Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 7803, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35551212

ABSTRACT

Sotatercept is an activin receptor type IIA-Fc (ActRIIA-Fc) fusion protein that improves cardiopulmonary function in patients with pulmonary arterial hypertension (PAH) by selectively trapping activins and growth differentiation factors. However, the cellular and molecular mechanisms of ActRIIA-Fc action are incompletely understood. Here, we determined through genome-wide expression profiling that inflammatory and immune responses are prominently upregulated in the lungs of a Sugen-hypoxia rat model of severe angio-obliterative PAH, concordant with profiles observed in PAH patients. Therapeutic treatment with ActRIIA-Fc-but not with a vasodilator-strikingly reversed proinflammatory and proliferative gene expression profiles and normalized macrophage infiltration in diseased rodent lungs. Furthermore, ActRIIA-Fc normalized pulmonary macrophage infiltration and corrected cardiopulmonary structure and function in Bmpr2 haploinsufficient mice subjected to hypoxia, a model of heritable PAH. Three high-affinity ligands of ActRIIA-Fc each induced macrophage activation in vitro, and their combined immunoneutralization in PAH rats produced cardiopulmonary benefits comparable to those elicited by ActRIIA-Fc. Our results in complementary experimental and genetic models of PAH reveal therapeutic anti-inflammatory activities of ActRIIA-Fc that, together with its known anti-proliferative effects on vascular cell types, could underlie clinical activity of sotatercept as either monotherapy or add-on to current PAH therapies.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Animals , Disease Models, Animal , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/drug therapy , Hypoxia/drug therapy , Inflammation/drug therapy , Mice , Pulmonary Arterial Hypertension/drug therapy , Rats , Recombinant Fusion Proteins
2.
Sci Rep ; 11(1): 18341, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526551

ABSTRACT

Ligands of the transforming growth factor-ß (TGF-ß) superfamily are important targets for therapeutic intervention but present challenges because they signal combinatorially and exhibit overlapping activities in vivo. To obtain agents capable of sequestering multiple TGF-ß superfamily ligands with novel selectivity, we generated soluble, heterodimeric ligand traps by pairing the extracellular domain (ECD) of the native activin receptor type IIB (ActRIIB) alternately with the ECDs of native type I receptors activin receptor-like kinase 4 (ALK4), ALK7, or ALK3. Systematic analysis of these heterodimeric constructs by surface plasmon resonance, and comparison with their homodimeric counterparts, revealed that each type I receptor partner confers a distinct ligand-binding profile to the heterodimeric construct. Additional characterization in cell-based reporter gene assays confirmed that the heterodimeric constructs possessed different profiles of signaling inhibition in vitro, which translated into altered patterns of pharmacological activity when constructs were administered systemically to wild-type mice. Our results detail a versatile platform for the modular recombination of naturally occurring receptor domains, giving rise to inhibitory ligand traps that could aid in defining the physiological roles of TGF-ß ligand sets or be directed therapeutically to human diseases arising from dysregulated TGF-ß superfamily signaling.


Subject(s)
Activin Receptors/metabolism , Drug Discovery/methods , Protein Engineering/methods , Activin Receptors/chemistry , Activin Receptors/genetics , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Humans , Ligands , Mice , Mice, Inbred C57BL , Protein Binding , Protein Multimerization , Transforming Growth Factor beta/metabolism
3.
J Clin Invest ; 131(4)2021 02 15.
Article in English | MEDLINE | ID: mdl-33586684

ABSTRACT

Patients with neuromuscular disorders suffer from a lack of treatment options for skeletal muscle weakness and disease comorbidities. Here, we introduce as a potential therapeutic agent a heterodimeric ligand-trapping fusion protein, ActRIIB:ALK4-Fc, which comprises extracellular domains of activin-like kinase 4 (ALK4) and activin receptor type IIB (ActRIIB), a naturally occurring pair of type I and II receptors belonging to the TGF-ß superfamily. By surface plasmon resonance (SPR), ActRIIB:ALK4-Fc exhibited a ligand binding profile distinctly different from that of its homodimeric variant ActRIIB-Fc, sequestering ActRIIB ligands known to inhibit muscle growth but not trapping the vascular regulatory ligand bone morphogenetic protein 9 (BMP9). ActRIIB:ALK4-Fc and ActRIIB-Fc administered to mice exerted differential effects - concordant with SPR results - on vessel outgrowth in a retinal explant assay. ActRIIB:ALK4-Fc induced a systemic increase in muscle mass and function in wild-type mice and in murine models of Duchenne muscular dystrophy (DMD), amyotrophic lateral sclerosis (ALS), and disuse atrophy. Importantly, ActRIIB:ALK4-Fc improved neuromuscular junction abnormalities in murine models of DMD and presymptomatic ALS and alleviated acute muscle fibrosis in a DMD model. Furthermore, in combination therapy ActRIIB:ALK4-Fc increased the efficacy of antisense oligonucleotide M12-PMO on dystrophin expression and skeletal muscle endurance in an aged DMD model. ActRIIB:ALK4-Fc shows promise as a therapeutic agent, alone or in combination with dystrophin rescue therapy, to alleviate muscle weakness and comorbidities of neuromuscular disorders.


