Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Nat Commun ; 12(1): 4950, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400635

ABSTRACT

Upon ligand binding, bone morphogenetic protein (BMP) receptors form active tetrameric complexes, comprised of two type I and two type II receptors, which then transmit signals to SMAD proteins. The link between receptor tetramerization and the mechanism of kinase activation, however, has not been elucidated. Here, using hydrogen deuterium exchange mass spectrometry (HDX-MS), small angle X-ray scattering (SAXS) and molecular dynamics (MD) simulations, combined with analysis of SMAD signaling, we show that the kinase domain of the type I receptor ALK2 and type II receptor BMPR2 form a heterodimeric complex via their C-terminal lobes. Formation of this dimer is essential for ligand-induced receptor signaling and is targeted by mutations in BMPR2 in patients with pulmonary arterial hypertension (PAH). We further show that the type I/type II kinase domain heterodimer serves as the scaffold for assembly of the active tetrameric receptor complexes to enable phosphorylation of the GS domain and activation of SMADs.


Subject(s)
Activin Receptors, Type I/chemistry , Activin Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type II/chemistry , Bone Morphogenetic Protein Receptors, Type II/metabolism , Signal Transduction/physiology , Activin Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Proteins/metabolism , Familial Primary Pulmonary Hypertension/metabolism , Humans , Ligands , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Domains , Pulmonary Arterial Hypertension , Scattering, Small Angle , Signal Transduction/genetics , Smad Proteins/metabolism , X-Ray Diffraction
2.
Dis Model Mech ; 13(9)2020 09 21.
Article in English | MEDLINE | ID: mdl-32988985

ABSTRACT

Heterotopic ossification (HO) is a disorder characterised by the formation of ectopic bone in soft tissue. Acquired HO typically occurs in response to trauma and is relatively common, yet its aetiology remains poorly understood. Genetic forms, by contrast, are very rare, but provide insights into the mechanisms of HO pathobiology. Fibrodysplasia ossificans progressiva (FOP) is the most debilitating form of HO. All patients reported to date carry heterozygous gain-of-function mutations in the gene encoding activin A receptor type I (ACVR1). These mutations cause dysregulated bone morphogenetic protein (BMP) signalling, leading to HO at extraskeletal sites including, but not limited to, muscles, ligaments, tendons and fascia. Ever since the identification of the causative gene, developing a cure for FOP has been a focus of investigation, and studies have decoded the pathophysiology at the molecular and cellular levels, and explored novel management strategies. Based on the established role of BMP signalling throughout HO in FOP, therapeutic modalities that target multiple levels of the signalling cascade have been designed, and some drugs have entered clinical trials, holding out hope of a cure. A potential role of other signalling pathways that could influence the dysregulated BMP signalling and present alternative therapeutic targets remains a matter of debate. Here, we review the recent FOP literature, including pathophysiology, clinical aspects, animal models and current management strategies. We also consider how this research can inform our understanding of other types of HO and highlight some of the remaining knowledge gaps.


Subject(s)
Myositis Ossificans/pathology , Translational Research, Biomedical , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/genetics , Animals , Bone Morphogenetic Proteins/metabolism , Disease Models, Animal , Humans , Mutation/genetics , Myositis Ossificans/diagnosis , Myositis Ossificans/genetics , Myositis Ossificans/physiopathology
3.
Cancer Cell ; 37(3): 308-323.e12, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32142668

ABSTRACT

Diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors for which there is currently no effective treatment. Some of these tumors combine gain-of-function mutations in ACVR1, PIK3CA, and histone H3-encoding genes. The oncogenic mechanisms of action of ACVR1 mutations are currently unknown. Using mouse models, we demonstrate that Acvr1G328V arrests the differentiation of oligodendroglial lineage cells, and cooperates with Hist1h3bK27M and Pik3caH1047R to generate high-grade diffuse gliomas. Mechanistically, Acvr1G328V upregulates transcription factors which control differentiation and DIPG cell fitness. Furthermore, we characterize E6201 as a dual inhibitor of ACVR1 and MEK1/2, and demonstrate its efficacy toward tumor cells in vivo. Collectively, our results describe an oncogenic mechanism of action for ACVR1 mutations, and suggest therapeutic strategies for DIPGs.


Subject(s)
Activin Receptors, Type I/chemistry , Activin Receptors, Type I/genetics , Brain Neoplasms/pathology , Glioma/pathology , Mutation , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Female , Glioma/drug therapy , Glioma/genetics , Histones/genetics , Histones/metabolism , Humans , Lactones/pharmacology , Male , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neuroglia/metabolism , Neuroglia/pathology , Oligodendroglia/pathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
4.
Sci Rep ; 9(1): 6896, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31053759

ABSTRACT

Neuronal activity is closely influenced by glia, especially microglia which are the resident immune cells in the central nervous system (CNS). Microglia in medicinal leech are the only cells able to migrate to the injury site within the 24 hours post-lesion. The microglia-neuron interactions constitute an important mechanism as there is neither astrocyte nor oligodendrocyte in the leech CNS. Given that axonal sprouting is impaired when microglia recruitment is inhibited, the crosstalk between microglia and neurons plays a crucial role in neuroprotection. The present results show that neurons and microglia both use ALK4/5 (a type of TGF-ß receptor) signaling in order to maintain mutual exchanges in an adult brain following an axonal injury. Indeed, a TGF-ß family member (nGDF) is immediately released by injured axons contributing to the early recruitment of ALK4/5+ microglia to the lesion site. Surprisingly, within the following hours, nGDF from microglia activates ALK4/5+ neurons to maintain a later microglia accumulation in lesion. Taken together, the results demonstrate that ALK4/5 signaling is essential throughout the response to the lesion in the leech CNS and gives a new insight in the understanding of this pathway. This latter is an important signal contributing to a correct sequential mobilization over time of microglia recruitment leading to axon regeneration.


Subject(s)
Activin Receptors, Type I/metabolism , Axons/pathology , Microglia/pathology , Neurons/pathology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Activin Receptors, Type I/chemistry , Amino Acid Sequence , Animals , Chemotaxis , Mice , Receptor, Transforming Growth Factor-beta Type I/chemistry
5.
Bioorg Med Chem Lett ; 28(20): 3356-3362, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30227946

ABSTRACT

The pyrazolo[1,5-a]pyrimidine LDN-193189 is a potent inhibitor of activin receptor-like kinase 2 (ALK2) but is nonselective for highly homologous ALK3 and shows only modest kinome selectivity. Herein, we describe the discovery of a novel series of potent and selective ALK2 inhibitors by replacing the quinolinyl with a 4-(sulfamoyl)naphthyl, yielding ALK2 inhibitors that exhibit not only excellent discrimination versus ALK3 but also high kinome selectivity. In addition, the optimized compound 23 demonstrates good ADME and in vivo pharmacokinetic properties.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Activin Receptors, Type I/chemistry , Animals , Binding Sites , Drug Discovery , Humans , Mice, Inbred C57BL , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics
6.
J Med Chem ; 61(16): 7261-7272, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30085668

ABSTRACT

Structure-activity relationship and crystallographic data revealed that quinazolinone-containing fragments flip between two distinct modes of binding to activin receptor-like kinase-2 (ALK2). We explored both binding modes to discover potent inhibitors and characterized the chemical modifications that triggered the flip in binding mode. We report kinase selectivity and demonstrate that compounds of this series modulate ALK2 in cancer cells. These inhibitors are attractive starting points for the discovery of more advanced ALK2 inhibitors.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Quinazolinones/chemistry , Structure-Activity Relationship , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/metabolism , Cell Line , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans
7.
J Mol Model ; 24(9): 262, 2018 Aug 29.
Article in English | MEDLINE | ID: mdl-30159679

ABSTRACT

Bone morphogenetic proteins (BMPs) are a family of more than 30 ligands and several receptors, such as activin like kinases (ALKs) and bone morphogenetic protein receptor (BMPR). Physiological significance of these proteins lies in their prominent role during homeostasis, apoptosis, tissue remodeling, embryonic patterning, and normal development. Fibrodysplasia ossificans progressive (FOP) is one among several other diseases caused by impaired BMP signaling. FOP is caused by the pathogenicity of activating mutation of ALK2. In order to treat FOP, a search for good inhibitors of ALK2 based on dorsomorphin and LDN substitution, which in essence is a ligand based search of inhibitors, is in progress. Contributing to this area of research we identified several lead molecules based on protein structure using virtual screening. After virtual screening of a huge library of small molecules and ab initio calculation of selected molecules for drug efficacy, we did molecular dynamic simulation of lead molecules and protein complexes. We identified five potential drug molecules that show very stable binding on the same binding site as LDN-213844. We also ranked these lead molecules based on MM/PBSA binding energy. This study provides a basis to think beyond the pyrimidine nucleus of dorsomorphin/LDN and design new chemical derivatives for effective treatment of FOP. Graphical abstract Small molecule inhibitors of ALK2.


Subject(s)
Activin Receptors, Type I , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/chemistry , Binding Sites , Drug Evaluation, Preclinical , Humans
9.
J Clin Endocrinol Metab ; 100(11): E1415-27, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26305619

ABSTRACT

CONTEXT: Activin A increases matrix metalloproteinase (MMP) 2 expression and cell invasion in human trophoblasts, but whether the expression of MMP2 is essential for the proinvasive effect of activin A has yet to be determined. Moreover, the identity of the activin receptor-like kinase (ALK; TGF-ß type I receptors) and downstream transcription factors (eg, SNAIL and SLUG) mediating the effects of activin on MMP2 expression and trophoblast cell invasion remains unknown. OBJECTIVE: To elucidate the role of MMP2 in activin A-induced human trophoblast cell invasion as well as the involvement of ALK4 and SNAIL. DESIGN: HTR8/SVneo immortalized human extravillous cytotrophoblast (EVT) cells and primary cultures of human first-trimester EVT cells were used as study models. Small interfering RNA (siRNA)-mediated knockdown approaches were used to investigate the molecular determinants of activin A-mediated functions. MAIN OUTCOME MEASURES: Levels of mRNA and protein were examined by reverse transcription-quantitative real-time PCR and Western blot, respectively. Cell invasiveness was measured by Matrigel-coated transwell assays. RESULTS: Treatment of HTR8/SVneo cells with activin A increased the production of SNAIL, SLUG, and MMP2 without altering that of MMP9, TIMP1, TIMP2, TWIST, RUNX2, ZEB1, or ZEB2. Similarly, activin A up-regulated the mRNA and protein levels of SNAIL and MMP2 in primary EVT cells. Knockdown of SNAIL attenuated activin A-induced MMP2 up-regulation in HTR8/SVneo and primary EVT cells. In HTR8/SVneo cells, activin A-induced production of SNAIL and MMP2 was abolished by pretreatment with the TGF-ß type I receptor (ALK4/5/7) inhibitor SB431542 or siRNA targeting ALK4, SMAD2/3, or common SMAD4. Likewise, knockdown of ALK4 or SMAD4 abolished the stimulatory effects of activin A on SNAIL and MMP2 expression in primary EVT cells. Importantly, activin A-induced HTR8/SVneo and primary EVT cell invasion were attenuated by siRNA-mediated depletion of ALK4 or MMP2. CONCLUSION: Activin A induces human trophoblast cell invasion by inducing SNAIL-mediated MMP2 expression through ALK4 in a SMAD2/3-SMAD4-dependent manner.


Subject(s)
Activin Receptors, Type I/metabolism , Activins/metabolism , Matrix Metalloproteinase 2/metabolism , Placentation , Transcription Factors/agonists , Trophoblasts/metabolism , Up-Regulation , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/genetics , Activins/antagonists & inhibitors , Adult , Cell Line, Transformed , Cell Movement , Cells, Cultured , Female , Humans , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/genetics , Pregnancy , Pregnancy Trimester, First , RNA Interference , RNA, Messenger/metabolism , Signal Transduction , Smad Proteins/agonists , Smad Proteins/antagonists & inhibitors , Smad Proteins/genetics , Smad Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/cytology
10.
PLoS One ; 10(7): e0132221, 2015.
Article in English | MEDLINE | ID: mdl-26133550

ABSTRACT

Abnormal alteration of bone morphogenetic protein (BMP) signaling is implicated in many types of diseases including cancer and heterotopic ossifications. Hence, small molecules targeting BMP type I receptors (BMPRI) to interrupt BMP signaling are believed to be an effective approach to treat these diseases. However, lack of understanding of the molecular determinants responsible for the binding selectivity of current BMP inhibitors has been a big hindrance to the development of BMP inhibitors for clinical use. To address this issue, we carried out in silico experiments to test whether computational methods can reproduce and explain the high selectivity of a small molecule BMP inhibitor DMH1 on BMPRI kinase ALK2 vs. the closely related TGF-ß type I receptor kinase ALK5 and vascular endothelial growth factor receptor type 2 (VEGFR2) tyrosine kinase. We found that, while the rigid docking method used here gave nearly identical binding affinity scores among the three kinases; free energy perturbation coupled with Hamiltonian replica-exchange molecular dynamics (FEP/H-REMD) simulations reproduced the absolute binding free energies in excellent agreement with experimental data. Furthermore, the binding poses identified by FEP/H-REMD led to a quantitative analysis of physical/chemical determinants governing DMH1 selectivity. The current work illustrates that small changes in the binding site residue type (e.g. pre-hinge region in ALK2 vs. ALK5) or side chain orientation (e.g. Tyr219 in caALK2 vs. wtALK2), as well as a subtle structural modification on the ligand (e.g. DMH1 vs. LDN193189) will cause distinct binding profiles and selectivity among BMP inhibitors. Therefore, the current computational approach represents a new way of investigating BMP inhibitors. Our results provide critical information for designing exclusively selective BMP inhibitors for the development of effective pharmacotherapy for diseases caused by aberrant BMP signaling.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Molecular Dynamics Simulation , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/genetics , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Substitution , Binding Sites/drug effects , Bone Morphogenetic Protein Receptors, Type I/chemistry , Crystallography, X-Ray , Drug Design , Humans , Ligands , Molecular Docking Simulation , Molecular Structure , Point Mutation , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Pyrazoles/chemistry , Pyrimidines/chemistry , Quinolines/chemistry , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/chemistry , Substrate Specificity , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/chemistry
11.
J Pept Sci ; 21(4): 283-93, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25588905

ABSTRACT

Nodal, a member of the TGF-ß superfamily, is a potent embryonic morphogen also implicated in tumor progression. As for other TGF-ßs, it triggers the signaling functions through the interaction with the extracellular domains of type I and type II serine/threonine kinase receptors and with the co-receptor Cripto. Recently, we reported the molecular models of Nodal in complex with its type I receptors (ALK4 and ALK7) as well as with Cripto, as obtained by homology modeling and docking simulations. From such models, potential binding epitopes have been identified. To validate such hypotheses, a series of mutated Nodal fragments have been synthesized. These peptide analogs encompass residues 44-67 of the Nodal protein, corresponding to the pre-helix loop and the H3 helix, and reproduce the wild-type sequence or bear some modifications to evaluate the hot-spot role of modified residues in the receptor binding. Here, we show the structural characterization in solution by CD and NMR of the Nodal peptides and the measurement of binding affinity toward Cripto by surface plasmon resonance. Data collected by both conformational analyses and binding measurements suggest a role for Y58 of Nodal in the recognition with Cripto and confirm that previously reported for E49 and E50. Surface plasmon resonance binding assays with recombinant proteins show that Nodal interacts in vitro also with ALK7 and ALK4 and preliminary data, generated using the Nodal synthetic fragments, suggest that Y58 of Nodal may also be involved in the recognition with these protein partners.


Subject(s)
Activin Receptors, Type I/chemistry , GPI-Linked Proteins/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Neoplasm Proteins/chemistry , Nodal Protein/chemistry , Peptides/chemistry , Peptides/metabolism , Activin Receptors, Type I/metabolism , Circular Dichroism , GPI-Linked Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Magnetic Resonance Imaging , Molecular Docking Simulation , Neoplasm Proteins/metabolism , Nodal Protein/metabolism , Protein Binding , Surface Plasmon Resonance
12.
Cell Signal ; 26(9): 1935-42, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24863882

ABSTRACT

Placentation is critical for establishing a healthy pregnancy. Trophoblasts mediate implantation and placentation and certain subtypes, most notably extravillous cytotrophoblast, are highly invasive. Trophoblast invasion is tightly regulated by microenvironmental cues that dictate placental morphology and depth. In choriocarcinomas, malignant trophoblast cells become hyperinvasive, breaching the myometrium and leading to major complications. Nodal, a member of the TGF-ß superfamily, is expressed throughout the endometrium during the peri-implantation period and in invasive trophoblast cells. Nodal promotes the invasion of numerous types of cancer cells. However, Nodal's role in trophoblast and choriocarcinoma cell invasion is unclear. Here we show that Nodal stimulates the invasion of both the non-malignant HTR-8SV/neo trophoblast and JAR choriocarcinoma cells in a dose-dependent manner. We found that endogenous ß-arrestins and Ral GTPases, key regulators of the cell cytoskeleton, are constitutively associated with Nodal receptors (ALK4 and ALK7) in trophoblast cells and that RalA is colocalized with ALK4 in endocytic vesicles. Nodal stimulates endogenous ß-arrestin2 to associate with phospho-ERK1/2, and knockdown of ß-arrestin or Ral proteins impairs Nodal-induced trophoblast and choriocarcinoma cell invasion. These results demonstrate, for the first time, that ß-arrestins and RalGTPases are important regulators of Nodal-induced invasion.


Subject(s)
Arrestins/metabolism , Nodal Protein/metabolism , Signal Transduction , ral GTP-Binding Proteins/metabolism , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/metabolism , Arrestins/antagonists & inhibitors , Arrestins/genetics , Cell Line , Cell Movement/drug effects , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nodal Protein/antagonists & inhibitors , Nodal Protein/genetics , Phosphorylation , Protein Binding , RNA Interference , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Transferrin/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , beta-Arrestins , ral GTP-Binding Proteins/antagonists & inhibitors , ral GTP-Binding Proteins/genetics
13.
Hum Mol Genet ; 23(20): 5364-77, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24852373

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a disabling genetic disorder of progressive heterotopic ossification (HO). Here, we report a patient with an ultra-rare point mutation [c.619C>G, p.Q207E] located in a codon adjacent to the most common FOP mutation [c.617G>A, p.R206H] of Activin A Receptor, type 1 (ACVR1) and that affects the same intracellular amino acid position in the GS activation domain as the engineered constitutively active (c.a.) variant p.Q207D. It was predicted that both mutations at residue 207 have similar functional effects by introducing a negative charge. Transgenic p.Q207D-c.a. mice have served as a model for FOP HO in several in vivo studies. However, we found that the engineered ACVR1(Q207D-c.a.) is significantly more active than the classic FOP mutation ACVR1(R206H) when overexpressed in chicken limbs and in differentiation assays of chondrogenesis, osteogenesis and myogenesis. Importantly, our studies reveal that the ACVR1(Q207E) resembles the classic FOP receptor in these assays, not the engineered ACVR1(Q207D-c.a.). Notably, reporter gene assays revealed that both naturally occurring FOP receptors (ACVR1(R206H) and ACVR1(Q207E)) were activated by BMP7 and were sensitive to deletion of the ligand binding domain, whereas the engineered ACVR1(Q207D-c.a.) exhibited ligand independent activity. We performed an in silico analysis and propose a structural model for p.Q207D-c.a. that irreversibly relocates the GS domain into an activating position, where it becomes ligand independent. We conclude that the engineered p.Q207D-c.a. mutation has severe limitations as a model for FOP, whereas the naturally occurring mutations p.R206H and p.Q207E facilitate receptor activation, albeit in a reversible manner.


Subject(s)
Activin Receptors, Type I/chemistry , Activin Receptors, Type I/genetics , Muscle, Skeletal/pathology , Myositis Ossificans/genetics , Myositis Ossificans/pathology , Point Mutation , Amino Acid Sequence , Animals , Chickens , Child , Disease Models, Animal , Genetic Variation , Glutamic Acid/metabolism , Glutamine/metabolism , Hindlimb/metabolism , Humans , Male , Mice , Mice, Transgenic , Molecular Sequence Data , NIH 3T3 Cells , Polymorphism, Single Nucleotide , Sequence Alignment
14.
Cell Stem Cell ; 13(6): 706-19, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24139759

ABSTRACT

During embryogenesis, the Activin/Nodal pathway promotes the mesendodermal lineage and inhibits neural fate. The molecular mechanisms underlying this role of the Activin/Nodal pathway are not clear. In this study, we report a role for protein tyrosine phosphatase 1B (PTP1B) in Activin-mediated early fate decisions during ESC differentiation and show that PTP1B acts as an effector of the Activin pathway to specify mesendodermal or neural fate. We found that the Activin/ALK4 pathway directly recruits PTP1B and stimulates its release from the endoplasmic reticulum through ALK4-mediated cleavage. Subsequently, PTP1B suppresses p-ERK1/2 signaling to inhibit neural specification and promote mesendodermal commitment. These findings suggest that a noncanonical Activin signaling pathway functions in lineage specification of mouse and human embryonic stem cells.


Subject(s)
Activins/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Neurons/cytology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/metabolism , Amino Acid Sequence , Animals , Benzamides/pharmacology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Dioxoles/pharmacology , Embryonic Stem Cells/drug effects , Endoderm/cytology , Endoderm/drug effects , Endoderm/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism
15.
PLoS One ; 8(4): e62721, 2013.
Article in English | MEDLINE | ID: mdl-23646137

ABSTRACT

Growth factor signaling pathways are tightly regulated by phosphorylation and include many important kinase targets of interest for drug discovery. Small molecule inhibitors of the bone morphogenetic protein (BMP) receptor kinase ALK2 (ACVR1) are needed urgently to treat the progressively debilitating musculoskeletal disease fibrodysplasia ossificans progressiva (FOP). Dorsomorphin analogues, first identified in zebrafish, remain the only BMP inhibitor chemotype reported to date. By screening an assay panel of 250 recombinant human kinases we identified a highly selective 2-aminopyridine-based inhibitor K02288 with in vitro activity against ALK2 at low nanomolar concentrations similar to the current lead compound LDN-193189. K02288 specifically inhibited the BMP-induced Smad pathway without affecting TGF-ß signaling and induced dorsalization of zebrafish embryos. Comparison of the crystal structures of ALK2 with K02288 and LDN-193189 revealed additional contacts in the K02288 complex affording improved shape complementarity and identified the exposed phenol group for further optimization of pharmacokinetics. The discovery of a new chemical series provides an independent pharmacological tool to investigate BMP signaling and offers multiple opportunities for pre-clinical development.


Subject(s)
Aminopyridines/pharmacology , Bone Morphogenetic Proteins/antagonists & inhibitors , Phenols/pharmacology , Signal Transduction/drug effects , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/chemistry , Aminopyridines/chemistry , Animals , Body Patterning/drug effects , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/metabolism , Humans , Models, Molecular , Molecular Conformation , Neovascularization, Physiologic/drug effects , Phenols/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Smad Proteins/metabolism , Zebrafish
16.
J Biol Chem ; 287(44): 36990-8, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22977237

