Bone Morphogenetic Protein (BMP) signaling in development and human diseases.
Journal: 2017/February - Genes and Diseases
ISSN: 2352-3042
Abstract:
Bone Morphogenetic Proteins (BMPs) are a group of signaling molecules that belongs to the Transforming Growth Factor-β (TGF-β) superfamily of proteins. Initially discovered for their ability to induce bone formation, BMPs are now known to play crucial roles in all organ systems. BMPs are important in embryogenesis and development, and also in maintenance of adult tissue homeostasis. Mouse knockout models of various components of the BMP signaling pathway result in embryonic lethality or marked defects, highlighting the essential functions of BMPs. In this review, we first outline the basic aspects of BMP signaling and then focus on genetically manipulated mouse knockout models that have helped elucidate the role of BMPs in development. A significant portion of this review is devoted to the prominent human pathologies associated with dysregulated BMP signaling.
Relations:
Content
Citations
(124)
References
(275)
Genes
(3)
Affiliates
(3)
Similar articles
Articles by the same authors
Discussion board
Genes & Diseases. Aug/31/2014; 1(1): 87-105
Published online Jul/26/2014

Bone Morphogenetic Protein (BMP) signaling in development and human diseases

+11 authors

Abstract

Introduction

The activity of Bone Morphogenetic Proteins (BMPs) was first observed in the mid-1960s when it was discovered they could induce ectopic bone formation.1 It was not until the late 1980s, however, when the first BMPs were characterized and cloned, that individual BMPs could be studied biochemically.2 Many studies have since demonstrated the ability of BMPs to induce mesenchymal stem cells to differentiate into bone, confirming their role in bone and cartilage formation. BMPs are part of the Transforming Growth Factor-β (TGF-β) superfamily of proteins (Fig. 1A), which includes TGF-βs, activins, inhibins, Growth Differentiation Factors (GDFs), Glial Derived Neurotrophic Factors (GDNFs), Nodal, Lefty, and anti-Müllerian hormone. Since their initial discovery, they have been shown to affect a wide variety of cell types and processes beyond bone and osteogenesis. They are important morphogens in embryogenesis and development, and also regulate the maintenance of adult tissue homeostasis.

Figure 1

BMP Family and Signaling Pathways. (A) Phylogenetic analysis of the human BMP family members. The human BMP full-length precursor protein sequences and coding region sequences were analyzed using Phylogeny.fr. The branch length is proportional to the number of substitutions per site. (a) Phylogenetic analysis was performed using the amino acid sequence of human BMPs. (b) Phylogenetic analysis was carried out using the coding region of human BMPs. (B) BMPs signal via the canonical, Smad-dependent pathway or various non-canonical pathways. In the canonical pathway, BMPs initiate the signal transduction cascade by binding to type I or type II serine/threonine kinase receptors and forming a heterotetrameric complex. The constitutively active type II receptor then transphosphorylates the type I receptor, and the type I receptor phosphorylates the R-Smads (Smad1/5/8). Phosphorylated Smad1/5/8 associates with the co-Smad (Smad4), and the complex translocates to the nucleus where it further associates with coactivators or corepressors to regulate gene expression. Various non-canonical pathways, including the MAPK cascade, can also lead to regulation of gene expression. BMP signaling is modulated extracellularly (e.g., Noggin), intracellularly (e.g., FKBP12, microRNAs, phosphatases, and I-Smads), and by co-receptors in the plasma membrane (e.g., Endoglin).

Many processes in early development are dependent on BMP signaling for cell growth, apoptosis, and differentiation.3, 4, 5, 6 BMPs also play important roles in maintaining adult tissue homeostasis, such as the maintenance of joint integrity, the initiation of fracture repair, and vascular remodeling.7, 8, 9 Because of these diverse functions in all organ systems, it has been suggested that BMPs deserve to be called body morphogenetic proteins.10 Due to their ubiquitous expression and importance as regulators throughout the body, deficiency in BMP production or functionality usually leads to marked defects or severe pathologies (Fig. 2). Here, we review the mouse knockout models that have helped elucidate the role of BMPs in development and also emphasize some of the prominent human pathologies associated with deficiencies related to BMP signaling.

Figure 2

Representative members of the BMP signaling pathway that have been demonstrated to cause or be associated with human diseases. The mutations associated with human pathologies may be gain-of-function or loss-of-function. In some instances, higher or lower gene expression is correlated with disease. Because BMP signaling is involved in multiple organ systems, there are associated pathologies with most organ systems. Abbreviations: LOF, loss-of-function; GOF: gain-of-function; CAKUT, congenital anomalies of the kidney and urinary tract; CKD, chronic kidney disease; FOP, fibrodysplasia ossificans progressiva; OI, osteogenesis imperfecta; OA, osteoarthritis; A–M, anophthalmia–microphthalmia; PAH, pulmonary arterial hypertension; HHT, hereditary hemorrhagic telangiectasia; BE, Barrett esophagus; JP, juvenile polyposis.

Bmp signaling: canonical and non-canonical pathways

BMPs are synthesized as precursor proteins with an N-terminal signal peptide, a prodomain for folding and secretion, and a C-terminal mature peptide.11 Precursors are formed in the cytoplasm as dimeric pro-protein complexes, which are cleaved by pro-protein convertases to generate N- and C-terminal fragments. The C-terminal mature fragment is capable of binding to its receptor, with the non-covalently associated prodomain playing an important regulatory role.

BMPs can signal through both canonical and non-canonical pathways. In the canonical signaling pathway, they initiate the signal transduction cascade by binding to cell surface receptors and forming a heterotetrameric complex comprised of two dimers of type I and type II serine/threonine kinase receptors (Fig. 1B).12 Both receptor types have a short extracellular domain, a single transmembrane domain, and an intracellular domain with serine/threonine kinase activity. There are a total of seven type I receptors (ALK1-7) for the TGF-β family of ligands, three of which bind BMPs: type 1A BMP receptor (BMPR-1A or ALK3), type 1B BMP receptor (BMPR-1B or ALK6), and type 1A activin receptor (ActR-1A or ALK2).13 There are a total of four type II receptors for the TGF-β family, three of which are known to interact with BMPs: type 2 BMP receptor (BMPR-2), type 2 activin receptor (ActR-2A), and type 2B activin receptor (ActR-2B). While BMPR-1A, BMPR-1B, and BMPR-2 are specific to BMPs, ActR-1A, ActR-2A, and ActR-2B can function as receptors for activins, which are also members of the TGF-β superfamily. The mechanism of the heterotetrameric signaling complex formation can vary. For example, BMP6 and BMP7 interact with type II receptors and recruit type I receptors, whereas BMP2 and BMP4 preferentially bind type I receptors and recruit type II receptors.14 The existence of preformed oligomeric complexes adds an additional layer of intricacy; indeed, binding to preformed receptor complexes versus BMP-induced receptor recruitment can activate different pathways.15

Upon formation of a heterotetrameric complex, the constitutively active type II receptor transphosphorylates the type I receptor at a glycine–serine rich motif known as the GS domain. This activates the type I receptor and allows phosphorylation of the immediately downstream substrate proteins known as the receptor-regulated Smads (R-Smads) at a C-terminal SSXS motif.13 The R-Smads involved in BMP signaling are Smad1, Smad5, and Smad8 (Smad1/5/8). R-Smads then associate with the co-mediator Smad (co-Smad) Smad4, and this complex translocates to the nucleus where it functions as a transcription factor with coactivators and corepressors to regulate gene expression. Inhibitory Smads (I-Smads), Smad6 and Smad7 (Smad6/7), are involved in feedback inhibition of the signaling pathway.16

Several non-canonical, Smad-independent signaling pathways for BMPs have been identified. BMP4, for example, was found to activate TAK-1, a serine–threonine kinase of the MAPKKK family.17, 18 In addition to the MAPK pathway, BMP signaling has been found to affect PI3K/Akt, P/kc, Rho-GTPases, and others.19 Interestingly, BMPs can have temporal regulation of signaling via the canonical Smad pathway or non-canonical pathways.20 The specific pathway that is activated upon ligand-receptor interaction is thus likely dependent upon the extracellular environment, other cellular activity, and crosstalk with other pathways, such as Wnt signaling.

BMP signaling is extensively regulated by extracellular, intracellular, and membrane modulators.21 Extracellular modulators act as agonists or antagonists of BMP signaling. For example, BMP antagonists include the CAN (Cerberus and DAN) family of proteins, Twisted gastrulation, Chordin and Crossveinless 2, and Noggin.22 Intracellular regulators of BMP signaling include microRNAs, I-SMADS, phosphatases such as PP1 and PP2A that dephosphorylate the receptor and R-Smad, and FK506-binding protein 1A (FKBP1A or FKBP12) that binds the GS domain of type I receptors to inhibit receptor internalization.23 Crosstalk with other signaling pathways, such as Wnt signaling, likely adds another layer of control. Co-receptors in the plasma membrane that interact with type I and type II receptors further add a level of regulation. For instance, Endoglin is a co-receptor that has been shown to be important in vascular growth and disease.24

Biological consequences of BMP signaling

More than 15 known BMPs are structurally related and can be further categorized into subgroups based on amino acid or nucleotide similarity. In particular, BMP2/4, BMP5/6/7/8, BMP9/BMP10, and BMP12/13/14 (GDF5/6/7) are subgroups based on phylogenetic analysis (Fig. 1A).25 Analysis with amino acid (Fig. 1A, panel a) or nucleotide (Fig. 1A, panel b) sequences of BMPs yields similar clustering patterns. BMP1, while able to induce bone and cartilage development, is a metalloprotease that functions in collagen maturation as a procollagen C-proteinase and is not part of the TGF-β superfamily.26 Although the name implies that all members are inducers of bone, some BMPs can act as inhibitors of bone formation. For example, BMP3 is a negative regulator of bone density, and BMP13 is a strong inhibitor of bone formation.27, 28 BMP2, 4, 6, 7, and 9 are commonly referred to as the osteogenic BMPs, based on their potent bone-inducing activity.29 For instance, BMP2 is indispensable for endochondral bone formation.30

Many organ systems have one or more BMPs that are critical for development. BMP4 serves to regulate limb development, and BMP13 has a modulatory role in the development of the eye.31, 32 BMP7, important for eye development, is also crucial in the kidney. BMP8 has a demonstrated role in spermatogenesis, BMP12 is needed for seminal vesicle development, and BMP15 is classically associated with ovarian function.33, 34, 35

BMP signaling in embryogenesis

BMPs are essential during embryogenesis, most prominently for mesoderm formation and cardiac development. Knocking out BMP2 or BMP4 results in embryonic lethality, and BMP1, BMP7, and BMP11 knockouts die shortly after birth (Table 1). BMP2 deficient mice have malformation of amnion and chorion and cardiac defects. BMP4 deficient mice do not have differentiation of mesoderm, implying that BMP4 is essential for developmental processes as early as gastrulation. These mice also have no primordial germ cells (PGCs).41 BMP4 heterozygotes are viable but have a wide variety of abnormalities.33 Mice lacking BMP1 fail to close the ventral body wall and have gut herniation as a result.36

Table 1
Knockout phenotypes and biological consequences for the major players in BMP signaling.
Smad factorsBMP receptors
Signaling moleculePhenotype
BMP1Die after birth, failure of ventral body wall closure36
BMP2Embryonically lethal, defects in amnion/chorion and cardiac development37; limb: spontaneous fractures and impaired fracture repair8; chondrocyte: severe chondrodysplasia30; cardiac progenitor: abnormal heart valve development38; myocardium: defects in myocardial patterning39
BMP3Increased bone density28
BMP4Embryonically lethal, lack of mesoderm formation,40 no PGCs,41, 42 no lens induction43; heterozygotes: various organ abnormalities44; hypomorph: AVCD,45 HSC microenvironment defect46; limb bud mesoderm: defective digit patterning31; adipocyte: enlarged adipocytes and impaired insulin sensitivity47; other targeted: loss-of-trachea phenotype,48 abnormal branchial arch arteries and outflow tract septation,49 defects in mandibular development,50 defects in vestibular apparatus51
BMP5Short ear phenotype52; smaller and weaker bones53
BMP6Delay in sternum ossification54; smaller long bones55; decreased fertility56
BMP7Die after birth, defects in kidney and eye development57; defects in skeletal patterning58; impaired corticogenesis59; decreased brown fat,60 diminished Langerhans cell number61; inducible deletion: precocious differentiation of kidney progenitor cells62; limb: no effect63; podocyte: defective kidney development64
BMP8Germ cell degeneration33; defective PGC formation65; germ cell deficiency and infertility66
BMP9/GDF2Abnormal lymphatic development67, 68
BMP10Reduced cardiomyocyte proliferation69
BMP11/GDF11Die after birth, defects in A-P patterning70; smaller pancreas71; reduced β-cell numbers72; kidney agenesis73; slower spinal cord neuron differentiation74; increased olfactory neurogenesis75; retinal abnormalities76
BMP12/GDF7Increased endochondral bone growth77; smaller bone cross-sectional parameters78; no effect on tail tendon phenotype79; subtle effects on Achilles tendon80; defective dorsal interneuron formation81; sterile with seminal vesicle defects34
BMP13/GDF6Bone fusions in wrists and ankles82; accelerated coronal suture fusion83; eye and neural defects84, 85; Klippel–Feil syndrome86; males: lower tail tendon collagen87, 88
BMP14/GDF5Brachypodism89; malformations in bones of limb, sternum, and digits90; delayed fracture healing91, 92; impaired joint formation and osteoarthritis93; weaker Achilles tendon94; increased scarring after myocardial infarction95; altered skin properties96
BMP15Males: normal and fertile; females: subfertile with decreased fertilization and ovulation rates97
Smad1Die mid-gestation, defects in allantois formation,98 no PGCs99, 100; chondrocyte: delayed calvarial bone development101; osteoblast: osteopenia101; lung epithelium: severe neonatal respiratory failure102
Smad5Die mid-gestation, multiple embryonic and extraembryonic defects103; defective PGC formation104; left-right asymmetry105
Smad8Dispensable for development106; defective pulmonary vascular remodeling9
Smad4Embryonically lethal, gastrulation defects107; heterozygotes: gastric and duodenal polyps108, 109; adult: anemia110; osteoblast: lower bone mass111; chondrocyte: hearing loss and inner ear malformation,112, 113 disorganized growth plate114; muscle: muscle atrophy115; cardiomyocyte: cardiac hypertrophy116; endothelial cell: embryonically lethal117, 118; vascular smooth muscle: embryonically lethal119; CNS: cerebellar defects120; eye: defects in lacrimal gland121; Sertoli and Leydig cells: testicular dysgenesis122; preovulatory follices: follicle atresia123; ovary: defects in folliculogenesis124; skin: aberrant wound healing,125 hair loss and squamous cell carcinoma126; keratinocyte: accelerated reepithelialization127; head and neck: head and neck squamous cell carcinoma128; odontoblast: keratocystic odontogenic tumors129
Smad6Defects in axial and appendicular skeletal development130; multiple cardiovascular abnormalities131
Smad7Embryonically lethal, cardiovascular defects132; defects in axial and appendicular skeletal development133; growth retardation and reduced viability134; renal fibrosis and inflammation135; defective eye development136; scleroderma137; altered B cell response138; T cell: reduced disease and CNS inflammation139
BMPR-1A (ALK3)Embryonically lethal, no mesoderm formation140; mesoderm: embryonically lethal,141 omphalocele-like defect142; osteoblast: increased bone mass143; chondrocytes: no long bone growth144; liver: iron overload145; cardiomyocyte: heart valve defects and EC defects146, 147; endocardium: EC defects148; vascular smooth muscle cells: embryonically lethal149; adult vascular smooth muscle cells: impaired vascular remodeling150; lung epithelium: defects in lung development,151 neonatal respiratory distress152; ureteric bud: dysplastic renal phenotype153; hypothalamus (neurons in feeding center): hypophagia and death154; eye: lack of lens and retina growth155; Leydig cell: abnormal Leydig cells156; granulosa cell: subfertile157; facial primordia: lip and palate defects158; hair follicle: impaired differentiation of inner root sheath159; dental epithelia: switched differentiation of crown epithelia to root lineage160
BMPR-1B (ALK6)Defects in appendicular skeleton161; retinal defects162; females: irregular estrous cycles163
ActR-1A (ALK2, ACVR1)Embryonically lethal, defects in mesoderm formation and gastrulation,164 no PGCs42; surface ectoderm: smaller lens165; neural crest: craniofacial defects,166 cardiac outflow tract defects167; liver: iron overload145; endothelium: defects in AV septa, valves, and EC formation168; cushion mesenchyme: bicuspid aortic valve169; uterus: delayed implantation and sterility,170 mid-gestation abnormalities171
BMPR-2Embryonically lethal, defects in mesoderm and gastrulation,172 cardiac defects173; heterozygotes: PAH174, 175; lung epithelium: predisposed to PAH176
ActR-2A (ACVR2A)Defective reproductive performance and sexual behavior177, 178, 179
ActR-2B (ACVR2B)Die after birth, complicated cardiac defects and left-right asymmetry180

Deleting BMPR-1A, ActR-1A, and BMPR-2 results in embryonic lethality, and ActR-2B knockouts die shortly after birth (Table 1). The defects seen in these mice are consistent with the above phenotypes due to lack of BMP signaling. For instance, BMPR-1A is necessary for gastrulation and mesoderm formation.140 ActR-1A is also necessary for gastrulation in the mouse embryo, with mice deficient in this receptor showing disruption of mesoderm formation and no PGCs.42, 164 These phenotypes are again recapitulated in mouse knockouts of intracellular regulators. Smad1, Smad5, Smad4, and Smad7-knockouts are all embryonically lethal, but interestingly Smad8 is dispensable for development (Table 1).106 Smad1 plays an essential role in fusion of amnion and chorion, and mutants have defects in extraembryonic structures and germ cell formation.99, 140 Smad5 knockouts exhibit multiple embryonic and extraembryonic defects as well, including left-right asymmetry and significantly reduced PGC number.103, 104, 105

BMP signaling in skeletal system

BMPs play a crucial role in bone and cartilage formation, providing the namesake for this family of proteins, as well as in adult homeostasis of bone function. Though BMPs were initially discovered to induce bone formation, BMP3 has been shown to be a negative regulator of bone density.28 Deletion of BMPR-1A in osteoblasts also surprisingly leads to increased bone mass.143 Some BMPs may have redundant roles in bone formation, as conditional deletion of BMP7 from limb has no noticeable effect.63 However, BMP7 deficient mice have skeletal patterning defects confined to the ribs, skull, and hindlimbs.58 Additionally, BMP11 plays important roles in skeletal patterning during development, as BMP11 mutants have altered Hox gene expression and abnormal anterior-posterior (A-P) patterning of the axial skeleton.70

The short ear and brachypodism mutations, associated with BMP5 and BMP14, respectively, are specific named loci and phenotypes in the mouse.52, 89 These mice are both viable and fertile, but have various skeletal defects. BMP5 mutants have shorter and weaker bones, while the brachypodism mutation results in mice having altered length and number of bones in the limb.53 Examination of BMP14 mutants revealed that BMP14 also coordinates bone and joint formation during digit development.90 BMP5/14 double mutants have additional defects compared to single mutants, suggesting a likely synergistic function.181 Along with BMP14, BMP4 regulates digit patterning and the apical ectodermal ridge.31 A BMP14 mutant with severe joint malformations exhibited early-onset osteoarthritis (OA), offering a potential model for the study of the disease.93 Interestingly, BMP14 is implicated in OA in humans, although the mechanism is not clear (see OA section). Mice lacking BMP6 are also viable and fertile, but show reduced size of long bones and delayed sternal ossification, which is slightly exacerbated in BMP5/BMP6 double mutants.54, 55

The GDF5/6/7 family of BMPs (BMP12/13/14) are important in normal formation of bones and joints, and there is increasing evidence of their role in tendon and ligament biology. BMP12 is thought to play a role in the structural integrity of bone.78 Mutation in BMP13 causes defects at multiple sites, including the wrist and ankle.82 These sites are distinct from BMP14 mutants, and BMP13/14 double mutants show additional defects. BMP13 knockout mice have accelerated coronal suture fusion, indicating an inhibitory role of BMP13 in osteogenic differentiation.83 Furthermore, mutations in BMP13 are distinguished by fusion of carpal and tarsal bones.86 An in vitro study confirmed the strong inhibitory role of this BMP.27 Examination of tendon phenotype in mice with mutations in this subgroup has revealed the role of BMP in tendon biology. BMP14 deficiency results in structurally weaker tendon and altered mechanics, BMP13 deficiency results in significantly lower tendon collagen in males, and BMP12 deficiency has only a subtle effect on tendon phenotype although BMP14 levels were higher in these mice.79, 87, 94 This suggests these proteins might have overlapping roles, and BMP14 may be able to compensate for BMP12 deficiency.

BMPs also regulate cartilage development, which is usually coordinated with bone formation. BMP2 is considered a main player during endochondral bone development for chondrocyte proliferation and maturation.30 Chondrocyte-specific knockout of BMP2 results in a severe chondrodysplasia phenotype. BMP12, on the other hand, may serve as a negative regulator of chondrogenesis, since BMP12 deficient mice have accelerated hypertrophic chondrocyte kinetics.77 The Smad pathway is essential to mediate signaling in chondrocytes. Chondrocyte-specific deletion of Smad1 leads to delayed calvarial bone development.101 I-Smads are significant during cartilage development as well. Smad6 is needed for inhibition of endochondral bone formation, with knockouts showing abnormal growth plates; loss of Smad7 also results in abnormalities at the growth plate.130, 133

BMPs play a prominent role in adult bone homeostasis and fracture healing. BMP2, while dispensable for bone formation, is required for the initiation of fracture healing.8 In limb-specific BMP2 knockouts, the bones lacking BMP2 have spontaneous fractures and an inability to initiate the early stages of fracture healing. BMP14 deficiency is associated with a delay in the fracture healing process, presumably due to a delay in the recruitment of cells and chondrocyte differentiation, but long-term healing is not compromised.91, 92 These results emphasize the importance of BMPs as inducers of proliferation and differentiation in post-natal life.

The ability of BMPs to induce bone and cartilage formation is the basis for understanding the mechanism of certain diseases, as well as the potential use of recombinant human BMPs in treatments. Cleft palate (CP) is a recognizable birth defect that has various etiologies. BMPs are known to have a role in palate morphogenesis in development, and haploinsufficiency of BMP2 has been associated with syndromic CP.182 A family with a BMP2 deletion was examined, with the conclusion that BMP2 haploinsufficiency has high penetrance but variable expressivity. BMP4 also plays an important role in maxillofacial development, as three variants in BMP4 were identified as potential risk factors for nonsyndromic cleft lip/palate.183 We further discuss three other bone diseases linked to BMP signaling.

Fibrodysplasia ossificans progressiva (FOP)

FOP is a congenital disease that causes heterotopic ossification of soft tissues, such as skeletal muscle, through the endochondral pathway.184 The classic phenotype is caused by a constitutively activating mutation in ACVR1, the gene for ActR-1A (ALK2), due to an R206H mutation in the GS domain, and accounts for at least 98% of classic presentations.185 Other gain-of-function mutations have also been identified.186, 187

The constitutive activity appears to be dependent on non-enzymatic cooperation with type II receptors, but not type II receptor kinase activity or ligand participation.188, 189 Determination of the crystal structure of the cytoplasmic domain of ActR-1A complexed with FKBP12, an intracellular negative regulator of BMP signaling, revealed that FOP mutations disrupt critical interactions and decrease FKBP12 binding, which is consistent with other findings.190 Impaired binding of FKBP12 likely contributes to leaky activity of the type I BMP receptor and increased BMP pathway activity.191 The subcellular distribution of ActR-1A and FKBP12 may also play an important role.192 In addition to the canonical pathway, the BMP-p38 MAPK signaling pathway is disrupted in FOP.193 Because there is no known effective treatment of FOP, the focus is on prevention of heterotopic ossification. A selective small-molecule inhibitor of the type I BMP receptor, LDN-193189, inhibits activation of Smad1/5/8 and results in reduction of ectopic ossification.194 Recently, a novel class of small-molecule inhibitor of BMP signaling was discovered.195 Anti-sense oligonucleotides targeting the overactive ActR-1A receptor are another avenue of investigation.196

Osteogenesis imperfecta (OI)

OI, or “brittle bone disease,” is a heritable disorder characterized by bone fragility, deformity, and growth deficiency, and is etiologically related to type I collagen, one of the main components of the extracellular matrix.197 Type I collagen is synthesized as a procollagen I precursor with N- and C-terminal propeptides that must be cleaved for maturation and proper fibril assembly. Classic OI has autosomal dominant inheritance and is due to mutations in type I collagen genes, but rare forms have been discovered with autosomal recessive inheritance and are indirectly related to type I collagen. Two children of a consanguineous Egyptian family were diagnosed with severe autosomal recessive OI, and found to have a F249L homozygous missense mutation in the protease domain of BMP1.198 BMP1 is a metalloproteinase known to have procollagen C-proteinase activity that cleaves the C-propeptides from procollagens I-III.199 Another study of two individuals of a consanguineous Turkish family with autosomal recessive OI found a homozygous G12R mutation in the signal peptide of BMP1.200

Osteoarthritis (OA)

OA is a disease involving degeneration of the articular cartilage in synovial joints, such as the knee, hip, and hand. The molecular mechanisms of pathogenesis are not fully understood, but there appears to be a genetic component.201 An association between two polymorphisms in intron I of the BMP5 gene and OA has been demonstrated, and suggests that variability in gene expression of BMP5 is a susceptibility factor for the disease.202 The 5’ UTR of BMP14 is also implicated as a susceptibility factor.203 Overexpression of BMP2 was found in OA tissues and may be involved in the response to cartilage degeneration.204 However, crosstalk between the BMP pathway and Wnt/β-catenin pathway revealed that BMP2 contributes to both chondrocyte hypertrophy and cartilage degradation.205 This dual role of BMPs in OA has been discussed and explains why both increased cartilage anabolism and catabolism are observed.204 Elevated serum BMP2 and BMP4 is evidence of disease and has been proposed as indicators for disease severity and joint arthroplasty.206 Only a couple of studies have investigated OA progression, and these found no association with BMP2 or BMP5.206, 207

BMP7 has been known to protect cartilage and inhibit degradation in models of OA.208, 209 Investigation into the potential applicability of this for treatment in humans is in progress. A Phase I trial to evaluate safety in using BMP7 for the treatment of OA found no major adverse events and also a dose-dependent trend in symptom improvement.210 These results have suggested potential therapeutic applications of BMP7 for OA. Investigations of BMP2 as an injectable therapeutic agent to stimulate cartilage repair have also shown promise in an animal model.211

BMP signaling in muscle

Not many pathways are known to regulate muscle growth in adulthood other than myostatin, which is a member of the TGF-β family and a known negative regulator.115 It binds to activin type II receptors (ActR-2A and ActR-2B) and activin type I receptors (ALK4 and ALK5), leading to Smad2/3 phosphorylation and subsequent complexing with Smad4 to affect gene expression. Recently, it has been shown that the BMP pathway is an important hypertrophic, as well as anti-atrophic, signal in adult muscle.115 Constitutively active type I BMP receptor can lead to substantial muscle hypertrophy. Decreased levels of phosphorylated Smad2/3 perhaps leads to release of Smad4, which is then recruited into BMP signaling. Another study also concluded BMP signaling was a positive regulator of skeletal muscle.212 These results demonstrate the emerging evidence of BMP signaling in adult muscle homeostasis.

Mutations in Smad4 in humans result in Myhre syndrome, which is associated with muscle hypertrophy.213 The Smad4 mutations lead to increased stability of the protein and result in downregulation of the Myostatin pathway and variable response of the BMP pathway. Further investigations need to be done, but it seems the muscle hypertrophy is mainly due to inhibition of Myostatin signaling. Muscle-specific knockout of Smad4 in mice leads to muscle atrophy, consistent with the above finding.115

BMP signaling in gastrointestinal system

BMP signaling is required for normal growth and morphogenesis of the developing gastrointestinal tract.214 The underlying smooth muscle of the embryonic gut is also dependent upon BMP signaling, especially BMP2, for proper development.215 Gastric patterning requires BMP signaling, as deletion of BMPR-1A in foregut endoderm leads to anteriorization of the stomach.216 BMP11 is important in pancreatic development, and BMP11 deficient mice have a pancreas half the size of wild-type mice.71

Barrett esophagus (BE)

BE is a metaplasia associated with esophagitis from gastroesophageal reflux disease, in which the squamous epithelium of the esophagus is replaced by columnar epithelium. BMP4 and the downstream phosphorylated Smad1/5/8 were found to be upregulated in BE patient samples.217, 218 The BMP pathway is thus implicated in the trans-differentiation of squamous epithelium to columnar epithelium in inflamed esophageal mucosa, but the pathogenesis is not well understood. Initial upregulation of the Hedgehog pathway may lead to upregulation of target genes, including BMP4, subsequently reprogramming the squamous epithelium to the more favorable columnar type for protection.219, 220 microRNA modulation of BMP4 is also implicated in the development of BE.221

Juvenile polyposis (JP) and colorectal cancer

It has been relatively well-established that deletion of Smad4 in mice leads to gastric and duodenal polyps. While homozygotes are embryonically lethal, heterozygotes develop polyps with loss of heterozygosity.108, 109 This is highlighted in the corresponding human disease called JP. JP is an autosomal dominant syndrome in which affected individuals develop cystic polyps in the stomach and intestines with an increased risk for colorectal cancer. Mutations in Smad4 and BMPR-1A have been linked to the development of JP, and together they are responsible for around 40% of JP cases.222, 223, 224, 225 The genetic mutations represent a downregulation of the BMP signaling pathway, indicating a significant event in the pathogenesis of JP. Both nonsense and missense mutations of Smad4 have been identified in patients and result in reduced BMP signaling, with nonsense mutations causing a more significant reduction.

Missense mutations in BMPR-1A do not lead to decreased expression of the receptor, but rather cause localization to the cytoplasm instead of the plasma membrane.226 Deletions in chromosome 10q23, encompassing the PTEN and BMPR-1A genes, cause aggressive polyposis and numerous congenital anomalies, such as facial dysmorphism.227 It was recently shown that the BMP-Smad1 pathway functions as a tumor suppressor and stabilizes the well-known p53 tumor suppressor.228 Disruption of this interaction is thus suspected to play a role in tumorigenesis and the development of JP and cancer. The discovery of the relationship between BMPR-1A mutations and JP led to speculation of the role of BMP in colorectal cancer. Indeed, the BMP pathway is inactivated in the majority of sporadic colorectal cancer cases.229 BMP signaling has now been suggested to be involved in the initiation and progression of gastrointestinal cancer.

Studies have identified numerous potential markers for colorectal cancer, but further investigation is needed to clarify the associated mechanism. BMP11 may be a diagnostic and prognostic marker in colorectal cancer patients, as tumors with high BMP11 expression have a higher frequency of lymph node metastases, more cancer-related deaths, and decreased overall survival.230 Another study investigated potential markers for colorectal neoplasia screening, identifying BMP3, as well as three other genes, as highly methylated and silenced compared with normal epithelia.231 Interestingly, BMP3 was also found to be a powerful methylation marker in a stool assay for the detection of pancreatic cancer.232 In both these studies, however, many other markers were also identified, so it remains to be seen which have real significance. The relevance of silencing BMP3 has been investigated in the onset of colorectal cancer development, but the lack of known BMP3-interacting proteins is a hindrance to understanding. BMP3 inactivation does appear to be important in early polyp formation and colorectal tumor development.233 One study using data from population-based case-control studies found significant variation in certain BMP genes in colon and rectal cancer.234 Genetic variations in BMPR-1A, BMPR-1B, BMPR-2, BMP2, BMP4 were all associated with risk of developing colon cancer, with the most high-risk phenotypes conferring a 20–30% increased risk. BMPR-2, BMPR-1B, and BMP2 were associated with rectal cancer.

BMP signaling in cardiovascular and pulmonary systems

BMP signaling has an established role in development of the heart, the first functional organ in the embryo, and is a continuing area of investigation. BMP2 homozygous mutants are embryonically lethal, with malformation of the amnion and chorion and also developmental abnormalities of the heart.37 BMP2 expression is detected in extraembryonic mesoderm as well as myocardium, and the signaling from myocardium has been demonstrated by multiple studies to be critical for endocardial cushion (EC) formation. Signaling from myocardium to the underlying endothelium to form ECs depends on an epithelial-mesenchymal transformation (EMT) mediated by BMP2.38 The ECs eventually give rise to the mature heart valves and septa, ultimately allowing for formation of a four-chambered heart. Conditional deletion of BMP2 in cardiac progenitors prevents this process, and the heart valve region becomes differentiated chamber myocardium. Deletion of BMP2 in atrioventricular (AV) myocardium further revealed the role of BMP2 in EC EMT, as well as in formation of cardiac jelly and patterning of AV myocardium.39

Together with BMP2, BMP4 plays an essential role in the AV septation of the heart. BMP4 signaling from the myocardium to endocardium is involved in the process, and conditional inactivation leads to AV canal defect (AVCD).45 BMP4 is also required for outflow tract septation as demonstrated by conditional knockout.49 Several other BMPs play a role in the developing heart. BMP6 and BMP7, for example, are expressed in the ECs, although neither is essential during cardiogenesis. However, double mutants have defective EC development.235 BMP10 plays an essential role in maintaining cardiac growth during cardiogenesis, as BMP10 knockout mice have dramatically reduced cardiomyocyte proliferation.69 Smad6 and Smad7 mutants both show developmental defects in the outflow tract.131, 132

Murine knockout models of BMP receptors and BMP modulators in cardiac development have also been explored. Since multiple BMP knockouts show developmental abnormalities of the heart, it is not surprising that lack of BMP receptors presents with similar phenotype. Mouse knockouts have shown that BMPR-2 regulates outflow tract and AV cushion development.173 Mutation of BMPR-1A is embryonically lethal, even if only in cardiomyocytes and vascular smooth muscle.149 BMPR-1A signaling in the AV myocardium is required for EC formation and development of AV valves from the ECs.146, 147 ActR-1A is also crucial for AV cushion development, specifically the EMT required in EC formation.168 Ablation of ActR-1A signaling in neural crest cells results in impaired migration of these cells to form the outflow tract.167 Deletion of ActR-1A in cushion mesenchyme results in bicuspid aortic valve.169

BMP4 is a prominent signaling molecule in lung development. Knocking out Smad1 in lung epithelium disrupts branching morphogenesis and ultimately results in severe neonatal respiratory failure.102 It is thought that BMP4 and Smad1 signaling crosstalks with Wnt signaling to regulate lung development. Knocking out BMPR-1A, expressed mainly in airway epithelial cells, also disrupts branching morphogenesis and airway formation and causes neonatal respiratory distress.151, 152 Smad8 mutants have defective pulmonary vascular remodeling, with a resulting pulmonary arterial hypertension (PAH) phenotype.9 This highlights the role of BMP signaling in adult tissue homeostasis, and the PAH phenotype due to Smad8 mutation is observed in humans as well. BMPR-2 deletion has also been established to give rise to PAH, and the mechanism is likely through a Smad-dependent manner.174, 175, 176

Pulmonary arterial hypertension (PAH)

PAH is characterized by high pulmonary artery pressure and resulting heart failure. There are two types: idiopathic pulmonary arterial hypertension (IPAH) and hereditary pulmonary arterial hypertension (HPAH), with heterozygous germline mutations in BMPR-2 found in more than 70% of patients with HPAH and 20% of patients with IPAH.236 The pathogenesis of disease is not well understood, and a variety of factors other than BMPR-2 are likely involved. In HPAH, the autosomal dominant disease, only about 20% of carriers get the disease, and the low penetrance might be explained by changes in BMPR-2 alternative splicing.237 BMPR-2 mutations influence the disease expression more obviously in males than in females.238

Mutations that lead to decreased BMP signaling have been found in the ligand-binding domain, kinase domain, and long cytoplasmic tail. Although mutant receptors may be expressed at lower level or not at all, it is also possible they have altered cellular localization. A few mutants were shown to associate abnormally with caveolae and clathrin-coated pits, and disruption of these domains restored BMP signaling to wild-type levels.239 BMPR-2 also interacts with the cytoskeleton, and mutant receptors may cause cytoskeletal defects related to the development of PAH.240 Several differences compared to wild-type are noted when BMPR-2 expression is disrupted in pulmonary arterial smooth muscle cells (PASMCs), including reduced BMP2 and BMP4 signaling but enhanced BMP6 and BMP7 signaling.241 This results in loss of the anti-proliferative effects of BMP4 and also activation of the p38-MAPK pathway, leading to aberrant PASMC proliferation and lack of apoptosis.242 In addition, there is increased TGF-β1 signaling and reduced BMP signaling, so the pathogenesis of disease is expected to be in part due to abnormal crosstalk between the two pathways.243 Mutations in Smad8 have been linked to PAH, possibly due to a non-redundant role of Smad8 in microRNA processing.244 Interestingly, PAH is sometimes associated with hereditary hemorrhagic telangiectasia (HHT). Defects in the ALK1 or Endoglin gene can lead to HHT, with associated PAH.24, 245, 246

Sildenafil is an established treatment for PAH, and was shown to enhance BMP4-induced phosphorylation of Smad1/5 and the resulting downstream BMP pathway.247 By restoring some of the BMP pathway function in BMPR-2 deficiency, the anti-proliferative effects of BMP4 were partly re-established. Especially important are the downstream Id proteins that regulate PASMC proliferation by inhibiting the cell cycle.248 A recent study showed that ataluren might be an effective treatment for HPAH caused by nonsense mutations by causing ribosomal read-through of the premature stop codon.249 This is promising in an age of personalized medicine in which treatments are tailored based on one's genetics.

Hereditary hemorrhagic telangiectasia (HHT)

HHT is an autosomal dominant disease associated with abnormal and fragile blood vessel formation in skin and mucosa. HHT type 1 is due to mutations in Endoglin, a BMP co-receptor, while HHT type 2 is due to loss-of-function in ALK1.236 Smad4 mutations are known to be associated with a combined JP-HHT syndrome that predisposes to thoracic aortic disease.250 The discovery of BMP9 as the ligand for ALK1 showed that mutations in the receptor lead to defective BMP9 signaling.251 ALK1 is mainly expressed in arterial endothelial cells, and the defective BMP9 signaling likely leads to decreased levels of Id1 and Id3. Subsequent increased expression of VEGR2 is suggested as the mechanism of abnormal endothelial cell sprouting. Other non-Smad pathways, such as the MAPK cascade, likely also play an important role in the process.252 This process reveals the role of BMP9 as a regulator of angiogenesis in the adult. Because ALK1 normally has anti-angiogenic effects and is mainly expressed in endothelial cells, it makes for a potentially interesting target in diseases related to angiogenesis, such as cancer.253 BMP9 might be considered in such treatments, since it is one of the main ligands of the receptor.

BMP signaling in urinary system

Initiation of mature kidney development involves the ureteric bud, which originates from the caudal end of the mesonephric duct, invaginating into metanephric mesenchyme. The metanephric mesenchyme ultimately gives rise to structures from the glomerulus to distal convoluted tubule, while the ureteric bud is the precursor to all structures distal to the collecting duct. BMP signaling is one mediator of the interaction between ureteric bud and metanephric mesenchyme. Abnormal interaction due to improper BMP signaling causes malformations of the kidney, in particular a group of disorders termed congenital anomalies of the kidney and urinary tract (CAKUT), which includes renal agenesis, dysplasia, ureteropelvic junction obstruction (UPJO), and others.254 BMP11 knockout mice, for example, have a spectrum of renal abnormalities, with the majority having bilateral renal agenesis.73 BMP11 likely plays a role in directing the ureteric bud from the mesonephric duct towards the metanephric mesenchyme, as mutant BMP11 embryos show a failure of ureteric bud formation. Control of ureteric bud branching is mediated by BMPR-1A signaling, as conditional knockout of the receptor in ureteric bud results in abnormal bud branching.153 These mice have renal dysplasia phenotype and decreased number of collecting ducts.

Renal hypodysplasia (RHD) is characterized by reduced kidney size or maldevelopment of the renal tissue. BMP4 mutations were identified in RHD patients, consistent with BMP4 being mainly expressed in the mesenchyme surrounding the branching ureter.255 Homozygous mutations in BMP4 and DACH1 were found in a patient with Bilateral Cystic Renal Dysplasia.256 Proper signaling of BMP4 and BMP7 is necessary for complete development of the urethra during embryonic development. BMP4 is a major factor in the signaling cascade involved in controlling embryonic urethral development, and BMP7 helps regulate the growth of the urethral plate epithelium as well as the proper closure of the distal urethra.257 This is consistent with missense mutations in BMP4 and BMP7 being strongly associated with hypospadias.258 Mutations in BMP4 can also lead to UPJO.259 The ureteropelvic junction is the last to canalize and the most common site of obstruction in the fetus. It is believed that these mutations cause UPJO due to loss of canonical Smad4 signaling mediated in part by decreased levels of BMP4, although the mechanism is at least partially Smad4-independent.260

BMP7 has emerged as one of the most critical BMPs for kidney development. Mice lacking BMP7 have severe kidney defects in addition to eye defects. Early nephrogenic tissue interactions establishing the organ do not appear to be affected, but renal dysplasia ultimately results from lack of continued growth and development.57 This indicates a role for BMP7 in maintaining proliferation. After establishing a small number of nephrons, further development is arrested, and there is massive apoptosis of kidney progenitor cells. Using an inducible BMP7 knockout system, it was shown that BMP7 preserves undifferentiated kidney progenitor cells by preventing their differentiation into nephron.62 In effect, this serves as a regulator of kidney nephron number. Podocyte-specific knockout of BMP7 revealed a BMP7-mediated regulatory axis between glomeruli and proximal tubules during kidney development.64

Recent research has shown that the presence of BMP signaling may be protective against renal disease, evidence of BMP function in adult homeostasis. Progression of chronic kidney disease (CKD) is mainly determined by renal fibrosis, which is characterized by an accumulation of extracellular components that leads to loss of renal parenchyma and function. BMP7, highly expressed in podocytes, distal tubules, and collecting ducts, has been shown to be protective against CKD.261 Exogenous administration of BMP7 or transgenic overexpression of BMP7 reduces overall renal fibrosis and nephrocyte apoptosis.262 Additionally, BMP7 signaling has been shown to be protective against hypertensive nephrosclerosis, another major cause of CKD.263 BMP5 may have a similar role.264

BMP signaling in neurological and ophthalmic systems

BMP signaling plays an important role in many aspects of the development of the eye, including the formation of the retina, lens, iris, and ciliary body. Eye development begins with paired optic vesicles, diverticula of the forebrain that contact the surface ectoderm and develop into optic cups. The surface ectoderm thickens to form the lens placode, which in part gives rise to the lens. There is signaling between the optic vesicle and surface ectoderm mediating these transitions. BMP4 and BMP7 are implicated in the progression from optic vesicle to optic cup.265 Furthermore, BMP4 is an important ligand from the optic cup in lens induction, and BMP7 is expressed in the lens placode to regulate lens induction as well.43 Germline mutations in BMP7 prevent lens formation.58 As in the case of the kidney, early tissue interactions establishing the organ are unaffected in BMP7 mutants, but the mice exhibit anophthalmia.57 BMPR-1A is essential for lens and retinal growth.155 Although lens formation occurred with conditional knockout of ActR-1A in surface ectoderm, the lenses were smaller, due to an increase in apoptosis of lens epithelial cells.165 BMP13 is implicated in maintaining cell survival in the eye and central nervous system, as loss of BMP13 results in retinal apoptosis and smaller eye size.84, 85 Retinal apoptosis is also associated with loss of BMPR-1B, which is exclusively expressed in the ventral retina during development.162

In humans, frameshift, missense, and Kozak sequence mutations have been identified in BMP7 in individuals with developmental eye abnormalities and extra-ocular features.266 BMP7 is suggested to have an important role in optic fissure closure, and thus may play a role in coloboma, a condition in which there is a hole or gap in some part of the eye. The incomplete penetrance and variability in phenotype expression in families examined demonstrates the complex nature of BMP-related diseases. There are not extensive reports of BMP4 loss-of-function mutations and the resulting effects, but it is known to be associated with ocular defects, as well as digit and brain anomalies. BMP4 is also known to be associated with anophthalmia–microphthalmia (A–M), but with large degrees of variable expressivity, sometimes with polysyndactyly.267, 268, 269 One report indicated a role for BMP4 in short stature, hyperextensibility, hernia, ocular depression, Rieger anomaly, and teething delay (SHORT) syndrome.270

Another role of BMP in the neurological system is neurogenesis, and neural defects are associated with loss of BMP function in mouse models. BMP12 signaling in the spinal cord leads to differentiation of specific neuron classes, suggesting a role for BMPs in determining neuron identity in the CNS.81 BMP11 is also involved in spinal cord neurogenesis through its ability to induce cell cycle exit and instead promote differentiation of progenitors.74 On the other hand BMP11, secreted from neurons themselves, serves as an inhibitory signal in the generation of new neurons from progenitors in the olfactory epithelium.75 It plays a role in retina development as well.76 Further highlighting the importance of BMP regulation in neural development is the role of BMP7 in corticogenesis. BMP7 deletion results in reduced cortical thickening and impaired neurogenesis.59 Interestingly, BMPR-1A is important in establishment of neurons involved in regulating feeding behavior.154

BMP signaling in reproductive system

Several BMPs play an important role in aspects of reproductive system development and biology, from PGC formation to seminal vesicle development.34 As mentioned previously, knocking out some BMPs, such as BMP4, results in lack of formation of PGCs. Fertility is affected by several BMPs. BMP15 is the prominent BMP associated with ovarian function, specifically granulosa cell proliferation. Male BMP15 knockout mice are normal and fertile, while females are subfertile and have decreased ovulation and fertilization rates.97 BMP6 knockout females are demonstrated to have decreased fertility as well, with a decrease in ovulated eggs.56 BMP8 is most often associated with male reproductive system development, including spermatogenesis and development of epididymis. Maintenance of germ cells and spermatogenesis, and formation of PGCs, are dependent on BMP8.33, 65, 66

A couple of BMP receptors have been shown to be important in pregnancy. BMPR-2 is required for maintenance of pregnancy and uterine function after implantation.171 Signaling in the uterus during implantation requires ActR-1A.170

BMP signaling in adipogenesis

Adipocyte development first involves the generation of pre-adipocytes from mesenchymal stem cells, followed by differentiation of pre-adipocytes into adipocytes. Mesenchymal stem cells have the ability to differentiate along several lineages, including osteocytes, chondrocytes, myocytes, fibroblasts, and adipocytes. The specific lineage is in part regulated by BMPs. The osteogenic BMPs, in particular BMP9, are strong inducers of osteocyte differentiation. However, BMP2 and BMP4 have been shown to be capable of inducing pluripotent stem cells into adipocytes, mediated primarily by the Smad pathway rather than a non-canonical pathway.271

Obesity is characterized by an increase in white adipose tissue accumulation, which results from an increase in adipocyte size and/or an increase in adipocyte number. Mice studies have generally led to the belief that BMP4 induces stem cells to differentiate along the white adipocyte lineage, whereas BMP7 induces brown adipocyte differentiation.60 However, recent studies have indicated that BMP4 induces white-to-brown transition.272 Expressing BMP4 in white adipocytes of mice leads to white adipocyte cells with brown adipocyte characteristics, suggesting a role for BMP4 in altering insulin sensitivity.47 BMPR-1A has been associated with obesity because its activity has been shown to favor differentiation into adipocytes as opposed to osteoblasts.273 BMPR-2 has also been implicated in obesity.274 BMP7 treatment of diet-induced obese mice leads to increased energy expenditure and decreased food intake.275 This is likely linked to the ability of BMP7 to induce brown adipogenesis, and presents as a potentially interesting avenue for disease treatment.

Conclusions and future directions

BMP signaling is critical in embryogenesis and is involved in development of many organ systems, as well as many aspects of adult tissue homeostasis. BMPs mediate processes important in development, such as cell proliferation, differentiation, and apoptosis. Deletion of various components of the BMP pathway is embryonically lethal or presents with marked abnormalities. Thus, conditional knockout mouse models have aided tremendously in studying BMP function, and have provided much insight into the roles of BMP signaling in development. Adult tissues also rely on BMP signaling for homeostasis, and examples include fracture repair initiation and pulmonary vascular remodeling. Several players in BMP signaling have been determined to be the causative agent of human disease, while others have been shown to have a strong association. While these associations have been determined, the mechanisms of pathogenesis need to be fully understood. Thus, many critical biological questions pertaining to BMP signaling remain unanswered: What are the upstream signals governing BMP signaling? How are the distinct biological outcomes of a given BMP in different cell and tissue types regulated? Conversely, it remains to be understood how the actions of different BMPs exerted on the same cell or tissue types are coordinated. Furthermore, the extensive crosstalk with other major signaling pathways, such as the Wnt pathway, needs to be fully elucidated. Ultimately, a better understanding of BMP signaling should facilitate the clinical management of the BMP signaling-associated diseases, and may lead to the development of innovative and efficacious therapies, especially in the field of regenerative medicine.

Conflicts of interest

All authors have none to declare.

Footnotes

Peer review under responsibility of Chongqing Medical University.

Acknowledgments

The reported work was in part supported by research grants from theNational Institutes of Health(AR50142 and AR054381 to RCH and HHL). RW, JG, and OI were recipients of the Pritzker Summer Research Fellowship funded through a NIH T-35 training grant (NIDDK). AH was a recipient of the Urban Leadership Fellowship from Miami University.

References

  • 1. UristM.R.Bone: formation by autoinductionScience15036981965893899[PubMed][Google Scholar]
  • 2. WozneyJ.M.RosenV.CelesteA.J.Novel regulators of bone formation: molecular clones and activitiesScience2424885198815281534[PubMed][Google Scholar]
  • 3. Hemmati-BrivanlouA.ThomsenG.H.Ventral mesodermal patterning in Xenopus embryos: expression patterns and activities of BMP-2 and BMP-4Dev Genet17119957889[PubMed][Google Scholar]
  • 4. ZouH.NiswanderL.Requirement for BMP signaling in interdigital apoptosis and scale formationScience27252621996738741[PubMed][Google Scholar]
  • 5. StewartA.GuanH.YangK.BMP-3 promotes mesenchymal stem cell proliferation through the TGF-beta/activin signaling pathwayJ Cell Physiol22332010658666[PubMed][Google Scholar]
  • 6. KobayashiT.LyonsK.M.McMahonA.P.KronenbergH.M.BMP signaling stimulates cellular differentiation at multiple steps during cartilage developmentProc Natl Acad Sci U S A1025020051802318027[PubMed][Google Scholar]
  • 7. BobaczK.GruberR.SoleimanA.ErlacherL.SmolenJ.S.GraningerW.B.Expression of bone morphogenetic protein 6 in healthy and osteoarthritic human articular chondrocytes and stimulation of matrix synthesis in vitroArthritis Rheum489200325012508[PubMed][Google Scholar]
  • 8. TsujiK.BandyopadhyayA.HarfeB.D.BMP2 activity, although dispensable for bone formation, is required for the initiation of fracture healingNat Genet3812200614241429[PubMed][Google Scholar]
  • 9. HuangZ.WangD.Ihida-StansburyK.JonesP.L.MartinJ.F.Defective pulmonary vascular remodeling in Smad8 mutant miceHum Mol Genet1815200927912801[PubMed][Google Scholar]
  • 10. WagnerD.O.SieberC.BhushanR.BörgermannJ.H.GrafD.KnausP.BMPs: from bone to body morphogenetic proteinsSci Signal31072010mr1[Google Scholar]
  • 11. HarrisonC.A.Al-MusawiS.L.WaltonK.L.Prodomains regulate the synthesis, extracellular localisation and activity of TGF-β superfamily ligandsGrowth Factors Chur Switz2952011174186[Google Scholar]
  • 12. HeldinC.H.MiyazonoK.ten DijkeP.TGF-beta signalling from cell membrane to nucleus through SMAD proteinsNature39066591997465471[PubMed][Google Scholar]
  • 13. HorbeltD.DenkisA.KnausP.A portrait of transforming growth factor β superfamily signalling: background mattersInt J Biochem Cell Biol4432012469474[PubMed][Google Scholar]
  • 14. De CaesteckerM.The transforming growth factor-beta superfamily of receptorsCytokine Growth Factor Rev1512004111[PubMed][Google Scholar]
  • 15. NoheA.HasselS.EhrlichM.The mode of bone morphogenetic protein (BMP) receptor oligomerization determines different BMP-2 signaling pathwaysJ Biol Chem2777200253305338[PubMed][Google Scholar]
  • 16. HeldinC.-H.MoustakasA.Role of Smads in TGFβ signalingCell Tissue Res347120122136[PubMed][Google Scholar]
  • 17. DerynckR.ZhangY.E.Smad-dependent and Smad-independent pathways in TGF-beta family signallingNature42569582003577584[PubMed][Google Scholar]
  • 18. YamaguchiK.ShirakabeK.ShibuyaH.Identification of a member of the MAPKKK family as a potential mediator of TGF-beta signal transductionScience2705244199520082011[PubMed][Google Scholar]
  • 19. ZhangY.E.Non-Smad pathways in TGF-beta signalingCell Res1912009128139[PubMed][Google Scholar]
  • 20. BroegeA.PhamL.JensenE.D.Bone morphogenetic proteins signal via SMAD and mitogen-activated protein (MAP) kinase pathways at distinct times during osteoclastogenesisJ Biol Chem2885220133723037240[PubMed][Google Scholar]
  • 21. CorradiniE.BabittJ.L.LinH.Y.The RGM/DRAGON family of BMP co-receptorsCytokine Growth Factor Rev205–62009389398[PubMed][Google Scholar]
  • 22. WalshD.W.GodsonC.BrazilD.P.MartinF.Extracellular BMP-antagonist regulation in development and disease: tied up in knotsTrends Cell Biol2052010244256[PubMed][Google Scholar]
  • 23. YaoD.DoréJ.J.Jr.LeofE.B.FKBP12 is a negative regulator of transforming growth factor-beta receptor internalizationJ Biol Chem2751720001314913154[PubMed][Google Scholar]
  • 24. ToporsianM.JerkicM.ZhouY.-Q.Spontaneous adult-onset pulmonary arterial hypertension attributable to increased endothelial oxidative stress in a murine model of hereditary hemorrhagic telangiectasiaArterioscler Thromb Vasc Biol3032010509517[PubMed][Google Scholar]
  • 25. MuellerT.D.NickelJ.Promiscuity and specificity in BMP receptor activationFEBS Lett58614201218461859[PubMed][Google Scholar]
  • 26. KesslerE.TakaharaK.BiniaminovL.BruselM.GreenspanD.S.Bone morphogenetic protein-1: the type I procollagen C-proteinaseScience27152471996360362[PubMed][Google Scholar]
  • 27. ShenB.BhargavD.WeiA.BMP-13 emerges as a potential inhibitor of bone formationInt J Biol Sci522009192200[PubMed][Google Scholar]
  • 28. DaluiskiA.EngstrandT.BahamondeM.E.Bone morphogenetic protein-3 is a negative regulator of bone densityNat Genet27120018488[PubMed][Google Scholar]
  • 29. LuuH.H.SongW.-X.LuoX.Distinct roles of bone morphogenetic proteins in osteogenic differentiation of mesenchymal stem cellsJ Orthop Res Off Publ Orthop Res Soc2552007665677[Google Scholar]
  • 30. ShuB.ZhangM.XieR.BMP2, but not BMP4, is crucial for chondrocyte proliferation and maturation during endochondral bone developmentJ Cell Sci124Pt 20201134283440[PubMed][Google Scholar]
  • 31. SeleverJ.LiuW.LuM.-F.BehringerR.R.MartinJ.F.Bmp4 in limb bud mesoderm regulates digit pattern by controlling AER developmentDev Biol27622004268279[PubMed][Google Scholar]
  • 32. Asai-CoakwellM.FrenchC.R.BerryK.M.GDF6, a novel locus for a spectrum of ocular developmental anomaliesAm J Hum Genet8022007306315[PubMed][Google Scholar]
  • 33. ZhaoG.Q.LiawL.HoganB.L.Bone morphogenetic protein 8A plays a role in the maintenance of spermatogenesis and the integrity of the epididymisDev Camb Engl1256199811031112[Google Scholar]
  • 34. SettleS.MarkerP.GurleyK.The BMP family member Gdf7 is required for seminal vesicle growth, branching morphogenesis, and cytodifferentiationDev Biol23412001138150[PubMed][Google Scholar]
  • 35. OtsukaF.YaoZ.LeeT.YamamotoS.EricksonG.F.ShimasakiS.Bone morphogenetic protein-15. Identification of target cells and biological functionsJ Biol Chem2755020003952339528[PubMed][Google Scholar]
  • 36. SuzukiN.LaboskyP.A.FurutaY.Failure of ventral body wall closure in mouse embryos lacking a procollagen C-proteinase encoded by Bmp1, a mammalian gene related to Drosophila tolloidDev Camb Engl12211199635873595[Google Scholar]
  • 37. ZhangH.BradleyA.Mice deficient for BMP2 are nonviable and have defects in amnion/chorion and cardiac developmentDev Camb Engl12210199629772986[Google Scholar]
  • 38. Rivera-FelicianoJ.TabinC.J.Bmp2 instructs cardiac progenitors to form the heart-valve-inducing fieldDev Biol29522006580588[PubMed][Google Scholar]
  • 39. MaL.LuM.-F.SchwartzR.J.MartinJ.F.Bmp2 is essential for cardiac cushion epithelial-mesenchymal transition and myocardial patterningDev Camb Engl13224200556015611[Google Scholar]
  • 40. WinnierG.BlessingM.LaboskyP.A.HoganB.L.Bone morphogenetic protein-4 is required for mesoderm formation and patterning in the mouseGenes Dev917199521052116[PubMed][Google Scholar]
  • 41. LawsonK.A.DunnN.R.RoelenB.A.Bmp4 is required for the generation of primordial germ cells in the mouse embryoGenes Dev1341999424436[PubMed][Google Scholar]
  • 42. De Sousa LopesS.M.C.RoelenB.A.J.MonteiroR.M.BMP signaling mediated by ALK2 in the visceral endoderm is necessary for the generation of primordial germ cells in the mouse embryoGenes Dev1815200418381849[PubMed][Google Scholar]
  • 43. FurutaY.HoganB.L.BMP4 is essential for lens induction in the mouse embryoGenes Dev1223199837643775[PubMed][Google Scholar]
  • 44. DunnN.R.WinnierG.E.HargettL.K.SchrickJ.J.FogoA.B.HoganB.L.Haploinsufficient phenotypes in Bmp4 heterozygous null mice and modification by mutations in Gli3 and Alx4Dev Biol18821997235247[PubMed][Google Scholar]
  • 45. JiaoK.KulessaH.TompkinsK.An essential role of Bmp4 in the atrioventricular septation of the mouse heartGenes Dev1719200323622367[PubMed][Google Scholar]
  • 46. GoldmanD.C.BaileyA.S.PfaffleD.L.Al MasriA.ChristianJ.L.Fleming WH. BMP4 regulates the hematopoietic stem cell nicheBlood11420200943934401[PubMed][Google Scholar]
  • 47. QianS.-W.TangY.LiX.BMP4-mediated brown fat-like changes in white adipose tissue alter glucose and energy homeostasisProc Natl Acad Sci U S A11092013E798E807[PubMed][Google Scholar]
  • 48. LiY.GordonJ.ManleyN.R.LitingtungY.ChiangC.Bmp4 is required for tracheal formation: a novel mouse model for tracheal agenesisDev Biol32212008145155[PubMed][Google Scholar]
  • 49. LiuW.SeleverJ.WangD.Bmp4 signaling is required for outflow-tract septation and branchial-arch artery remodelingProc Natl Acad Sci U S A10113200444894494[PubMed][Google Scholar]
  • 50. LiuW.SeleverJ.MuraliD.Threshold-specific requirements for Bmp4 in mandibular developmentDev Biol28322005282293[PubMed][Google Scholar]
  • 51. ChangW.LinZ.KulessaH.HebertJ.HoganB.L.M.WuD.K.Bmp4 is essential for the formation of the vestibular apparatus that detects angular head movementsPLoS Genet442008e1000050[PubMed][Google Scholar]
  • 52. KingsleyD.M.BlandA.E.GrubberJ.M.The mouse short ear skeletal morphogenesis locus is associated with defects in a bone morphogenetic member of the TGF beta superfamilyCell7131992399410[PubMed][Google Scholar]
  • 53. MikićB.van der MeulenM.C.KingsleyD.M.CarterD.R.Long bone geometry and strength in adult BMP-5 deficient miceBone1641995445454[PubMed][Google Scholar]
  • 54. SollowayM.J.DudleyA.T.BikoffE.K.LyonsK.M.HoganB.L.RobertsonE.J.Mice lacking Bmp6 functionDev Genet2241998321339[PubMed][Google Scholar]
  • 55. PerryM.J.McDougallK.E.HouS.-C.TobiasJ.H.Impaired growth plate function in bmp-6 null miceBone4212008216225[PubMed][Google Scholar]
  • 56. SugiuraK.SuY.-Q.EppigJ.J.Does bone morphogenetic protein 6 (BMP6) affect female fertility in the mouse?Biol Reprod83620109971004[PubMed][Google Scholar]
  • 57. DudleyA.T.LyonsK.M.RobertsonE.J.A requirement for bone morphogenetic protein-7 during development of the mammalian kidney and eyeGenes Dev922199527952807[PubMed][Google Scholar]
  • 58. LuoG.HofmannC.BronckersA.L.SohockiM.BradleyA.KarsentyG.BMP-7 is an inducer of nephrogenesis, and is also required for eye development and skeletal patterningGenes Dev922199528082820[PubMed][Google Scholar]
  • 59. SegkliaA.SeuntjensE.ElkourisM.Bmp7 regulates the survival, proliferation, and neurogenic properties of neural progenitor cells during corticogenesis in the mousePloS One732012e34088[PubMed][Google Scholar]
  • 60. TsengY.-H.KokkotouE.SchulzT.J.New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditureNature4547207200810001004[PubMed][Google Scholar]
  • 61. YasminN.BauerT.ModakM.Identification of bone morphogenetic protein 7 (BMP7) as an instructive factor for human epidermal Langerhans cell differentiationJ Exp Med21012201325972610[PubMed][Google Scholar]
  • 62. TomitaM.AsadaM.AsadaN.Bmp7 maintains undifferentiated kidney progenitor population and determines nephron numbers at birthPloS One882013e73554[PubMed][Google Scholar]
  • 63. TsujiK.CoxK.GamerL.GrafD.EconomidesA.RosenV.Conditional deletion of BMP7 from the limb skeleton does not affect bone formation or fracture repairJ Orthop Res Off Publ Orthop Res Soc2832010384389[Google Scholar]
  • 64. KazamaI.MahoneyZ.MinerJ.H.GrafD.EconomidesA.N.KreidbergJ.A.Podocyte-derived BMP7 is critical for nephron developmentJ Am Soc Nephrol JASN1911200821812191[PubMed][Google Scholar]
  • 65. YingY.LiuX.M.MarbleA.LawsonK.A.ZhaoG.Q.Requirement of Bmp8b for the generation of primordial germ cells in the mouseMol Endocrinol Balt Md147200010531063[Google Scholar]
  • 66. ZhaoG.Q.DengK.LaboskyP.A.LiawL.HoganB.L.The gene encoding bone morphogenetic protein 8B is required for the initiation and maintenance of spermatogenesis in the mouseGenes Dev1013199616571669[PubMed][Google Scholar]
  • 67. LevetS.CiaisD.MerdzhanovaG.Bone morphogenetic protein 9 (BMP9) controls lymphatic vessel maturation and valve formationBlood12242013598607[PubMed][Google Scholar]
  • 68. YoshimatsuY.LeeY.G.AkatsuY.Bone morphogenetic protein-9 inhibits lymphatic vessel formation via activin receptor-like kinase 1 during development and cancer progressionProc Natl Acad Sci U S A1104720131894018945[PubMed][Google Scholar]
  • 69. ChenH.ShiS.AcostaL.BMP10 is essential for maintaining cardiac growth during murine cardiogenesisDev Camb Engl1319200422192231[Google Scholar]
  • 70. McPherronA.C.LawlerA.M.LeeS.J.Regulation of anterior/posterior patterning of the axial skeleton by growth/differentiation factor 11Nat Genet2231999260264[PubMed][Google Scholar]
  • 71. DichmannD.S.YassinH.SerupP.Analysis of pancreatic endocrine development in GDF11-deficient miceDev Dyn Off Publ Am Assoc Anat23511200630163025[Google Scholar]
  • 72. HarmonE.B.ApelqvistA.A.SmartN.G.GuX.OsborneD.H.KimS.K.GDF11 modulates NGN3+ islet progenitor cell number and promotes beta-cell differentiation in pancreas developmentDev Camb Engl13124200461636174[Google Scholar]
  • 73. EsquelaA.F.LeeS.-J.Regulation of metanephric kidney development by growth/differentiation factor 11Dev Biol25722003356370[PubMed][Google Scholar]
  • 74. ShiY.LiuJ.-P.Gdf11 facilitates temporal progression of neurogenesis in the developing spinal cordJ Neurosci Off J Soc Neurosci3132011883893[Google Scholar]
  • 75. WuH.-H.IvkovicS.MurrayR.C.Autoregulation of neurogenesis by GDF11Neuron3722003197207[PubMed][Google Scholar]
  • 76. KimJ.WuH.-H.LanderA.D.LyonsK.M.MatzukM.M.CalofA.L.GDF11 controls the timing of progenitor cell competence in developing retinaScience3085730200519271930[PubMed][Google Scholar]
  • 77. MikicB.FerreiraM.P.BattagliaT.C.HunzikerE.B.Accelerated hypertrophic chondrocyte kinetics in GDF-7 deficient murine tibial growth platesJ Orthop Res Off Publ Orthop Res Soc2672008986990[Google Scholar]
  • 78. MaloulA.RossmeierK.MikicB.PogueV.BattagliaT.Geometric and material contributions to whole bone structural behavior in GDF-7-deficient miceConnect Tissue Res4732006157162[PubMed][Google Scholar]
  • 79. MikicB.EntwistleR.RossmeierK.BierwertL.Effect of GDF-7 deficiency on tail tendon phenotype in miceJ Orthop Res Off Publ Orthop Res Soc2662008834839[Google Scholar]
  • 80. MikicB.BierwertL.TsouD.Achilles tendon characterization in GDF-7 deficient miceJ Orthop Res Off Publ Orthop Res Soc2442006831841[Google Scholar]
  • 81. LeeK.J.MendelsohnM.JessellT.M.Neuronal patterning by BMPs: a requirement for GDF7 in the generation of a discrete class of commissural interneurons in the mouse spinal cordGenes Dev1221199833943407[PubMed][Google Scholar]
  • 82. SettleS.H.RountreeR.B.SinhaA.ThackerA.HigginsK.KingsleyD.M.Multiple joint and skeletal patterning defects caused by single and double mutations in the mouse Gdf6 and Gdf5 genesDev Biol25412003116130[PubMed][Google Scholar]
  • 83. ClendenningD.E.MortlockD.P.The BMP ligand Gdf6 prevents differentiation of coronal suture mesenchyme in early cranial developmentPloS One752012e36789[PubMed][Google Scholar]
  • 84. Asai-CoakwellM.MarchL.DaiX.H.Contribution of growth differentiation factor 6-dependent cell survival to early-onset retinal dystrophiesHum Mol Genet227201314321442[PubMed][Google Scholar]
  • 85. HanelM.L.HenseyC.Eye and neural defects associated with loss of GDF6BMC Dev Biol6200643[PubMed][Google Scholar]
  • 86. TassabehjiM.FangZ.M.HiltonE.N.Mutations in GDF6 are associated with vertebral segmentation defects in Klippel-Feil syndromeHum Mutat298200810171027[PubMed][Google Scholar]
  • 87. MikicB.RossmeierK.BierwertL.Identification of a tendon phenotype in GDF6 deficient miceAnat Rec Hoboken29232009396400[PubMed][Google Scholar]
  • 88. MikicB.RossmeierK.BierwertL.Sexual dimorphism in the effect of GDF-6 deficiency on murine tendonJ Orthop Res Off Publ Orthop Res Soc2712200916031611[Google Scholar]
  • 89. StormE.E.HuynhT.V.CopelandN.G.JenkinsN.A.KingsleyD.M.LeeS.J.Limb alterations in brachypodism mice due to mutations in a new member of the TGF beta-superfamilyNature36864721994639643[PubMed][Google Scholar]
  • 90. StormE.E.KingsleyD.M.GDF5 coordinates bone and joint formation during digit developmentDev Biol209119991127[PubMed][Google Scholar]
  • 91. ChhabraA.ZijerdiD.ZhangJ.KlineA.BalianG.HurwitzS.BMP-14 deficiency inhibits long bone fracture healing: a biochemical, histologic, and radiographic assessmentJ Orthop Trauma1992005629634[PubMed][Google Scholar]
  • 92. ColemanC.M.ScheremetaB.H.BoyceA.T.MauckR.L.TuanR.S.Delayed fracture healing in growth differentiation factor 5-deficient mice: a pilot studyClin Orthop46910201129152924[PubMed][Google Scholar]
  • 93. MasuyaH.NishidaK.FuruichiT.A novel dominant-negative mutation in Gdf5 generated by ENU mutagenesis impairs joint formation and causes osteoarthritis in miceHum Mol Genet1619200723662375[PubMed][Google Scholar]
  • 94. MikicB.SchaletB.J.ClarkR.T.GaschenV.HunzikerE.B.GDF-5 deficiency in mice alters the ultrastructure, mechanical properties and composition of the Achilles tendonJ Orthop Res Off Publ Orthop Res Soc1932001365371[Google Scholar]
  • 95. ZaidiS.H.E.HuangQ.MomenA.RiaziA.HusainM.Growth differentiation factor 5 regulates cardiac repair after myocardial infarctionJ Am Coll Cardiol5522010135143[PubMed][Google Scholar]
  • 96. BattagliaT.C.GDF-5 deficiency alters stress-relaxation properties in mouse skinJ Dermatol Sci3932005192195[PubMed][Google Scholar]
  • 97. YanC.WangP.DeMayoJ.Synergistic roles of bone morphogenetic protein 15 and growth differentiation factor 9 in ovarian functionMol Endocrinol Balt Md1562001854866[Google Scholar]
  • 98. LechleiderR.J.RyanJ.L.GarrettL.Targeted mutagenesis of Smad1 reveals an essential role in chorioallantoic fusionDev Biol24012001157167[PubMed][Google Scholar]
  • 99. TremblayK.D.DunnN.R.RobertsonE.J.Mouse embryos lacking Smad1 signals display defects in extra-embryonic tissues and germ cell formationDev Camb Engl12818200136093621[Google Scholar]
  • 100. HayashiK.KobayashiT.UminoT.GoitsukaR.MatsuiY.KitamuraD.SMAD1 signaling is critical for initial commitment of germ cell lineage from mouse epiblastMech Dev1181–2200299109[PubMed][Google Scholar]
  • 101. WangM.JinH.TangD.HuangS.ZuscikM.J.ChenD.Smad1 plays an essential role in bone development and postnatal bone formationOsteoarthr Cartil OARS Osteoarthr Res Soc1962011751762[Google Scholar]
  • 102. XuB.ChenC.ChenH.Smad1 and its target gene Wif1 coordinate BMP and Wnt signaling activities to regulate fetal lung developmentDev Camb Engl13852011925935[Google Scholar]
  • 103. ChangH.HuylebroeckD.VerschuerenK.GuoQ.MatzukM.M.ZwijsenA.Smad5 knockout mice die at mid-gestation due to multiple embryonic and extraembryonic defectsDev Camb Engl1268199916311642[Google Scholar]
  • 104. ChangH.MatzukM.M.Smad5 is required for mouse primordial germ cell developmentMech Dev1041–220016167[PubMed][Google Scholar]
  • 105. ChangH.ZwijsenA.VogelH.HuylebroeckD.MatzukM.M.Smad5 is essential for left-right asymmetry in miceDev Biol219120007178[PubMed][Google Scholar]
  • 106. ArnoldS.J.MarettoS.IslamA.BikoffE.K.RobertsonE.J.Dose-dependent Smad1, Smad5 and Smad8 signaling in the early mouse embryoDev Biol29612006104118[PubMed][Google Scholar]
  • 107. SirardC.de la PompaJ.L.EliaA.The tumor suppressor gene Smad4/Dpc4 is required for gastrulation and later for anterior development of the mouse embryoGenes Dev1211998107119[PubMed][Google Scholar]
  • 108. TakakuK.MiyoshiH.MatsunagaA.OshimaM.SasakiN.TaketoM.M.Gastric and duodenal polyps in Smad4 (Dpc4) knockout miceCancer Res5924199961136117[PubMed][Google Scholar]
  • 109. TaketoM.M.TakakuK.Gastro-intestinal tumorigenesis in Smad4 mutant miceCytokine Growth Factor Rev111–22000147157[PubMed][Google Scholar]
  • 110. PanD.SchomberT.KalbererC.P.Normal erythropoiesis but severe polyposis and bleeding anemia in Smad4-deficient miceBlood1108200730493055[PubMed][Google Scholar]
  • 111. TanX.WengT.ZhangJ.Smad4 is required for maintaining normal murine postnatal bone homeostasisJ Cell Sci120Pt 13200721622170[PubMed][Google Scholar]
  • 112. YangS.HouZ.YangG.Chondrocyte-specific Smad4 gene conditional knockout results in hearing loss and inner ear malformation in miceDev Dyn Off Publ Am Assoc Anat2388200918971908[Google Scholar]
  • 113. YangS.DengA.HuangD.The role of Smad4 in vestibular development in miceInt J Dev Neurosci Off J Int Soc Dev Neurosci29120111523[Google Scholar]
  • 114. ZhangJ.TanX.LiW.Smad4 is required for the normal organization of the cartilage growth plateDev Biol28422005311322[PubMed][Google Scholar]
  • 115. SartoriR.SchirwisE.BlaauwB.BMP signaling controls muscle massNat Genet4511201313091318[PubMed][Google Scholar]
  • 116. WangJ.XuN.FengX.Targeted disruption of Smad4 in cardiomyocytes results in cardiac hypertrophy and heart failureCirc Res9782005821828[PubMed][Google Scholar]
  • 117. LanY.LiuB.YaoH.Essential role of endothelial Smad4 in vascular remodeling and integrityMol Cell Biol2721200776837692[PubMed][Google Scholar]
  • 118. QiX.YangG.YangL.Essential role of Smad4 in maintaining cardiomyocyte proliferation during murine embryonic heart developmentDev Biol31112007136146[PubMed][Google Scholar]
  • 119. MaoX.DebenedittisP.SunY.Vascular smooth muscle cell Smad4 gene is important for mouse vascular developmentArterioscler Thromb Vasc Biol329201221712177[PubMed][Google Scholar]
  • 120. ZhouY.-X.ZhaoM.LiD.Cerebellar deficits and hyperactivity in mice lacking Smad4J Biol Chem2784320034231342320[PubMed][Google Scholar]
  • 121. LiuY.LinD.Necessity of Smad4 for the normal development of the mouse lacrimal glandJpn J Ophthalmol5832014298306[PubMed][Google Scholar]
  • 122. ArchambeaultD.R.YaoH.H.-C.Loss of smad4 in Sertoli and Leydig cells leads to testicular dysgenesis and hemorrhagic tumor formation in miceBiol Reprod903201462[PubMed][Google Scholar]
  • 123. YuC.ZhangY.-L.FanH.-Y.Selective Smad4 knockout in ovarian preovulatory follicles results in multiple defects in ovulationMol Endocrinol Balt Md2762013966978[Google Scholar]
  • 124. PangasS.A.LiX.RobertsonE.J.MatzukM.M.Premature luteinization and cumulus cell defects in ovarian-specific Smad4 knockout miceMol Endocrinol Balt Md206200614061422[Google Scholar]
  • 125. OwensP.EngelkingE.HanG.HaegerS.M.WangX.-J.Epidermal Smad4 deletion results in aberrant wound healingAm J Pathol17612010122133[PubMed][Google Scholar]
  • 126. QiaoW.LiA.G.OwensP.XuX.WangX.-J.DengC.-X.Hair follicle defects and squamous cell carcinoma formation in Smad4 conditional knockout mouse skinOncogene2522006207217[PubMed][Google Scholar]
  • 127. YangL.LiW.WangS.Smad4 disruption accelerates keratinocyte reepithelialization in murine cutaneous wound repairHistochem Cell Biol13842012573582[PubMed][Google Scholar]
  • 128. BornsteinS.WhiteR.MalkoskiS.Smad4 loss in mice causes spontaneous head and neck cancer with increased genomic instability and inflammationJ Clin Invest11911200934083419[PubMed][Google Scholar]
  • 129. GaoY.YangG.WengT.Disruption of Smad4 in odontoblasts causes multiple keratocystic odontogenic tumors and tooth malformation in miceMol Cell Biol2921200959415951[PubMed][Google Scholar]
  • 130. EstradaK.D.RettingK.N.ChinA.M.LyonsK.M.Smad6 is essential to limit BMP signaling during cartilage developmentJ Bone Min Res Off J Am Soc Bone Min Res2610201124982510[Google Scholar]
  • 131. GalvinK.M.DonovanM.J.LynchC.A.A role for smad6 in development and homeostasis of the cardiovascular systemNat Genet2422000171174[PubMed][Google Scholar]
  • 132. ChenQ.ChenH.ZhengD.Smad7 is required for the development and function of the heartJ Biol Chem28412009292300[PubMed][Google Scholar]
  • 133. EstradaK.D.WangW.RettingK.N.Smad7 regulates terminal maturation of chondrocytes in the growth plateDev Biol38222013375384[PubMed][Google Scholar]
  • 134. TojoM.TakebeA.TakahashiS.Smad7-deficient mice show growth retardation with reduced viabilityJ Biochem (Tokyo)15162012621631[PubMed][Google Scholar]
  • 135. ChungA.C.K.HuangX.R.ZhouL.HeuchelR.LaiK.N.LanH.Y.Disruption of the Smad7 gene promotes renal fibrosis and inflammation in unilateral ureteral obstruction (UUO) in miceNephrol Dial Transpl Off Publ Eur Dial Transpl Assoc - Eur Ren Assoc245200914431454[Google Scholar]
  • 136. ZhangR.HuangH.CaoP.WangZ.ChenY.PanY.Sma- and Mad-related protein 7 (Smad7) is required for embryonic eye development in the mouseJ Biol Chem2881520131027510285[PubMed][Google Scholar]
  • 137. DongC.ZhuS.WangT.Deficient Smad7 expression: a putative molecular defect in sclerodermaProc Natl Acad Sci U S A996200239083913[PubMed][Google Scholar]
  • 138. LiR.RosendahlA.BrodinG.Deletion of exon I of SMAD7 in mice results in altered B cell responsesJ Immunol Balt Md17611200667776784[Google Scholar]
  • 139. KleiterI.SongJ.LukasD.Smad7 in T cells drives T helper 1 responses in multiple sclerosis and experimental autoimmune encephalomyelitisBrain J Neurol133Pt 4201010671081[Google Scholar]
  • 140. MishinaY.SuzukiA.UenoN.BehringerR.R.Bmpr encodes a type I bone morphogenetic protein receptor that is essential for gastrulation during mouse embryogenesisGenes Dev924199530273037[PubMed][Google Scholar]
  • 141. ParkC.LavineK.MishinaY.DengC.-X.OrnitzD.M.ChoiK.Bone morphogenetic protein receptor 1A signaling is dispensable for hematopoietic development but essential for vessel and atrioventricular endocardial cushion formationDev Camb Engl13317200634733484[Google Scholar]
  • 142. SunJ.LiuY.-H.ChenH.Deficient Alk3-mediated BMP signaling causes prenatal omphalocele-like defectBiochem Biophys Res Commun36012007238243[PubMed][Google Scholar]
  • 143. KamiyaN.YeL.KobayashiT.BMP signaling negatively regulates bone mass through sclerostin by inhibiting the canonical Wnt pathwayDev Camb Engl13522200838013811[Google Scholar]
  • 144. JingJ.RenY.ZongZ.BMP receptor 1A determines the cell fate of the postnatal growth plateInt J Biol Sci992013895906[PubMed][Google Scholar]
  • 145. SteinbickerA.U.BartnikasT.B.LohmeyerL.K.Perturbation of hepcidin expression by BMP type I receptor deletion induces iron overload in miceBlood11815201142244230[PubMed][Google Scholar]
  • 146. GaussinV.Van de PutteT.MishinaY.Endocardial cushion and myocardial defects after cardiac myocyte-specific conditional deletion of the bone morphogenetic protein receptor ALK3Proc Natl Acad Sci U S A995200228782883[PubMed][Google Scholar]
  • 147. GaussinV.MorleyG.E.CoxL.Alk3/Bmpr1a receptor is required for development of the atrioventricular canal into valves and annulus fibrosusCirc Res9732005219226[PubMed][Google Scholar]
  • 148. SongL.FässlerR.MishinaY.JiaoK.BaldwinH.S.Essential functions of Alk3 during AV cushion morphogenesis in mouse embryonic heartsDev Biol30112007276286[PubMed][Google Scholar]
  • 149. El-BizriN.GuignabertC.WangL.SM22alpha-targeted deletion of bone morphogenetic protein receptor 1A in mice impairs cardiac and vascular development, and influences organogenesisDev Camb Engl13517200829812991[Google Scholar]
  • 150. El-BizriN.WangL.MerklingerS.L.Smooth muscle protein 22alpha-mediated patchy deletion of Bmpr1a impairs cardiac contractility but protects against pulmonary vascular remodelingCirc Res10232008380388[PubMed][Google Scholar]
  • 151. EblaghieM.C.ReedyM.OliverT.MishinaY.HoganB.L.M.Evidence that autocrine signaling through Bmpr1a regulates the proliferation, survival and morphogenetic behavior of distal lung epithelial cellsDev Biol291120066782[PubMed][Google Scholar]
  • 152. SunJ.ChenH.ChenC.Prenatal lung epithelial cell-specific abrogation of Alk3-bone morphogenetic protein signaling causes neonatal respiratory distress by disrupting distal airway formationAm J Pathol17232008571582[PubMed][Google Scholar]
  • 153. HartwigS.BridgewaterD.Di GiovanniV.CainJ.MishinaY.RosenblumN.D.BMP receptor ALK3 controls collecting system developmentJ Am Soc Nephrol JASN1912008117124[PubMed][Google Scholar]
  • 154. PengC.-Y.MukhopadhyayA.JarrettJ.C.YoshikawaK.KesslerJ.A.BMP receptor 1A regulates development of hypothalamic circuits critical for feeding behaviorJ Neurosci Off J Soc Neurosci324820121721117224[Google Scholar]
  • 155. ZhaoQ.ZhaoJ.-Y.WuD.LuX.-C.ZhangJ.-S.ZhangZ.-Y.Mutually inductive interactions between the lens and retina require ALK3 functions during mouse embryonic developmentInt J Ophthalmol522012119124[PubMed][Google Scholar]
  • 156. WuX.ZhangN.LeeM.M.Mullerian inhibiting substance recruits ALK3 to regulate Leydig cell differentiationEndocrinology15310201249294937[PubMed][Google Scholar]
  • 157. EdsonM.A.NalamR.L.ClementiC.Granulosa cell-expressed BMPR1A and BMPR1B have unique functions in regulating fertility but act redundantly to suppress ovarian tumor developmentMol Endocrinol Balt Md246201012511266[Google Scholar]
  • 158. LiuW.SunX.BrautA.Distinct functions for Bmp signaling in lip and palate fusion in miceDev Camb Engl1326200514531461[Google Scholar]
  • 159. YuhkiM.YamadaM.KawanoM.BMPR1A signaling is necessary for hair follicle cycling and hair shaft differentiation in miceDev Camb Engl1318200418251833[Google Scholar]
  • 160. YangZ.HaiB.QinL.Cessation of epithelial Bmp signaling switches the differentiation of crown epithelia to the root lineage in a β-catenin-dependent mannerMol Cell Biol3323201347324744[PubMed][Google Scholar]
  • 161. YiS.E.DaluiskiA.PedersonR.RosenV.LyonsK.M.The type I BMP receptor BMPRIB is required for chondrogenesis in the mouse limbDev Camb Engl12732000621630[Google Scholar]
  • 162. LiuJ.WilsonS.RehT.BMP receptor 1b is required for axon guidance and cell survival in the developing retinaDev Biol256120033448[PubMed][Google Scholar]
  • 163. YiS.E.LaPoltP.S.YoonB.S.ChenJ.Y.LuJ.K.LyonsK.M.The type I BMP receptor BmprIB is essential for female reproductive functionProc Natl Acad Sci U S A9814200179947999[PubMed][Google Scholar]
  • 164. KomatsuY.ScottG.NagyA.KaartinenV.MishinaY.BMP type I receptor ALK2 is essential for proper patterning at late gastrulation during mouse embryogenesisDev Dyn Off Publ Am Assoc Anat23622007512517[Google Scholar]
  • 165. RajagopalR.DattiloL.K.KaartinenV.Functions of the type 1 BMP receptor Acvr1 (Alk2) in lens development: cell proliferation, terminal differentiation, and survivalInvest Ophthalmol Vis Sci4911200849534960[PubMed][Google Scholar]
  • 166. DudasM.SridurongritS.NagyA.OkazakiK.KaartinenV.Craniofacial defects in mice lacking BMP type I receptor Alk2 in neural crest cellsMech Dev12122004173182[PubMed][Google Scholar]
  • 167. KaartinenV.DudasM.NagyA.SridurongritS.LuM.M.EpsteinJ.A.Cardiac outflow tract defects in mice lacking ALK2 in neural crest cellsDev Camb Engl13114200434813490[Google Scholar]
  • 168. WangJ.SridurongritS.DudasM.Atrioventricular cushion transformation is mediated by ALK2 in the developing mouse heartDev Biol28612005299310[PubMed][Google Scholar]
  • 169. ThomasP.S.SridurongritS.Ruiz-LozanoP.KaartinenV.Deficient signaling via Alk2 (Acvr1) leads to bicuspid aortic valve developmentPloS One742012e35539[PubMed][Google Scholar]
  • 170. ClementiC.TripuraniS.K.LargeM.J.Activin-like kinase 2 functions in peri-implantation uterine signaling in mice and humansPLoS Genet9112013e1003863[PubMed][Google Scholar]
  • 171. NagashimaT.LiQ.ClementiC.LydonJ.P.DeMayoF.J.MatzukM.M.BMPR2 is required for postimplantation uterine function and pregnancy maintenanceJ Clin Invest1236201325392550[PubMed][Google Scholar]
  • 172. BeppuH.KawabataM.HamamotoT.BMP type II receptor is required for gastrulation and early development of mouse embryosDev Biol22112000249258[PubMed][Google Scholar]
  • 173. BeppuH.MalhotraR.BeppuY.LeporeJ.J.ParmacekM.S.BlochK.D.BMP type II receptor regulates positioning of outflow tract and remodeling of atrioventricular cushion during cardiogenesisDev Biol33122009167175[PubMed][Google Scholar]
  • 174. BeppuH.IchinoseF.KawaiN.BMPR-II heterozygous mice have mild pulmonary hypertension and an impaired pulmonary vascular remodeling response to prolonged hypoxiaAm J Physiol Lung Cell Mol Physiol28762004L1241L1247[PubMed][Google Scholar]
  • 175. WestJ.HarralJ.LaneK.Mice expressing BMPR2R899X transgene in smooth muscle develop pulmonary vascular lesionsAm J Physiol Lung Cell Mol Physiol29552008L744L755[PubMed][Google Scholar]
  • 176. HongK.-H.LeeY.J.LeeE.Genetic ablation of the BMPR2 gene in pulmonary endothelium is sufficient to predispose to pulmonary arterial hypertensionCirculation11872008722730[PubMed][Google Scholar]
  • 177. MatzukM.M.KumarT.R.BradleyA.Different phenotypes for mice deficient in either activins or activin receptor type IINature37465201995356360[PubMed][Google Scholar]
  • 178. MaX.ReynaA.ManiS.K.MatzukM.M.KumarT.R.Impaired male sexual behavior in activin receptor type II knockout miceBiol Reprod736200511821190[PubMed][Google Scholar]
  • 179. WrefordN.G.Rajendra KumarT.MatzukM.M.de KretserD.M.Analysis of the testicular phenotype of the follicle-stimulating hormone beta-subunit knockout and the activin type II receptor knockout mice by stereological analysisEndocrinology1427200129162920[PubMed][Google Scholar]
  • 180. OhS.P.LiE.The signaling pathway mediated by the type IIB activin receptor controls axial patterning and lateral asymmetry in the mouseGenes Dev1114199718121826[PubMed][Google Scholar]
  • 181. StormE.E.KingsleyD.M.Joint patterning defects caused by single and double mutations in members of the bone morphogenetic protein (BMP) familyDev Camb Engl12212199639693979[Google Scholar]
  • 182. WilliamsE.S.UhasK.A.BunkeB.P.GarberK.B.MartinC.L.Cleft palate in a multigenerational family with a microdeletion of 20p12.3 involving BMP2Am J Med Genet A158A10201226162620[PubMed][Google Scholar]
  • 183. SuazoJ.TapiaJ.C.SantosJ.L.CastroV.G.ColomboA.BlancoR.Risk variants in BMP4 promoters for nonsyndromic cleft lip/palate in a Chilean populationBMC Med Genet122011163[PubMed][Google Scholar]
  • 184. KaplanF.S.Le MerrerM.GlaserD.L.Fibrodysplasia ossificans progressivaBest Pract Res Clin Rheumatol2212008191205[PubMed][Google Scholar]
  • 185. KaplanF.S.XuM.SeemannP.Classic and atypical fibrodysplasia ossificans progressiva (FOP) phenotypes are caused by mutations in the bone morphogenetic protein (BMP) type I receptor ACVR1Hum Mutat3032009379390[PubMed][Google Scholar]
  • 186. OhteS.ShinM.SasanumaH.A novel mutation of ALK2, L196P, found in the most benign case of fibrodysplasia ossificans progressiva activates BMP-specific intracellular signaling equivalent to a typical mutation, R206HBiochem Biophys Res Commun40712011213218[PubMed][Google Scholar]
  • 187. FukudaT.KanomataK.NojimaJ.A unique mutation of ALK2, G356D, found in a patient with fibrodysplasia ossificans progressiva is a moderately activated BMP type I receptorBiochem Biophys Res Commun37732008905909[PubMed][Google Scholar]
  • 188. BagarovaJ.VonnerA.J.ArmstrongK.A.Constitutively active ALK2 receptor mutants require type II receptor cooperationMol Cell Biol3312201324132424[PubMed][Google Scholar]
  • 189. LeV.Q.WhartonK.A.Hyperactive BMP signaling induced by ALK2(R206H) requires type II receptor function in a Drosophila model for classic fibrodysplasia ossificans progressivaDev Dyn Off Publ Am Assoc Anat24112012200214[Google Scholar]
  • 190. ChaikuadA.AlfanoI.KerrG.Structure of the bone morphogenetic protein receptor ALK2 and implications for fibrodysplasia ossificans progressivaJ Biol Chem2874420123699036998[PubMed][Google Scholar]
  • 191. ShenQ.LittleS.C.XuM.The fibrodysplasia ossificans progressiva R206H ACVR1 mutation activates BMP-independent chondrogenesis and zebrafish embryo ventralizationJ Clin Invest11911200934623472[PubMed][Google Scholar]
  • 192. SongG.-A.KimH.-J.WooK.-M.Molecular consequences of the ACVR1(R206H) mutation of fibrodysplasia ossificans progressivaJ Biol Chem2852920102254222553[PubMed][Google Scholar]
  • 193. FioriJ.L.BillingsP.C.de la PeñaL.S.KaplanF.S.ShoreE.M.Dysregulation of the BMP-p38 MAPK signaling pathway in cells from patients with fibrodysplasia ossificans progressiva (FOP)J Bone Min Res Off J Am Soc Bone Min Res2162006902909[Google Scholar]
  • 194. YuP.B.DengD.Y.LaiC.S.BMP type I receptor inhibition reduces heterotopic [corrected] ossificationNat Med1412200813631369[PubMed][Google Scholar]
  • 195. SanvitaleC.E.KerrG.ChaikuadA.A new class of small molecule inhibitor of BMP signalingPloS One842013e62721[PubMed][Google Scholar]
  • 196. ShiS.CaiJ.de GorterD.J.J.Antisense-oligonucleotide mediated exon skipping in activin-receptor-like kinase 2: inhibiting the receptor that is overactive in fibrodysplasia ossificans progressivaPloS One872013e69096[PubMed][Google Scholar]
  • 197. MariniJ.C.BlissettA.R.New genes in bone development: what’s new in osteogenesis imperfectaJ Clin Endocrinol Metab988201330953103[PubMed][Google Scholar]
  • 198. Martínez-GlezV.ValenciaM.Caparrós-MartínJ.A.Identification of a mutation causing deficient BMP1/mTLD proteolytic activity in autosomal recessive osteogenesis imperfectaHum Mutat3322012343350[PubMed][Google Scholar]
  • 199. HopkinsD.R.KelesS.GreenspanD.S.The bone morphogenetic protein 1/Tolloid-like metalloproteinasesMatrix Biol J Int Soc Matrix Biol2672007508523[Google Scholar]
  • 200. AsharaniP.V.KeuppK.SemlerO.Attenuated BMP1 function compromises osteogenesis, leading to bone fragility in humans and zebrafishAm J Hum Genet9042012661674[PubMed][Google Scholar]
  • 201. WilkinsJ.M.SouthamL.MustafaZ.ChapmanK.LoughlinJ.Association of a functional microsatellite within intron 1 of the BMP5 gene with susceptibility to osteoarthritisBMC Med Genet102009141[PubMed][Google Scholar]
  • 202. SouthamL.DowlingB.FerreiraA.Microsatellite association mapping of a primary osteoarthritis susceptibility locus on chromosome 6p12.3-q13Arthritis Rheum5012200439103914[PubMed][Google Scholar]
  • 203. MiyamotoY.MabuchiA.ShiD.A functional polymorphism in the 5’ UTR of GDF5 is associated with susceptibility to osteoarthritisNat Genet3942007529533[PubMed][Google Scholar]
  • 204. NakaseT.MiyajiT.TomitaT.Localization of bone morphogenetic protein-2 in human osteoarthritic cartilage and osteophyteOsteoarthr Cartil OARS Osteoarthr Res Soc1142003278284[Google Scholar]
  • 205. PapathanasiouI.MalizosK.N.TsezouA.Bone morphogenetic protein-2-induced Wnt/β-catenin signaling pathway activation through enhanced low-density-lipoprotein receptor-related protein 5 catabolic activity contributes to hypertrophy in osteoarthritic chondrocytesArthritis Res Ther1422012R82[PubMed][Google Scholar]
  • 206. AlbiliaJ.B.TenenbaumH.C.ClokieC.M.L.Serum levels of BMP-2, 4, 7 and AHSG in patients with degenerative joint disease requiring total arthroplasty of the hip and temporomandibular jointsJ Orthop Res Off Publ Orthop Res Soc31120134452[Google Scholar]
  • 207. BijsterboschJ.KloppenburgM.ReijnierseM.Association study of candidate genes for the progression of hand osteoarthritisOsteoarthr Cartil OARS Osteoarthr Res Soc2142013565569[Google Scholar]
  • 208. BadlaniN.OshimaY.HealeyR.CouttsR.AmielD.Use of bone morphogenic protein-7 as a treatment for osteoarthritisClin Orthop46712200932213229[PubMed][Google Scholar]