Subject(s)
Activin Receptors, Type II/pharmacology , Activin Receptors, Type I/pharmacology , Amyotrophic Lateral Sclerosis/drug therapy , Immunoglobulin Fc Fragments/pharmacology , Muscle, Skeletal/metabolism , Muscular Disorders, Atrophic/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Recombinant Fusion Proteins/pharmacology , Activin Receptors, Type I/genetics , Activin Receptors, Type II/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , CHO Cells , Cricetulus , Disease Models, Animal , Humans , Immunoglobulin Fc Fragments/genetics , Male , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/metabolism , Muscular Disorders, Atrophic/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Recombinant Fusion Proteins/genetics
4.
Sci Transl Med ; 12(543)2020 05 13.
Article in English | MEDLINE | ID: mdl-32404506

ABSTRACT

Human genetics, biomarker, and animal studies implicate loss of function in bone morphogenetic protein (BMP) signaling and maladaptive transforming growth factor-ß (TGFß) signaling as drivers of pulmonary arterial hypertension (PAH). Although sharing common receptors and effectors with BMP/TGFß, the function of activin and growth and differentiation factor (GDF) ligands in PAH are less well defined. Increased expression of GDF8, GDF11, and activin A was detected in lung lesions from humans with PAH and experimental rodent models of pulmonary hypertension (PH). ACTRIIA-Fc, a potent GDF8/11 and activin ligand trap, was used to test the roles of these ligands in animal and cellular models of PH. By blocking GDF8/11- and activin-mediated SMAD2/3 activation in vascular cells, ACTRIIA-Fc attenuated proliferation of pulmonary arterial smooth muscle cells and pulmonary microvascular endothelial cells. In several experimental models of PH, prophylactic administration of ACTRIIA-Fc markedly improved hemodynamics, right ventricular (RV) hypertrophy, RV function, and arteriolar remodeling. When administered after the establishment of hemodynamically severe PH in a vasculoproliferative model, ACTRIIA-Fc was more effective than vasodilator in attenuating PH and arteriolar remodeling. Potent antiremodeling effects of ACTRIIA-Fc were associated with inhibition of SMAD2/3 activation and downstream transcriptional activity, inhibition of proliferation, and enhancement of apoptosis in the vascular wall. ACTRIIA-Fc reveals an unexpectedly prominent role of GDF8, GDF11, and activin as drivers of pulmonary vascular disease and represents a therapeutic strategy for restoring the balance between SMAD1/5/9 and SMAD2/3 signaling in PAH.


Subject(s)
Hypertension, Pulmonary , Activins , Animals , Cell Differentiation , Endothelial Cells , Hypertension, Pulmonary/drug therapy , Signal Transduction
5.
J Cell Mol Med ; 24(11): 6162-6177, 2020 06.
Article in English | MEDLINE | ID: mdl-32351032

ABSTRACT

In ß-thalassaemia, anaemia results from ineffective erythropoiesis characterized by inhibition of late-stage erythroid differentiation. We earlier used luspatercept and RAP-536 protein traps for certain Smad2/3-pathway ligands to implicate Smad2/3-pathway overactivation in dysregulated erythroid differentiation associated with murine ß-thalassaemia and myelodysplasia. Importantly, luspatercept alleviates anaemia and has been shown to reduce transfusion burden in patients with ß-thalassaemia or myelodysplasia. Here, we investigated the molecular mechanisms underlying luspatercept action and pSmad2/3-mediated inhibition of erythroid differentiation. In murine erythroleukemic (MEL) cells in vitro, ligand-mediated overactivation of the Smad2/3 pathway reduced nuclear levels of GATA-1 (GATA-binding factor-1) and its transcriptional activator TIF1γ (transcription intermediary factor 1γ), increased levels of reactive oxygen species, reduced cell viability and haemoglobin levels, and inhibited erythroid differentiation. Co-treatment with luspatercept in MEL cells partially or completely restored each of these. In ß-thalassaemic mice, RAP-536 up-regulated Gata1 and its target gene signature in erythroid precursors determined by transcriptional profiling and gene set enrichment analysis, restored nuclear levels of GATA-1 in erythroid precursors, and nuclear distribution of TIF1γ in erythroblasts. Bone marrow cells from ß-thalassaemic mice treated with luspatercept also exhibited restored nuclear availability of GATA-1 ex vivo. Our results implicate GATA-1, and likely TIF1γ, as key mediators of luspatercept/RAP-536 action in alleviating ineffective erythropoiesis.


Subject(s)
Activin Receptors, Type II/pharmacology , Cell Differentiation , Erythroid Cells/pathology , GATA1 Transcription Factor/metabolism , Immunoglobulin Fc Fragments/pharmacology , Recombinant Fusion Proteins/pharmacology , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , beta-Thalassemia/pathology , Anemia/complications , Anemia/drug therapy , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Disease Models, Animal , Erythroblasts , Erythroid Cells/drug effects , Hemoglobins/metabolism , Leukemia, Erythroblastic, Acute/pathology , Ligands , Mice, Inbred C57BL , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , beta-Thalassemia/complications , beta-Thalassemia/genetics
6.
Genetics ; 214(3): 691-702, 2020 03.
Article in English | MEDLINE | ID: mdl-31879319

ABSTRACT

The azoxymethane model of colorectal cancer (CRC) was used to gain insights into the genetic heterogeneity of nonfamilial CRC. We observed significant differences in susceptibility parameters across 40 mouse inbred strains, with 6 new and 18 of 24 previously identified mouse CRC modifier alleles detected using genome-wide association analysis. Tumor incidence varied in F1 as well as intercrosses and backcrosses between resistant and susceptible strains. Analysis of inheritance patterns indicates that resistance to CRC development is inherited as a dominant characteristic genome-wide, and that susceptibility appears to occur in individuals lacking a large-effect, or sufficient numbers of small-effect, polygenic resistance alleles. Our results suggest a new polygenic model for inheritance of nonfamilial CRC, and that genetic studies in humans aimed at identifying individuals with elevated susceptibility should be pursued through the lens of absence of dominant resistance alleles rather than for the presence of susceptibility alleles.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Multifactorial Inheritance/genetics , Alleles , Animals , Azoxymethane/toxicity , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Disease Models, Animal , Drug Resistance, Neoplasm , Genetic Heterogeneity , Heredity , Humans , Mice , Mice, Inbred Strains/genetics , Models, Genetic
7.
J Pharmacol Exp Ther ; 368(3): 435-445, 2019 03.
Article in English | MEDLINE | ID: mdl-30563942

ABSTRACT

Follistatin is an endogenous glycoprotein that promotes growth and repair of skeletal muscle by sequestering inhibitory ligands of the transforming growth factor-ß superfamily and may therefore have therapeutic potential for neuromuscular diseases. Here, we sought to determine the suitability of a newly engineered follistatin fusion protein (FST288-Fc) to promote localized, rather than systemic, growth of skeletal muscle by capitalizing on the intrinsic heparin-binding ability of the follistatin-288 isoform. As determined by surface plasmon resonance and cell-based assays, FST288-Fc binds to activin A, activin B, myostatin (growth differentiation factor GDF8), and GDF11 with high affinity and neutralizes their activity in vitro. Intramuscular administration of FST288-Fc in mice induced robust, dose-dependent growth of the targeted muscle but not of surrounding or contralateral muscles, in contrast to the systemic effects of a locally administered fusion protein incorporating activin receptor type IIB (ActRIIB-Fc). Furthermore, systemic administration of FST288-Fc in mice did not alter muscle mass or body composition as determined by NMR, which again contrasts with the pronounced systemic activity of ActRIIB-Fc when administered by the same route. Subsequent analysis revealed that FST288-Fc in the circulation undergoes rapid proteolysis, thereby restricting its activity to individual muscles targeted by intramuscular administration. These results indicate that FST288-Fc can produce localized growth of skeletal muscle in a targeted manner with reduced potential for undesirable systemic effects. Thus, FST288-Fc and similar agents may be beneficial in the treatment of disorders with muscle atrophy that is focal, asymmetric, or otherwise heterogeneous.


Subject(s)
Follistatin/administration & dosage , Immunoglobulin G/administration & dosage , Muscle, Skeletal/drug effects , Muscle, Skeletal/growth & development , Recombinant Fusion Proteins/administration & dosage , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Follistatin/genetics , Follistatin/metabolism , Humans , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Injections, Intramuscular , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Protein Structure, Secondary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
8.
Muscle Nerve ; 58(5): 718-725, 2018 11.
Article in English | MEDLINE | ID: mdl-29981243

ABSTRACT

INTRODUCTION: Difficulty in modeling congenital contractures (deformities of muscle-tendon unit development that include shortened muscles and lengthened tendons) has limited research of new treatments. METHODS: Early immobilization of the ankle in prepuberal mice was used to produce deformities similar to congenital contractures. Stretch treatment, electrostimulation, and local intramuscular injection of a follistatin analog (FST-288) were assessed as therapeutic interventions for these deformities. RESULTS: Ankle immobilization at full plantarflexion and 90 ° created tendon lengthening and muscle shortening in the tibialis anterior and soleus. Stretch treatment produced minimal evidence for longitudinal muscle growth and electrostimulation provided no additional benefit. Stretch treatment with FST-288 produced greater longitudinal muscle growth and less tendon lengthening, constituting the best treatment response. DISCUSSION: Ankle immobilization recapitulates key morphologic features of congenital contracture, and these features can be mitigated by a combination of stretch and pharmacological approaches that may be useful in patients. Muscle Nerve 58: 718-725, 2018.


Subject(s)
Ankle Injuries/etiology , Ankle Injuries/pathology , Immobilization/adverse effects , Muscle, Skeletal/physiopathology , Outcome Assessment, Health Care/methods , Animals , Ankle Injuries/therapy , Biomechanical Phenomena , Disease Models, Animal , Electric Stimulation Therapy , Female , Follistatin/therapeutic use , Male , Mice , Muscle Contraction , Sarcomeres/pathology , Splints , Statistics, Nonparametric , Tendons , Time Factors
9.
J Bone Miner Res ; 33(10): 1760-1772, 2018 10.
Article in English | MEDLINE | ID: mdl-29813187

ABSTRACT

Osteogenesis imperfecta (OI) is a heritable connective tissue disorder primarily due to mutations in the type I collagen genes (COL1A1 and COL1A2), leading to compromised biomechanical integrity in type I collagen-containing tissues such as bone. Bone is inherently mechanosensitive and thus responds and adapts to external stimuli, such as muscle mass and contractile strength, to alter its mass and shape. Myostatin, a member of the TGF-ß superfamily, signals through activin receptor type IIB to negatively regulate muscle fiber growth. Because of the positive impact of myostatin deficiency on bone mass, we utilized a soluble activin receptor type IIB-mFc (sActRIIB-mFc) fusion protein in two molecularly distinct OI mouse models (G610C and oim) and evaluated their bone properties. Wild-type (WT), +/G610C, and oim/oim mice were treated from 2 to 4 months of age with either vehicle (Tris-buffered saline) or sActRIIB-mFc (10 mg/kg). Femurs of sActRIIB-mFc-treated mice exhibited increased trabecular bone volume regardless of genotype, whereas the cortical bone microarchitecture and biomechanical strength were only improved in WT and +/G610C mice. Dynamic histomorphometric analyses suggest the improved cortical bone geometry and biomechanical integrity reflect an anabolic effect due to increased mineral apposition and bone formation rates, whereas static histomorphometric analyses supported sActRIIB-mFc treatment also having an anti-catabolic impact with decreased osteoclast number per bone surface on trabecular bone regardless of sex and genotype. Together, our data suggest that sActRIIB-mFc may provide a new therapeutic direction to improve both bone and muscle properties in OI. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Activin Receptors, Type II/therapeutic use , Bone and Bones/pathology , Osteogenesis Imperfecta/drug therapy , Osteogenesis Imperfecta/pathology , Activin Receptors, Type II/pharmacology , Animals , Biomarkers/blood , Biomechanical Phenomena , Bone and Bones/physiopathology , Disease Models, Animal , Female , Femur/pathology , Male , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteocytes/metabolism , Osteogenesis Imperfecta/blood , Osteogenesis Imperfecta/physiopathology , Peptide Fragments/blood , Procollagen/blood , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Solubility , Torsion, Mechanical
10.
Muscle Nerve ; 57(2): 294-304, 2018 02.
Article in English | MEDLINE | ID: mdl-28555931

ABSTRACT

INTRODUCTION: Osteogenesis imperfecta (OI) is characterized by skeletal fragility and muscle weakness. In this study we investigated the effects of soluble activin type IIB receptor (sActRIIB-mFc) on muscle mass and function in 2 distinct mouse models of OI: osteogenesis imperfecta murine (oim) and +/G610C. METHODS: Wild-type (WT), +/G610C, and oim/oim mice were treated from 2 to 4 months of age with Tris-buffered saline (vehicle) or sActRIIB-mFc and their hindlimb muscles evaluated for mass, morphology, and contractile function. RESULTS: sActRIIB-mFc-treated WT, +/G610C, and oim/oim mice had increased hindlimb muscle weights and myofiber cross-sectional area compared with vehicle-treated counterparts. sActRIIB-mFc-treated oim/oim mice also exhibited increased contractile function relative to vehicle-treated counterparts. DISCUSSION: Blocking endogenous ActRIIB was effective at increasing muscle size in mouse models of OI, and increasing contractile function in oim/oim mice. ActRIIB inhibitors may provide a potential mutation-specific therapeutic option for compromised muscle function in OI. Muscle Nerve 57: 294-304, 2018.


Subject(s)
Activin Receptors, Type II/genetics , Muscle, Skeletal/physiopathology , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/physiopathology , Anatomy, Cross-Sectional , Animals , Citrate (si)-Synthase/metabolism , Collagen Type I/genetics , Female , Hindlimb/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Muscle Contraction , Muscle Fibers, Skeletal/pathology , Muscle Strength , Mutation , Organ Size , Osteogenesis Imperfecta/pathology
11.
Oncotarget ; 7(27): 41857-41869, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27248821

ABSTRACT

Treatment of metastatic renal cell carcinoma (mRCC) with agents that block signaling through vascular endothelial growth factor receptor 2 (VEGFR2) induces disease regression or stabilization in some patients; however, these responses tend to be short-lived. Therefore, development of combination therapies that can extend the efficacy of VEGFR antagonists in mRCC remains a priority.We studied murine xenograft models of RCC that become refractory to treatment with the VEGFR tyrosine kinase inhibitor (TKI) sunitinib. Dalantercept is a novel antagonist of Activin receptor-like kinase 1 (ALK1)/Bone morphogenetic protein (BMP) 9 signaling. Dalantercept inhibited growth in the murine A498 xenograft model which correlated with hyperdilation of the tumor vasculature and an increase in tumor hypoxia. When combined with sunitinib, dalantercept induced tumor necrosis and prevented tumor regrowth and revascularization typically seen with sunitinib monotherapy in two RCC models. Combination therapy led to significant downregulation of angiogenic genes as well as downregulation of endothelial specific gene expression particularly of the Notch signaling pathway. We demonstrate that simultaneous targeting of molecules that control distinct phases of angiogenesis, such as ALK1 and VEGFR, is a valid strategy for treatment of mRCC. At the molecular level, combination therapy leads to downregulation of Notch signaling.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Activin Receptors, Type II/administration & dosage , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Axitinib , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/administration & dosage , Immunoglobulin Fc Fragments/administration & dosage , Indazoles/administration & dosage , Indoles/administration & dosage , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Kinase Inhibitors/administration & dosage , Pyrroles/administration & dosage , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Fusion Proteins/administration & dosage , Sunitinib , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
12.
Am J Respir Crit Care Med ; 194(9): 1140-1151, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27115515

ABSTRACT

RATIONALE: Transforming growth factor-ß (TGF-ß) ligands signal via type I and type II serine-threonine kinase receptors to regulate broad transcriptional programs. Excessive TGF-ß-mediated signaling is implicated in the pathogenesis of pulmonary arterial hypertension, based in part on the ability of broad inhibition of activin-like kinase (ALK) receptors 4/5/7 recognizing TGF-ß, activin, growth and differentiation factor, and nodal ligands to attenuate experimental pulmonary hypertension (PH). These broad inhibition strategies do not delineate the specific contribution of TGF-ß versus a multitude of other ligands, and their translation is limited by cardiovascular and systemic toxicity. OBJECTIVES: We tested the impact of a soluble TGF-ß type II receptor extracellular domain expressed as an immunoglobulin-Fc fusion protein (TGFBRII-Fc), serving as a selective TGF-ß1/3 ligand trap, in several experimental PH models. METHODS: Signaling studies used cultured human pulmonary artery smooth muscle cells. PH was studied in monocrotaline-treated Sprague-Dawley rats, SU5416/hypoxia-treated Sprague-Dawley rats, and SU5416/hypoxia-treated C57BL/6 mice. PH, cardiac function, vascular remodeling, and valve structure were assessed by ultrasound, invasive hemodynamic measurements, and histomorphometry. MEASUREMENTS AND MAIN RESULTS: TGFBRII-Fc is an inhibitor of TGF-ß1 and TGF-ß3, but not TGF-ß2, signaling. In vivo treatment with TGFBRII-Fc attenuated Smad2 phosphorylation, normalized expression of plasminogen activator inhibitor-1, and mitigated PH and pulmonary vascular remodeling in monocrotaline-treated rats, SU5416/hypoxia-treated rats, and SU5416/hypoxia-treated mice. Administration of TGFBRII-Fc to monocrotaline-treated or SU5416/hypoxia-treated rats with established PH improved right ventricular systolic pressures, right ventricular function, and survival. No cardiac structural or valvular abnormalities were observed after treatment with TGFBRII-Fc. CONCLUSIONS: Our findings are consistent with a pathogenetic role of TGF-ß1/3, demonstrating the efficacy and tolerability of selective TGF-ß ligand blockade for improving hemodynamics, remodeling, and survival in multiple experimental PH models.


Subject(s)
Hypertension, Pulmonary/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Disease Models, Animal , Heart/physiopathology , Hemodynamics/physiology , Hypertension, Pulmonary/physiopathology , Immunoglobulin Fc Fragments/metabolism , Ligands , Male , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Vascular Remodeling/physiology
13.
Am J Pathol ; 186(6): 1568-81, 2016 06.
Article in English | MEDLINE | ID: mdl-27102768

ABSTRACT

Nemaline myopathies (NMs) are a group of congenital muscle diseases caused by mutations in at least 10 genes and associated with a range of clinical symptoms. NM is defined on muscle biopsy by the presence of cytoplasmic rod-like structures (nemaline rods) composed of cytoskeletal material. Myofiber smallness is also found in many cases of NM and may represent a cause of weakness that can be counteracted by treatment. We have used i.p. injection of activin type IIB receptor (ActRIIB)-mFc (an inhibitor of myostatin signaling) to promote hypertrophy and increase strength in our prior murine work; we therefore tested whether ActRIIB-mFc could improve weakness in NM mice through myofiber hypertrophy. We report a study of ActRIIB-mFc treatment in the Acta1 H40Y mouse model of NM. Treatment of Acta1 H40Y mice produced significant increases in body mass, muscle mass, quadriceps myofiber size, and survival, but other measurements of strength (forelimb grip strength, ex vivo measurements of contractile function) did not improve. Our studies also identified that the complications of urethral obstruction are associated with mortality in male hemizygote Acta1 H40Y mice. The incidence of urethral obstruction and histologic evidence of chronic obstruction (inflammation) were significantly lower in Acta1 H40Y mice that had been treated with ActRIIB-mFc. ActRIIB-mFc treatment produces a mild benefit to the disease phenotype in Acta1 H40Y mice.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Myofibrils/drug effects , Myopathies, Nemaline/pathology , Animals , Blotting, Western , Disease Models, Animal , Male , Mice , Mice, Mutant Strains , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Myofibrils/pathology
14.
Cancer Res ; 75(12): 2445-56, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26077471

ABSTRACT

Exploration of new strategies for the prevention of breast cancer metastasis is justifiably at the center of clinical attention. In this study, we combined a computational biology approach with mechanism-based preclinical trials to identify inhibitors of activin-like receptor kinase (ALK) 1 as effective agents for blocking angiogenesis and metastasis in breast cancer. Pharmacologic targeting of ALK1 provided long-term therapeutic benefit in mouse models of mammary carcinoma, accompanied by strikingly reduced metastatic colonization as a monotherapy or part of combinations with chemotherapy. Gene-expression analysis of breast cancer specimens from a population-based nested case-control study encompassing 768 subjects defined endothelial expression of ALK1 as an independent and highly specific prognostic factor for metastatic manifestation, a finding that was corroborated in an independent clinical cohort. Overall, our results suggest that pharmacologic inhibition of endothelial ALK1 constitutes a tractable strategy for interfering with metastatic dissemination of breast cancer.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Activin Receptors, Type II/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Endothelium/enzymology , Female , Humans , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Neoplasm Metastasis
15.
Blood ; 123(25): 3864-72, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24795345

ABSTRACT

In ß-thalassemia, unequal production of α- and ß-globin chains in erythroid precursors causes apoptosis and inhibition of late-stage erythroid differentiation, leading to anemia, ineffective erythropoiesis (IE), and dysregulated iron homeostasis. Here we used a murine model of ß-thalassemia intermedia (Hbb(th1/th1) mice) to investigate effects of a modified activin receptor type IIB (ActRIIB) ligand trap (RAP-536) that inhibits Smad2/3 signaling. In Hbb(th1/th1) mice, treatment with RAP-536 reduced overactivation of Smad2/3 in splenic erythroid precursors. In addition, treatment of Hbb(th1/th1) mice with RAP-536 reduced α-globin aggregates in peripheral red cells, decreased the elevated reactive oxygen species present in erythroid precursors and peripheral red cells, and alleviated anemia by promoting differentiation of late-stage erythroid precursors and reducing hemolysis. Notably, RAP-536 treatment mitigated disease complications of IE, including iron overload, splenomegaly, and bone pathology, while reducing erythropoietin levels, improving erythrocyte morphology, and extending erythrocyte life span. These results implicate signaling by the transforming growth factor-ß superfamily in late-stage erythropoiesis and reveal potential of a modified ActRIIB ligand trap as a novel therapeutic agent for thalassemia syndrome and other red cell disorders characterized by IE.


Subject(s)
Activin Receptors, Type II/genetics , Erythropoiesis/drug effects , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , beta-Thalassemia/drug therapy , Activin Receptors, Type II/metabolism , Anemia/blood , Anemia/genetics , Anemia/prevention & control , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/genetics , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Hemolysis/drug effects , Hemolysis/genetics , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Iron Overload/metabolism , Iron Overload/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/blood , beta-Thalassemia/genetics
16.
Nat Med ; 20(4): 408-14, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24658078

ABSTRACT

Erythropoietin (EPO) stimulates proliferation of early-stage erythrocyte precursors and is widely used for the treatment of chronic anemia. However, several types of EPO-resistant anemia are characterized by defects in late-stage erythropoiesis, which is EPO independent. Here we investigated regulation of erythropoiesis using a ligand-trapping fusion protein (ACE-536) containing the extracellular domain of human activin receptor type IIB (ActRIIB) modified to reduce activin binding. ACE-536, or its mouse version RAP-536, produced rapid and robust increases in erythrocyte numbers in multiple species under basal conditions and reduced or prevented anemia in murine models. Unlike EPO, RAP-536 promoted maturation of late-stage erythroid precursors in vivo. Cotreatment with ACE-536 and EPO produced a synergistic erythropoietic response. ACE-536 bound growth differentiation factor-11 (GDF11) and potently inhibited GDF11-mediated Smad2/3 signaling. GDF11 inhibited erythroid maturation in mice in vivo and ex vivo. Expression of GDF11 and ActRIIB in erythroid precursors decreased progressively with maturation, suggesting an inhibitory role for GDF11 in late-stage erythroid differentiation. RAP-536 treatment also reduced Smad2/3 activation, anemia, erythroid hyperplasia and ineffective erythropoiesis in a mouse model of myelodysplastic syndromes (MDS). These findings implicate transforming growth factor-ß (TGF-ß) superfamily signaling in erythroid maturation and identify ACE-536 as a new potential treatment for anemia, including that caused by ineffective erythropoiesis.


Subject(s)
Activin Receptors, Type II , Anemia/blood , Bone Morphogenetic Proteins/drug effects , Erythroid Precursor Cells/drug effects , Erythropoiesis/drug effects , Growth Differentiation Factors/drug effects , Hematinics/pharmacology , Myelodysplastic Syndromes/blood , Recombinant Fusion Proteins/pharmacology , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Disease Models, Animal , Drug Therapy, Combination , Erythrocyte Count , Erythropoietin/pharmacology , Growth Differentiation Factors/antagonists & inhibitors , Haplorhini , Humans , Ligands , Mice , Rats , Reticulocyte Count , Signal Transduction/drug effects , Smad2 Protein/drug effects , Smad3 Protein/drug effects
17.
Proc Natl Acad Sci U S A ; 109(30): 12207-12, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22761317

ABSTRACT

Diseases such as osteoporosis are associated with reduced bone mass. Therapies to prevent bone loss exist, but there are few that stimulate bone formation and restore bone mass. Bone morphogenetic proteins (BMPs) are members of the TGFß superfamily, which act as pleiotropic regulators of skeletal organogenesis and bone homeostasis. Ablation of the BMPR1A receptor in osteoblasts increases bone mass, suggesting that inhibition of BMPR1A signaling may have therapeutic benefit. The aim of this study was to determine the skeletal effects of systemic administration of a soluble BMPR1A fusion protein (mBMPR1A-mFc) in vivo. mBMPR1A-mFc was shown to bind BMP2/4 specifically and with high affinity and prevent downstream signaling. mBMPR1A-mFc treatment of immature and mature mice increased bone mineral density, cortical thickness, trabecular bone volume, thickness and number, and decreased trabecular separation. The increase in bone mass was due to an early increase in osteoblast number and bone formation rate, mediated by a suppression of Dickkopf-1 expression. This was followed by a decrease in osteoclast number and eroded surface, which was associated with a decrease in receptor activator of NF-κB ligand (RANKL) production, an increase in osteoprotegerin expression, and a decrease in serum tartrate-resistant acid phosphatase (TRAP5b) concentration. mBMPR1A treatment also increased bone mass and strength in mice with bone loss due to estrogen deficiency. In conclusion, mBMPR1A-mFc stimulates osteoblastic bone formation and decreases bone resorption, which leads to an increase in bone mass, and offers a promising unique alternative for the treatment of bone-related disorders.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone and Bones/drug effects , Osteogenesis/drug effects , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , Analysis of Variance , Animals , Blotting, Western , Bone Density/drug effects , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Resorption/drug therapy , Bone and Bones/anatomy & histology , Bone and Bones/physiology , Chromatography, Gel , Cloning, Molecular , Densitometry , Electrophoresis, Polyacrylamide Gel , Intercellular Signaling Peptides and Proteins/metabolism , Luciferases , Mice , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoclasts/physiology , Osteoprotegerin/metabolism , Polymerase Chain Reaction , RANK Ligand/metabolism , Real-Time Polymerase Chain Reaction , Recombinant Fusion Proteins/administration & dosage , Signal Transduction/physiology
18.
G3 (Bethesda) ; 2(12): 1577-84, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23275880

ABSTRACT

Colorectal cancer (CRC) has a complex etiology resulting from the combination of multiple genetic and environmental factors, each with small effects. Interactions among susceptibility modifier loci make many of the loci difficult to detect in human genome-wide association studies. Previous analyses in mice have used classical inbred strains, which share large portions of their genomes due to common ancestry. Herein, we used an interspecific backcross between the Mus musculus strain A/J and the Mus spretus strain SPRET/EiJ to map 6 additional CRC modifier loci (Scc16-21) and 2 suggestive loci. Three loci modify the location of tumors along the proximal-distal axis of the colon. Six CRC modifiers previously mapped in intraspecific crosses were also replicated. This work confirms genetic models suggesting that CRC is caused by many small effect alleles and brings the catalog of reported CRC modifier loci to 23 spread across 13 chromosomes. Furthermore, this work provides the foundation for large population-level epistatic interaction tests to identify combinations of low effect alleles that may have large effects on CRC susceptibility.


Subject(s)
Chromosome Mapping , Colonic Neoplasms/genetics , Genetic Predisposition to Disease , Alleles , Animals , Chromosomes/genetics , Colonic Neoplasms/metabolism , Crosses, Genetic , Genome , Genotype , Mice , Phenotype , Quantitative Trait Loci
19.
Curr Mol Pharmacol ; 5(2): 195-204, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21787285

ABSTRACT

Current antiresorptive therapies not only prevent bone loss by decreasing osteoclastic bone resorption but also inhibit bone formation. Dual anabolic antiresorptive agents may be required to cure severe osteoporosis by preventing further bone loss and increasing bone mass to normal levels. Recent studies have demonstrated that activin signaling plays a crucial role in the skeleton. Activins, like other TGF-ß superfamily members, transduce their signals through type I and II receptor serine/threonine kinases. The binding of activins to activin type IIA (ActRIIA) or type IIB (ActRIIB) receptors induces the recruitment and phosphorylation of an activin type I receptor (ALK4 and/or ALK7), which then phosphorylates the Smad2 and Smad3 intracellular signaling proteins. Activin signaling is down-regulated by inhibins, follistatin and other proteins, which antagonize activin signaling by a variety of mechanisms. A soluble chimeric protein composed of the extracellular domain of ActRIIA fused to IgG-Fc binds to circulating ligands such as activin A and prevents signaling through the endogenous receptor. In cynomolgus monkeys, the ActRIIA soluble receptor increases bone volume by decreasing bone resorption and increasing bone formation, leading to enhanced mechanical strength and bone quality. In addition, a single dose of the soluble ActRIIA-Fc fusion protein increased serum BSALP and PINP and decreased serum CTX and TRACP 5b in postmenopausal women. These data provide evidence of a dual anabolic antiresorptive effect of the soluble ActRIIA-Fc fusion protein in the skeleton. Therefore, targeting activin receptor signaling may be useful for therapeutic intervention in osteoporosis.


Subject(s)
Activin Receptors/metabolism , Osteoporosis/drug therapy , Activin Receptors/antagonists & inhibitors , Activin Receptors, Type I/metabolism , Activin Receptors, Type II/metabolism , Activins/antagonists & inhibitors , Activins/genetics , Activins/metabolism , Animals , Bone Density Conservation Agents/pharmacology , Bone Density Conservation Agents/therapeutic use , Bone Remodeling/drug effects , Bone and Bones/metabolism , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/drug effects
20.
J Biol Chem ; 286(34): 30034-46, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21737454

ABSTRACT

Endoglin (CD105), a transmembrane protein of the transforming growth factor ß superfamily, plays a crucial role in angiogenesis. Mutations in endoglin result in the vascular defect known as hereditary hemorrhagic telangiectasia (HHT1). The soluble form of endoglin was suggested to contribute to the pathogenesis of preeclampsia. To obtain further insight into its function, we cloned, expressed, purified, and characterized the extracellular domain (ECD) of mouse and human endoglin fused to an immunoglobulin Fc domain. We found that mouse and human endoglin ECD-Fc bound directly, specifically, and with high affinity to bone morphogenetic proteins 9 and 10 (BMP9 and BMP10) in surface plasmon resonance (Biacore) and cell-based assays. We performed a function mapping analysis of the different domains of endoglin by examining their contributions to the selectivity and biological activity of the protein. The BMP9/BMP10 binding site was localized to the orphan domain of human endoglin composed of the amino acid sequence 26-359. We established that endoglin and type II receptors bind to overlapping sites on BMP9. In the in vivo chick chorioallantoic membrane assay, the mouse and the truncated human endoglin ECD-Fc both significantly reduced VEGF-induced vessel formation. Finally, murine endoglin ECD-Fc acted as an anti-angiogenic factor that decreased blood vessel sprouting in VEGF/FGF-induced angiogenesis in in vivo angioreactors and reduced the tumor burden in the colon-26 mouse tumor model. Together our findings indicate an important role of soluble endoglin ECD in the regulation of angiogenesis and highlight efficacy of endoglin-Fc as a potential anti-angiogenesis therapeutic agent.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antigens, CD/pharmacology , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factor 2/metabolism , Growth Differentiation Factors/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Binding Sites , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Proteins/genetics , Cell Line , Endoglin , Growth Differentiation Factor 2/genetics , Growth Differentiation Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neoplasms/genetics , Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...