1366. https://doi.org/10.3390/cells8111366

AMA Style

Valer JA, Sánchez-de-Diego C, Pimenta-Lopes C, Rosa JL, Ventura F. ACVR1 Function in Health and Disease. Cells. 2019; 8(11):1366. https://doi.org/10.3390/cells8111366

Chicago/Turabian Style

Valer, José Antonio, Cristina Sánchez-de-Diego, Carolina Pimenta-Lopes, Jose Luis Rosa, and Francesc Ventura. 2019. "ACVR1 Function in Health and Disease" Cells 8, no. 11: 1366. https://doi.org/10.3390/cells8111366

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop