1
|
Zhou Y, Shi C, Sun H. Advancements in mechanisms and drug treatments for fibrodysplasia ossificans progressiva. J Zhejiang Univ Sci B 2025; 26:317-332. [PMID: 40274382 PMCID: PMC12021541 DOI: 10.1631/jzus.b2300779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 01/12/2024] [Indexed: 04/26/2025]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder characterized by congenital bilateral malformation of the large toe and progressive, extensive, and irreversible heterotopic ossification (HO) of soft tissues throughout the body, leading to severe disabilities. FOP is caused primarily by mutations in activin A receptor type 1 (ACVR1), also known as activin-like kinase 2 (ALK2), which encodes a receptor belonging to the bone morphogenetic protein (BMP) type I family. However, the continuous and complex process of HO in FOP is not yet fully understood, which has impeded the development of therapeutic drugs. Despite surgical removal of HO, which often results in recurrence and expansion of ossification, there is currently no definitive drug treatment available to completely prevent, halt, or reverse the progression of HO in FOP. Currently, researchers are intensively studying the pathogenesis of FOP at various stages and developing promising drug candidates, including saracatinib, palovarotene, and rapamycin. This review provides an overview of progress in understanding the mechanism of FOP and the development of therapeutic drugs, with the goal of providing insights for further research and the development of new treatment methods.
Collapse
Affiliation(s)
- Yijun Zhou
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun 130021, China
| | - Ce Shi
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun 130021, China.
| | - Hongchen Sun
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun 130021, China.
| |
Collapse
|
2
|
Yang YO, Gong X, Getsy J, Wang P, Liu X, Sheng J, Chen X, Rockich K. Pharmacokinetics of Zilurgisertib With and Without Food from Single and Multiple Ascending Dose Phase 1 Studies in Healthy Adults. Eur J Drug Metab Pharmacokinet 2025; 50:65-80. [PMID: 39652202 PMCID: PMC11802711 DOI: 10.1007/s13318-024-00926-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 02/07/2025]
Abstract
BACKGROUND AND OBJECTIVES The oral, potent, and highly selective activin receptor-like kinase 2 (ALK2) inhibitor zilurgisertib (INCB000928) is in development as a treatment for fibrodysplasia ossificans progressiva (FOP), and for anemia due to myelofibrosis, myelodysplastic syndromes, and multiple myeloma. Saliva is an attractive alternative to blood for drug monitoring and pharmacokinetic analysis, as it is non-invasive to retrieve. This is beneficial for patients, such as those with FOP, for whom blood draws can be challenging due to soft tissue damage susceptibility that can cause progressive heterotopic ossification, and for whom tourniquet time and blood draws must be minimized. The objectives of these studies were to evaluate zilurgisertib pharmacokinetics, safety, tolerability, and the effect of food in healthy participants from phase 1 single ascending dose (SAD) and multiple ascending dose (MAD) studies. METHODS Both the SAD and MAD studies were double-blind, randomized, placebo-controlled dose escalation studies. In the SAD study, healthy participants received a single oral dose of zilurgisertib (10, 25, 50, 100, 175, 250, or 500 mg) or placebo in the fasted state. A further group of healthy participants were enrolled into an additional "food effect" cohort and randomized to receive a single oral dose of zilurgisertib (100 mg) after either an overnight fast or a high-fat meal in a 2-way crossover manner. In the MAD study, healthy participants received oral zilurgisertib at 50, 100, 150, 200, or 400 mg once daily or 300 mg twice daily in the fasted state. Blood, saliva, and urine samples were collected for zilurgisertib pharmacokinetic analysis. Safety was assessed throughout both studies. RESULTS Overall, 91 participants (70 active, 21 placebo) were enrolled and randomized to the SAD study and 79 participants (59 active, 20 placebo) were enrolled and randomized to the MAD study. Zilurgisertib was generally well tolerated, and adverse events were generally of mild-to-moderate severity. Zilurgisertib was rapidly absorbed, with median time to maximum plasma drug concentration (Cmax) of 2.0-4.1 h post-dose. Zilurgisertib exposure was more than dose proportional after single and multiple doses over the dose range tested, suggesting non-linear pharmacokinetics. Plasma half-life values ranged from 22.8 to 31.4 h, supporting once-daily dosing. There was a strong correlation between zilurgisertib concentrations in saliva and plasma. No food effect was observed on zilurgisertib pharmacokinetics, with geometric mean ratio (90% confidence interval) Cmax and area under the plasma concentration-time curve values of 0.98 (0.91 to1.06) and 1.03 (0.97 to 1.10). Renal excretion under fasted conditions was 16% and 27% of total drug clearance with single and multiple doses, respectively; therefore, it was not the predominant pathway for zilurgisertib elimination. CONCLUSIONS Zilurgisertib exhibited a favorable pharmacokinetic profile amenable to once-daily dosing that can be administered without regard to food. Study results support further clinical development of zilurgisertib in patients.
Collapse
Affiliation(s)
- Yan-Ou Yang
- Department of Clinical Pharmacology and Pharmacometrics, Incyte Research Institute, Wilmington, DE, USA.
| | - Xiaohua Gong
- Department of Clinical Pharmacology and Pharmacometrics, Incyte Research Institute, Wilmington, DE, USA
| | - Jay Getsy
- Department of Early Development, Incyte Corporation, Wilmington, DE, USA
| | - Phillip Wang
- Department of Bioanalysis, Incyte Research Institute, Wilmington, DE, USA
| | - Xiang Liu
- Department of Clinical Pharmacology and Pharmacometrics, Incyte Research Institute, Wilmington, DE, USA
| | - Jennifer Sheng
- Department of Clinical Pharmacology and Pharmacometrics, Incyte Research Institute, Wilmington, DE, USA
| | - Xuejun Chen
- Department of Clinical Pharmacology and Pharmacometrics, Incyte Research Institute, Wilmington, DE, USA
| | - Kevin Rockich
- Department of Early Development, Incyte Corporation, Wilmington, DE, USA
| |
Collapse
|
3
|
Tang W, Gu Z, Guo J, Lin M, Tao H, Jia D, Jia P. Activins and Inhibins in Cardiovascular Pathophysiology. Biomolecules 2024; 14:1462. [PMID: 39595638 PMCID: PMC11592067 DOI: 10.3390/biom14111462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Activins and inhibins, members of the transforming growth factor β (TGFβ) superfamily, were initially recognized for their opposing effects on the secretion of follicle-stimulating hormone. Subsequent research has demonstrated their broader biological roles across various tissue types. Primarily, activins and inhibins function through the classical TGFβ SMAD signaling pathway, but studies suggest that they also act through other pathways, with their specific signaling being complex and context-dependent. Recent research has identified significant roles for activins and inhibins in the cardiovascular system. Their actions in other systems and their signaling pathways show strong correlations with the development and progression of cardiovascular diseases, indicating potential broader roles in the cardiovascular system. This review summarizes the progress in research on the biological functions and mechanisms of activins and inhibins and their signaling pathways in cardiovascular diseases, offering new insights for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Dalin Jia
- Department of Cardiology, The First Hospital of China Medical University, Shenyang 110001, China; (W.T.); (Z.G.); (J.G.); (M.L.); (H.T.)
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, Shenyang 110001, China; (W.T.); (Z.G.); (J.G.); (M.L.); (H.T.)
| |
Collapse
|
4
|
Siraj Y, Aprile D, Alessio N, Peluso G, Di Bernardo G, Galderisi U. IGFBP7 is a key component of the senescence-associated secretory phenotype (SASP) that induces senescence in healthy cells by modulating the insulin, IGF, and activin A pathways. Cell Commun Signal 2024; 22:540. [PMID: 39533382 PMCID: PMC11558980 DOI: 10.1186/s12964-024-01921-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Senescent cells exert their effects through the release of various factors, collectively referred to as the senescence-associated secretory phenotype (SASP). The SASP can induce senescence in healthy cells (secondary senescence), modulate immune system function, reshape the extracellular matrix, and facilitate cancer progression.Among SASP components, certain factors act as key regulators in the induction of secondary senescence. In this study, we evaluated the role of IGFBP7, a crucial SASP component. Our results demonstrated that ROS-prostaglandin signaling is involved in the release of IGFBP7. Furthermore, neutralizing antibodies targeting IGFBP7 attenuated the SASP's pro-senescence activity. Cells incubated with IGFBP7 also entered a state of senescence.The senescence induced by IGFBP7 appears to be mediated through three primary pathways. First, IGFBP7 can bind to insulin, thereby inhibiting its anti-senescence and pro-growth effects. In addition to this inhibitory effect on the insulin pathway, IGFBP7 may enhance IGFII pro-senescence signaling by promoting its interaction with IGF2R while blocking IGF1R. These activities are dependent on ERK and AKT signaling pathways. Finally, IGFBP7 and Activin A, both of which can induce cellular senescence, appear to regulate and inhibit each other, suggesting a compensatory mechanism to prevent excessive senescence. Notably, our preliminary data indicate that IGFBP7, in addition to blocking Activin A, may interact with its receptors and induce senescence via SMAD pathways.Our findings highlight that IGFBP7, along with other members of the IGFBP family, plays a pivotal role in senescence-related signaling pathways. Therefore, IGFBP7 may serve as a potential target for anti-aging strategies aimed at reducing the burden of senescence on tissues and organs.
Collapse
Affiliation(s)
- Yesuf Siraj
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
- College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | | | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.
- Department of Experimental Medicine, Luigi Vanvitelli Campania Univeristy, Via Luigi De Crecchio 7, Napoli, 80138, Italy.
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.
- Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Turkey.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, USA.
- Department of Experimental Medicine, Luigi Vanvitelli Campania Univeristy, Via Luigi De Crecchio 7, Napoli, 80138, Italy.
| |
Collapse
|
5
|
Stump B, Waxman AB. Pulmonary Arterial Hypertension and TGF-β Superfamily Signaling: Focus on Sotatercept. BioDrugs 2024; 38:743-753. [PMID: 39292393 DOI: 10.1007/s40259-024-00680-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and progressive disease that continues to remain highly morbid despite multiple advances in medical therapies. There remains a persistent and desperate need to identify novel methods of treating and, ideally, reversing the pathologic vasculopathy that results in PAH development and progression. Sotatercept is a first-in-class fusion protein that is believed to primarily inhibit activin signaling resulting in decreased cell proliferation and differentiation, though the exact mechanism remains uncertain. Here, we review the currently available PAH therapies, data highlighting the importance of transforming growth factor-β (TGF-β) superfamily signaling in the development of PAH, and the published and on-going clinical trials evaluating sotatercept in the treatment of PAH. We will also discuss preclinical data supporting the potential use of the fusion protein KER-012 in the inhibition of aberrant TGF-β superfamily signaling to ameliorate the obstructive vasculopathy of PAH.
Collapse
|
6
|
Pinjusic K, Bulliard M, Rothé B, Ansaryan S, Liu YC, Ginefra P, Schmuziger C, Altug H, Constam DB. Stepwise release of Activin-A from its inhibitory prodomain is modulated by cysteines and requires furin coexpression to promote melanoma growth. Commun Biol 2024; 7:1383. [PMID: 39448726 PMCID: PMC11502825 DOI: 10.1038/s42003-024-07053-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
The Activin-A precursor dimer can be cleaved by furin, but how this proteolytic maturation is regulated in vivo and how it facilitates access to signaling receptors is unclear. Here, analysis in a syngeneic melanoma grafting model shows that without furin coexpression, Activin-A failed to accelerate tumor growth, correlating with failure of one or both subunits to undergo cleavage in signal-sending cells, even though compensatory processing by host cells nonetheless sustained elevated circulating Activin-A levels. In reporter assays, furin-independent cleavage of one subunit enabled juxtacrine Activin-A signaling, whereas completion of proteolytic maturation by coexpressed furin or by recipient cells stimulated contact-independent activity, crosstalk with BMP receptors, and signal inhibition by follistatin. Mechanistically, Activin-A processing was modulated by allosteric disulfide bonds flanking the furin site. Disruption of these disulfide linkages with the prodomain enabled Activin-A binding to cognate type II receptors independently of proteolytic maturation. Stepwise proteolytic maturation is a novel mechanism to control Activin-A protein interactions and signaling.
Collapse
Affiliation(s)
- Katarina Pinjusic
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, Lausanne, Switzerland
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Manon Bulliard
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, Lausanne, Switzerland
| | - Benjamin Rothé
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, Lausanne, Switzerland
| | - Saeid Ansaryan
- Ecole Polytechnique Fédérale de Lausanne (EPFL) STI IBI-STI BIOS BM, Station 17, Lausanne, Switzerland
| | - Yeng-Cheng Liu
- Ecole Polytechnique Fédérale de Lausanne (EPFL) STI IBI-STI BIOS BM, Station 17, Lausanne, Switzerland
| | - Pierpaolo Ginefra
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, Lausanne, Switzerland
- University of Lausanne, Department of Oncology, Ludwig Cancer Institute, Epalinges, Switzerland
| | - Céline Schmuziger
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, Lausanne, Switzerland
| | - Hatice Altug
- Ecole Polytechnique Fédérale de Lausanne (EPFL) STI IBI-STI BIOS BM, Station 17, Lausanne, Switzerland
| | - Daniel B Constam
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, Lausanne, Switzerland.