ABSTRACT

Bone morphogenetic protein (BMP) receptor kinases are tightly regulated to control development and tissue homeostasis. Mutant receptor kinase domains escape regulation leading to severely degenerative diseases and represent an important therapeutic target. Fibrodysplasia ossificans progressiva (FOP) is a rare but devastating disorder of extraskeletal bone formation. FOP-associated mutations in the BMP receptor ALK2 reduce binding of the inhibitor FKBP12 and promote leaky signaling in the absence of ligand. To establish structural mechanisms of receptor regulation and to address the effects of FOP mutation, we determined the crystal structure of the cytoplasmic domain of ALK2 in complex with the inhibitors FKBP12 and dorsomorphin. FOP mutations break critical interactions that stabilize the inactive state of the kinase, thereby facilitating structural rearrangements that diminish FKBP12 binding and promote the correct positioning of the glycine-serine-rich loop and αC helix for kinase activation. The balance of these effects accounts for the comparable activity of R206H and L196P. Kinase activation in the clinically benign mutant L196P is far weaker than R206H but yields equivalent signals due to the stronger interaction of FKBP12 with R206H. The presented ALK2 structure offers a valuable template for the further design of specific inhibitors of BMP signaling.


Subject(s)
Activin Receptors, Type I/chemistry , Myositis Ossificans/enzymology , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Amino Acid Motifs , Animals , Bone Morphogenetic Protein 4/physiology , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Gene Expression Regulation , Genes, Reporter , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Mice , Models, Molecular , Mutation, Missense , Myositis Ossificans/genetics , Protein Binding , Pyrazoles/chemistry , Pyrimidines/chemistry , Signal Transduction , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/antagonists & inhibitors , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Protein 1A/metabolism
17.
Blood ; 119(25): 6162-71, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22566602

ABSTRACT

ALK1 is a type I receptor of the TGF-ß family that is involved in angiogenesis. Circulating BMP9 was identified as a specific ligand for ALK1 inducing vascular quiescence. In this work, we found that blocking BMP9 with a neutralizing antibody in newborn mice significantly increased retinal vascular density. Surprisingly, Bmp9-KO mice did not show any defect in retinal vascularization. However, injection of the extracellular domain of ALK1 impaired retinal vascularization in Bmp9-KO mice, implicating another ligand for ALK1. Interestingly, we detected a high level of circulating BMP10 in WT and Bmp9-KO pups. Further, we found that injection of a neutralizing anti-BMP10 antibody to Bmp9-KO pups reduced retinal vascular expansion and increased vascular density, whereas injection of this antibody to WT pups did not affect the retinal vasculature. These data suggested that BMP9 and BMP10 are important in postnatal vascular remodeling of the retina and that BMP10 can substitute for BMP9. In vitro stimulation of endothelial cells by BMP9 and BMP10 increased the expression of genes involved in the Notch signaling pathway (Jagged1, Dll4, Hey1, Hey2, Hes1) and decreased apelin expression, suggesting a possible cross-talk between these pathways and the BMP pathway.


Subject(s)
Bone Morphogenetic Proteins/physiology , Growth Differentiation Factor 2/physiology , Retinal Vessels/physiology , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/pharmacology , Activin Receptors, Type II , Animals , Animals, Newborn , Antibodies/pharmacology , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Count , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Growth Differentiation Factor 2/antagonists & inhibitors , Growth Differentiation Factor 2/genetics , Growth Differentiation Factor 2/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Structure, Tertiary , Recombinant Proteins/pharmacology , Retinal Vessels/cytology , Retinal Vessels/drug effects , Retinal Vessels/metabolism
18.
J Mol Model ; 18(8): 3617-25, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22354277

ABSTRACT

Transforming growth factor-beta (TGF-ß) proteins are a family of structurally related extracellular proteins that trigger their signaling functions through interaction with the extracellular domains of their cognate serine/threonine kinase receptors. The specificity of TGF-ß/receptor binding is complex and gives rise to multiple functional roles. Additionally, it is not completely understood at the atomic level. Here, we use the most reliable computational methods currently available to study systems involving activin-like kinase (ALK) receptors ALK4 and ALK7 and their multiple TGF-ß ligands. We built models for all these proteins and their complexes for which experimental structures are not available. By analyzing the surfaces of interaction in six different TGF-ß/ALK complexes we could infer which are the structural distinctive features of the ligand-receptor binding mode. Furthermore, this study allowed us to rationalize why binding of the growth factors GDF3 and Nodal to the ALK4 receptor requires the Cripto co-factor, whilst binding to the ALK7 receptor does not.


Subject(s)
Activin Receptors, Type I/chemistry , Computer Simulation , Growth Differentiation Factors/chemistry , Models, Molecular , Nodal Protein/chemistry , Binding Sites , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , TGF-beta Superfamily Proteins/chemistry
19.
Cell Signal ; 24(2): 476-483, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21983015

ABSTRACT

Transforming growth factor-ß (TGFß) receptor kinase inhibitors have a great therapeutic potential. SB431542 is one of the mainly used kinase inhibitors of the TGFß/Activin pathway receptors, but needs improvement of its EC(50) (EC(50)=1 µM) to be translated to clinical use. A key feature of SB431542 is that it specifically targets receptors from the TGFß/Activin pathway but not the closely related receptors from the bone morphogenic proteins (BMP) pathway. To understand the mechanisms of this selectivity, we solved the crystal structure of the TGFß type I receptor (TßRI) kinase domain in complex with SB431542. We mutated TßRI residues coordinating SB431542 to their counterparts in activin-receptor like kinase 2 (ALK2), a BMP receptor kinase, and tested the kinase activity of mutated TßRI. We discovered that a Ser280Thr mutation yielded a TßRI variant that was resistant to SB431542 inhibition. Furthermore, the corresponding Thr283Ser mutation in ALK2 yielded a BMP receptor sensitive to SB431542. This demonstrated that Ser280 is the key determinant of selectivity for SB431542. This work provides a framework for optimising the SB431542 scaffold to more potent and selective inhibitors of the TGFß/Activin pathway.


Subject(s)
Activin Receptors, Type I/metabolism , Benzamides/pharmacology , Bone Morphogenetic Proteins/metabolism , Dioxoles/pharmacology , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Serine/metabolism , Signal Transduction , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/genetics , Activins/metabolism , Benzamides/chemistry , Benzamides/metabolism , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/genetics , Crystallography, X-Ray , Dioxoles/chemistry , Dioxoles/metabolism , Drug Design , HEK293 Cells , Humans , Inhibitory Concentration 50 , Models, Molecular , Mutation , Phosphorylation , Plasmids , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/genetics , Serine/genetics , Substrate Specificity , Transfection , Transforming Growth Factor beta/metabolism
20.
Eur J Hum Genet ; 19(4): 389-93, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21248739

ABSTRACT

Down's syndrome (DS), resulting from an additional copy of chromosome 21 (trisomy 21), is frequently associated with congenital heart defects (CHDs). Although the increased dosage of chromosome 21 sequences is likely to be part of the etiology of cardiac defects, only a proportion of DS patients exhibit a congenital heart defect (birth prevalence 40-60%). Through a large-candidate gene-sequencing screen in patients with atrioventricular septal defects, substitutions were identified in bone morphogenetic protein (BMP) type I receptor ALK2 and two other genes in a patient with DS and a primum-type atrial septal defect. Structural modeling of the cytoplasmic domain of the ALK2 receptor suggests that H286 is in close proximity to the nucleotide-binding site of the kinase domain. We investigated whether this p.His286Asp substitution altered ALK2 function by using both in vitro as well as in vivo assays. The p.His286Asp variant demonstrated impaired functional activity as measured by BMP-specific transcriptional response assays. Furthermore, mild dominant-interfering activity was observed in vivo compared with wild-type ALK2 as determined by RNA injection into zebrafish embryos. These data indicate that in the context of a DS background, ALK2-mediated reduction of BMP signaling may contribute to CHDs.


Subject(s)
Activin Receptors, Type I/genetics , Down Syndrome/genetics , Heart Defects, Congenital/etiology , Heart Defects, Congenital/genetics , Mutation/genetics , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Protein 1/metabolism , Cattle , Down Syndrome/complications , Female , Heart Defects, Congenital/diagnosis , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Atrial/pathology , Humans , Male , Protein Conformation , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...