  • 209. HayashiM.MunetaT.TakahashiT.JuY.-J.TsujiK.SekiyaI.Intra-articular injections of bone morphogenetic protein-7 retard progression of existing cartilage degenerationJ Orthop Res Off Publ Orthop Res Soc2811201015021506[Google Scholar]
  • 210. HunterD.J.PikeM.C.JonasB.L.KissinE.KropJ.McAlindonT.Phase 1 safety and tolerability study of BMP-7 in symptomatic knee osteoarthritisBMC Musculoskelet Disord112322010[Google Scholar]
  • 211. SrinivasanP.P.McCoyS.Y.JhaA.K.Injectable perlecan domain 1-hyaluronan microgels potentiate the cartilage repair effect of BMP2 in a murine model of early osteoarthritisBiomed Mater Bristol Engl722012024109[Google Scholar]
  • 212. WinbanksC.E.ChenJ.L.QianH.The bone morphogenetic protein axis is a positive regulator of skeletal muscle massJ Cell Biol20322013345357[PubMed][Google Scholar]
  • 213. Le GoffC.MahautC.AbhyankarA.Mutations at a single codon in Mad homology 2 domain of SMAD4 cause Myhre syndromeNat Genet44120128588[Google Scholar]
  • 214. BattsL.E.PolkD.B.DuboisR.N.KulessaH.Bmp signaling is required for intestinal growth and morphogenesisDev Dyn Off Publ Am Assoc Anat2356200615631570[Google Scholar]
  • 215. TorihashiS.HattoriT.HasegawaH.KurahashiM.OgaeriT.FujimotoT.The expression and crucial roles of BMP signaling in development of smooth muscle progenitor cells in the mouse embryonic gutDiffer Res Biol Divers7732009277289[Google Scholar]
  • 216. MaloumF.AllaireJ.M.Gagné-SansfaçonJ.Epithelial BMP signaling is required for proper specification of epithelial cell lineages and gastric endocrine cellsAm J Physiol Gastrointest Liver Physiol30062011G1065G1079[PubMed][Google Scholar]
  • 217. CastilloD.PuigS.IglesiasM.Activation of the BMP4 pathway and early expression of CDX2 characterize non-specialized columnar metaplasia in a human model of Barrett’s esophagusJ Gastrointest Surg Off J Soc Surg Aliment Tract1622012227237discussion 237[Google Scholar]
  • 218. MilanoF.van BaalJ.W.P.M.ButtarN.S.Bone morphogenetic protein 4 expressed in esophagitis induces a columnar phenotype in esophageal squamous cellsGastroenterology1327200724122421[PubMed][Google Scholar]
  • 219. YamanakaY.ShiotaniA.FujimuraY.Expression of Sonic hedgehog (SHH) and CDX2 in the columnar epithelium of the lower oesophagusDig Liver Dis Off J Ital Soc Gastroenterol Ital Assoc Study Liver43120115459[Google Scholar]
  • 220. WangD.H.ClemonsN.J.MiyashitaT.Aberrant epithelial-mesenchymal Hedgehog signaling characterizes Barrett’s metaplasiaGastroenterology1385201018101822[PubMed][Google Scholar]
  • 221. Van BaalJ.W.P.M.VerbeekR.E.BusP.microRNA-145 in Barrett's oesophagus: regulating BMP4 signalling via GATA6Gut6252013664675[PubMed][Google Scholar]
  • 222. JeeM.J.YoonS.M.KimE.J.A novel germline mutation in exon 10 of the SMAD4 gene in a familial juvenile polyposisGut Liver762013747751[PubMed][Google Scholar]
  • 223. HoweJ.R.RothS.RingoldJ.C.Mutations in the SMAD4/DPC4 gene in juvenile polyposisScience2805366199810861088[PubMed][Google Scholar]
  • 224. HoweJ.R.BairJ.L.SayedM.G.Germline mutations of the gene encoding bone morphogenetic protein receptor 1A in juvenile polyposisNat Genet2822001184187[PubMed][Google Scholar]
  • 225. CarrJ.C.DahdalehF.S.WangD.HoweJ.R.Germline mutations in SMAD4 disrupt bone morphogenetic protein signalingJ Surg Res17422012211214[PubMed][Google Scholar]
  • 226. HoweJ.R.DahdalehF.S.CarrJ.C.WangD.ShermanS.K.HoweJ.R.BMPR1A mutations in juvenile polyposis affect cellular localizationJ Surg Res18422013739745[PubMed][Google Scholar]
  • 227. SepterS.ZhangL.LawsonC.E.CocjinJ.AttardT.ArdingerH.H.Aggressive juvenile polyposis in children with chromosome 10q23 deletionWorld J Gastroenterol WJG1914201322862292[PubMed][Google Scholar]
  • 228. ChauJ.F.L.JiaD.WangZ.A crucial role for bone morphogenetic protein-Smad1 signalling in the DNA damage responseNat Commun38362012[Google Scholar]
  • 229. KodachL.L.WiercinskaE.de MirandaN.F.C.C.The bone morphogenetic protein pathway is inactivated in the majority of sporadic colorectal cancersGastroenterology1345200813321341[PubMed][Google Scholar]
  • 230. YokoeT.OhmachiT.InoueH.Clinical significance of growth differentiation factor 11 in colorectal cancerInt J Oncol315200710971101[PubMed][Google Scholar]
  • 231. ZouH.HarringtonJ.J.ShireA.M.Highly methylated genes in colorectal neoplasia: implications for screeningCancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol1612200726862696[Google Scholar]
  • 232. KisielJ.B.YabT.C.TaylorW.R.Stool DNA testing for the detection of pancreatic cancer: assessment of methylation marker candidatesCancer11810201226232631[PubMed][Google Scholar]
  • 233. LohK.ChiaJ.A.GrecoS.Bone morphogenic protein 3 inactivation is an early and frequent event in colorectal cancer developmentGenes Chromosom Cancer4762008449460[PubMed][Google Scholar]
  • 234. SlatteryM.L.LundgreenA.HerrickJ.S.Genetic variation in bone morphogenetic protein and colon and rectal cancerInt J Cancer J Int Cancer13032012653664[Google Scholar]
  • 235. KimR.Y.RobertsonE.J.SollowayM.J.Bmp6 and Bmp7 are required for cushion formation and septation in the developing mouse heartDev Biol23522001449466[PubMed][Google Scholar]
  • 236. CaiJ.PardaliE.Sánchez-DuffhuesG.ten DijkeP.BMP signaling in vascular diseasesFEBS Lett58614201219932002[PubMed][Google Scholar]
  • 237. CoganJ.AustinE.HedgesL.Role of BMPR2 alternative splicing in heritable pulmonary arterial hypertension penetranceCirculation12615201219071916[PubMed][Google Scholar]
  • 238. LiuD.WuW.-H.MaoY.-M.BMPR2 mutations influence phenotype more obviously in male patients with pulmonary arterial hypertensionCirc Cardiovasc Genet552012511518[PubMed][Google Scholar]
  • 239. JiangY.NoheA.BragdonB.Trapping of BMP receptors in distinct membrane domains inhibits their function in pulmonary arterial hypertensionAm J Physiol Lung Cell Mol Physiol30122011L218L227[PubMed][Google Scholar]
  • 240. JohnsonJ.A.HemnesA.R.PerrienD.S.Cytoskeletal defects in Bmpr2-associated pulmonary arterial hypertensionAm J Physiol Lung Cell Mol Physiol30252012L474L484[PubMed][Google Scholar]
  • 241. YuP.B.BeppuH.KawaiN.LiE.BlochK.D.Bone morphogenetic protein (BMP) type II receptor deletion reveals BMP ligand-specific gain of signaling in pulmonary artery smooth muscle cellsJ Biol Chem2802620052444324450[PubMed][Google Scholar]
  • 242. DewachterL.AdnotS.GuignabertC.Bone morphogenetic protein signalling in heritable versus idiopathic pulmonary hypertensionEur Respir J345200911001110[PubMed][Google Scholar]
  • 243. UptonP.D.DaviesR.J.TajsicT.MorrellN.W.Transforming growth factor-β(1) represses bone morphogenetic protein-mediated Smad signaling in pulmonary artery smooth muscle cells via Smad3Am J Respir Cell Mol Biol496201311351145[PubMed][Google Scholar]
  • 244. DrakeK.M.ZygmuntD.MavrakisL.Altered MicroRNA processing in heritable pulmonary arterial hypertension: an important role for Smad-8Am J Respir Crit Care Med18412201114001408[PubMed][Google Scholar]
  • 245. HarrisonR.E.FlanaganJ.A.SankeloM.Molecular and functional analysis identifies ALK-1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic telangiectasiaJ Med Genet40122003865871[PubMed][Google Scholar]
  • 246. AbdallaS.A.GallioneC.J.BarstR.J.Primary pulmonary hypertension in families with hereditary haemorrhagic telangiectasiaEur Respir J2332004373377[PubMed][Google Scholar]
  • 247. YangJ.LiX.Al-LamkiR.S.Sildenafil potentiates bone morphogenetic protein signaling in pulmonary arterial smooth muscle cells and in experimental pulmonary hypertensionArterioscler Thromb Vasc Biol33120133442[PubMed][Google Scholar]
  • 248. YangJ.LiX.LiY.Id proteins are critical downstream effectors of BMP signaling in human pulmonary arterial smooth muscle cellsAm J Physiol Lung Cell Mol Physiol30542013L312L321[PubMed][Google Scholar]
  • 249. DrakeK.M.DunmoreB.J.McNellyL.N.MorrellN.W.AldredM.A.Correction of nonsense BMPR2 and SMAD9 mutations by ataluren in pulmonary arterial hypertensionAm J Respir Cell Mol Biol4932013403409[PubMed][Google Scholar]
  • 250. TeekakirikulP.MilewiczD.M.MillerD.T.Thoracic aortic disease in two patients with juvenile polyposis syndrome and SMAD4 mutationsAm J Med Genet A161A12013185191[PubMed][Google Scholar]
  • 251. RicardN.BidartM.MalletC.Functional analysis of the BMP9 response of ALK1 mutants from HHT2 patients: a diagnostic tool for novel ACVRL1 mutationsBlood1169201016041612[PubMed][Google Scholar]
  • 252. ChoiE.-J.KimY.H.ChoeS.Enhanced responses to angiogenic cues underlie the pathogenesis of hereditary hemorrhagic telangiectasia 2PloS One852013e63138[PubMed][Google Scholar]
  • 253. MitchellD.PobreE.G.MulivorA.W.ALK1-Fc inhibits multiple mediators of angiogenesis and suppresses tumor growthMol Cancer Ther922010379388[PubMed][Google Scholar]
  • 254. TabatabaeifarM.SchlingmannK.-P.LitwinM.Functional analysis of BMP4 mutations identified in pediatric CAKUT patientsPediatr Nephrol Berl Ger2412200923612368[Google Scholar]
  • 255. WeberS.TaylorJ.C.WinyardP.SIX2 and BMP4 mutations associate with anomalous kidney developmentJ Am Soc Nephrol JASN1952008891903[PubMed][Google Scholar]
  • 256. SchildR.KnüppelT.KonradM.Double homozygous missense mutations in DACH1 and BMP4 in a patient with bilateral cystic renal dysplasiaNephrol Dial Transpl Off Publ Eur Dial Transpl Assoc - Eur Ren Assoc2812013227232[Google Scholar]
  • 257. MorganE.A.NguyenS.B.ScottV.StadlerH.S.Loss of Bmp7 and Fgf8 signaling in Hoxa13-mutant mice causes hypospadiaDev Camb Engl13014200330953109[Google Scholar]
  • 258. ChenT.LiQ.XuJ.Mutation screening of BMP4, BMP7, HOXA4 and HOXB6 genes in Chinese patients with hypospadiasEur J Hum Genet EJHG15120072328[PubMed][Google Scholar]
  • 259. HeJ.L.LiuJ.H.LiuF.TanP.LinT.LiX.L.Mutation screening of BMP4 and Id2 genes in Chinese patients with congenital ureteropelvic junction obstructionEur J Pediatr17132012451456[PubMed][Google Scholar]
  • 260. TripathiP.WangY.CaseyA.M.ChenF.Absence of canonical Smad signaling in ureteral and bladder mesenchyme causes ureteropelvic junction obstructionJ Am Soc Nephrol JASN2342012618628[PubMed][Google Scholar]
  • 261. WetzelP.HaagJ.CâmpeanV.Bone morphogenetic protein-7 expression and activity in the human adult normal kidney is predominantly localized to the distal nephronKidney Int7042006717723[PubMed][Google Scholar]
  • 262. MituG.HirschbergR.Bone morphogenetic protein-7 (BMP7) in chronic kidney diseaseFront Biosci J Virtual Libr13200847264739[Google Scholar]
  • 263. BramlageC.P.TampeB.KoziolekM.Bone morphogenetic protein (BMP)-7 expression is decreased in human hypertensive nephrosclerosisBMC Nephrol11201031[PubMed][Google Scholar]
  • 264. BramlageC.P.MüllerG.A.TampeB.The role of bone morphogenetic protein-5 (BMP-5) in human nephrosclerosisJ Nephrol2452011647655[PubMed][Google Scholar]
  • 265. GrawJ.Eye developmentCurr Top Dev Biol902010343386[PubMed][Google Scholar]
  • 266. WyattA.W.OsborneR.J.StewartH.RaggeN.K.Bone morphogenetic protein 7 (BMP7) mutations are associated with variable ocular, brain, ear, palate, and skeletal anomaliesHum Mutat3172010781787[PubMed][Google Scholar]
  • 267. TakenouchiT.NishinaS.KosakiR.Concurrent deletion of BMP4 and OTX2 genes, two master genes in ophthalmogenesisEur J Med Genet56120135053[PubMed][Google Scholar]
  • 268. BakraniaP.EfthymiouM.KleinJ.C.Mutations in BMP4 cause eye, brain, and digit developmental anomalies: overlap between the BMP4 and hedgehog signaling pathwaysAm J Hum Genet8222008304319[PubMed][Google Scholar]
  • 269. HayashiS.OkamotoN.MakitaY.HataA.ImotoI.InazawaJ.Heterozygous deletion at 14q22.1-q22.3 including the BMP4 gene in a patient with psychomotor retardation, congenital corneal opacity and feet polysyndactylyAm J Med Genet A146A22200829052910[PubMed][Google Scholar]
  • 270. ReisL.M.TylerR.C.SchilterK.F.BMP4 loss-of-function mutations in developmental eye disorders including SHORT syndromeHum Genet13042011495504[PubMed][Google Scholar]
  • 271. HuangH.SongT.-J.LiX.BMP signaling pathway is required for commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineageProc Natl Acad Sci U S A1063120091267012675[PubMed][Google Scholar]
  • 272. ElsenM.RaschkeS.TennagelsN.BMP4 and BMP7 induce the white-to-brown transition of primary human adipose stem cellsAm J Physiol Cell Physiol30652014C431C440[PubMed][Google Scholar]
  • 273. BöttcherY.UnbehauenH.KlötingN.Adipose tissue expression and genetic variants of the bone morphogenetic protein receptor 1A gene (BMPR1A) are associated with human obesityDiabetes589200921192128[PubMed][Google Scholar]
  • 274. SchleinitzD.KlötingN.BöttcherY.Genetic and evolutionary analyses of the human bone morphogenetic protein receptor 2 (BMPR2) in the pathophysiology of obesityPloS One622011e16155[PubMed][Google Scholar]
  • 275. TownsendK.L.SuzukiR.HuangT.L.Bone morphogenetic protein 7 (BMP7) reverses obesity and regulates appetite through a central mTOR pathwayFASEB J Off Publ Fed Am Soc Exp Biol265201221872196[Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.