| |
Collapse
|
7
|
Towler OW, Shore EM, Kaplan FS. Molecular Developmental Biology of Fibrodysplasia Ossificans Progressiva: Measuring the Giant by Its Toe. Biomolecules 2024; 14:1009. [PMID: 39199396 PMCID: PMC11353020 DOI: 10.3390/biom14081009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
When a genetic disease is characterized by the abnormal activation of normal molecular pathways and cellular events, it is illuminating to critically examine the places and times of these activities both in health and disease. Therefore, because heterotopic ossification (HO) in fibrodysplasia ossificans progressiva (FOP) is by far the disease's most prominent symptom, attention is also directed toward the pathways and processes of bone formation during skeletal development. FOP is recognizable by effects of the causative mutation on skeletal development even before HO manifests, specifically in the malformation of the great toes. This signature skeletal phenotype is the most highly penetrant, but is only one among several skeletal abnormalities associated with FOP. Patients may present clinically with joint malformation and ankylosis, particularly in the cervical spine and costovertebral joints, as well as characteristic facial features and a litany of less common, non-skeletal symptoms, all stemming from missense mutations in the ACVR1 gene. In the same way that studying the genetic cause of HO advanced our understanding of HO initiation and progression, insight into the roles of ACVR1 signaling during tissue development, particularly in the musculoskeletal system, can be gained from examining altered skeletal development in individuals with FOP. This review will detail what is known about the molecular mechanisms of developmental phenotypes in FOP and the early role of ACVR1 in skeletal patterning and growth, as well as highlight how better understanding these processes may serve to advance patient care, assessments of patient outcomes, and the fields of bone and joint biology.
Collapse
Affiliation(s)
- O. Will Towler
- Division of Plastic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Eileen M. Shore
- Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frederick S. Kaplan
- Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
8
|
He K, Jiang H, Li W, Toutounchi S, Huang Y, Wu J, Ma X, Baehr W, Pignolo RJ, Ling K, Zhou X, Wang H, Hu J. Primary cilia mediate skeletogenic BMP and Hedgehog signaling in heterotopic ossification. Sci Transl Med 2024; 16:eabn3486. [PMID: 39047114 DOI: 10.1126/scitranslmed.abn3486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/04/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
Heterotopic ossification (HO), defined as the formation of extraskeletal bone in muscle and soft tissues, is a diverse pathological process caused by either genetic mutations or inciting trauma. Fibrodysplasia ossificans progressiva (FOP) is a genetic form of HO caused by mutations in the bone morphogenetic protein (BMP) type I receptor gene activin A receptor type 1 (ACVR1). These mutations make ACVR1 hypersensitive to BMP and responsive to activin A. Hedgehog (Hh) signaling also contributes to HO development. However, the exact pathophysiology of how skeletogenic cells contribute to endochondral ossification in FOP remains unknown. Here, we showed that the wild-type or FOP-mutant ACVR1 localized in the cilia of stem cells from human exfoliated deciduous teeth with key FOP signaling components, including activin A receptor type 2A/2B, SMAD family member 1/5, and FK506-binding protein 12kD. Cilia suppression by deletion of intraflagellar transport 88 or ADP ribosylation factor like GTPase 3 effectively inhibited pathological BMP and Hh signaling, subdued aberrant chondro-osteogenic differentiation in primary mouse or human FOP cells, and diminished in vivo extraskeletal ossification in Acvr1Q207D, Sox2-Cre; Acvr1R206H/+ FOP mice and in burn tenotomy-treated wild-type mice. Our results provide a rationale for early and localized suppression of cilia in affected tissues after injury as a therapeutic strategy against either genetic or acquired HO.
Collapse
Affiliation(s)
- Kai He
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Heng Jiang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Weijun Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905 USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Saman Toutounchi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905 USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Yan Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Jianfeng Wu
- Department of Orthopedics, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Xiaoyu Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Wolfgang Baehr
- Department of Ophthalmology, University of Utah, Salt Lake City, UT 84132, USA
| | - Robert J Pignolo
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905 USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Xuhui Zhou
- Translational Research Center of Orthopedics, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Haitao Wang
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905 USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Robert M. and Billie Kelley Pirnie Translational Polycystic Kidney Disease Center, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
9
|
Li W, Quigley K. Bone morphogenetic protein signalling in pulmonary arterial hypertension: revisiting the BMPRII connection. Biochem Soc Trans 2024; 52:1515-1528. [PMID: 38716930 PMCID: PMC11346422 DOI: 10.1042/bst20231547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and life-threatening vascular disorder, characterised by abnormal remodelling of the pulmonary vessels and elevated pulmonary artery pressure, leading to right ventricular hypertrophy and right-sided heart failure. The importance of bone morphogenetic protein (BMP) signalling in the pathogenesis of PAH is demonstrated by human genetic studies. Many PAH risk genes are involved in the BMP signalling pathway and are highly expressed or preferentially act on vascular endothelial cells. Endothelial dysfunction is recognised as an initial trigger for PAH, and endothelial BMP signalling plays a crucial role in the maintenance of endothelial integrity. BMPR2 is the most prevalent PAH gene, found in over 80% of heritable cases. As BMPRII protein is the major type II receptor for a large family of BMP ligands and expressed ubiquitously in many tissues, dysregulated BMP signalling in other cells may also contribute to PAH pathobiology. Sotatercept, which contains the extracellular domain of another transforming growth factor-β family type II receptor ActRIIA fused to immunoglobin Fc domain, was recently approved by the FDA as a treatment for PAH. Neither its target cells nor its mechanism of action is fully understood. This review will revisit BMPRII function and its extracellular regulation, summarise how dysregulated BMP signalling in endothelial cells and smooth muscle cells may contribute to PAH pathogenesis, and discuss how novel therapeutics targeting the extracellular regulation of BMP signalling, such as BMP9 and Sotatercept, can be related to restoring BMPRII function.
Collapse
Affiliation(s)
- Wei Li
- VPD Heart and Lung Research Institute, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0BB, U.K
| | - Kate Quigley
- VPD Heart and Lung Research Institute, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0BB, U.K
| |
Collapse
|
10
|
Gao P, Inada Y, Hotta A, Sakurai H, Ikeya M. iMSC-mediated delivery of ACVR2B-Fc fusion protein reduces heterotopic ossification in a mouse model of fibrodysplasia ossificans progressiva. Stem Cell Res Ther 2024; 15:83. [PMID: 38500216 PMCID: PMC10949803 DOI: 10.1186/s13287-024-03691-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/07/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease caused by a gain-of-function mutation in ACVR1, which is a bone morphogenetic protein (BMP) type I receptor. Moreover, it causes progressive heterotopic ossification (HO) in connective tissues. Using FOP patient-derived induced pluripotent stem cells (FOP-iPSCs) and mouse models, we elucidated the underlying mechanisms of FOP pathogenesis and identified a candidate drug for FOP. METHODS In the current study, healthy mesenchymal stem/stromal cells derived from iPSCs (iMSCs) expressing ACVR2B-Fc (iMSCACVR2B-Fc), which is a neutralizing receptobody, were constructed. Furthermore, patient-derived iMSCs and FOP mouse model (ACVR1R206H, female) were used to confirm the inhibitory function of ACVR2B-Fc fusion protein secreted by iMSCACVR2B-Fc on BMP signaling pathways and HO development, respectively. RESULTS We found that secreted ACVR2B-Fc attenuated BMP signaling initiated by Activin-A and BMP-9 in both iMSCs and FOP-iMSCs in vitro. Transplantation of ACVR2B-Fc-expressing iMSCs reduced primary HO in a transgenic mouse model of FOP. Notably, a local injection of ACVR2B-Fc-expressing iMSCs and not an intraperitoneal injection improved the treadmill performance, suggesting compound effects of ACVR2B-Fc and iMSCs. CONCLUSIONS These results offer a new perspective for treating FOP through stem cell therapy.
Collapse
Affiliation(s)
- Pan Gao
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and, Department of General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53, Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshiko Inada
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53, Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akitsu Hotta
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53, Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hidetoshi Sakurai
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53, Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53, Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
11
|
Burdick LN, DelVichio AH, Hanson LR, Griffith BB, Bouchard KR, Hunter JW, Goldhamer DJ. Sex as a Critical Variable in Basic and Pre-Clinical Studies of Fibrodysplasia Ossificans Progressiva. Biomolecules 2024; 14:177. [PMID: 38397414 PMCID: PMC10886767 DOI: 10.3390/biom14020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Heterotopic ossification (HO) is most dramatically manifested in the rare and severely debilitating disease, fibrodysplasia ossificans progressiva (FOP), in which heterotopic bone progressively accumulates in skeletal muscles and associated soft tissues. The great majority of FOP cases are caused by a single amino acid substitution in the type 1 bone morphogenetic protein (BMP) receptor ACVR1, a mutation that imparts responsiveness to activin A. Although it is well-established that biological sex is a critical variable in a range of physiological and disease processes, the impact of sex on HO in animal models of FOP has not been explored. We show that female FOP mice exhibit both significantly greater and more variable HO responses after muscle injury. Additionally, the incidence of spontaneous HO was significantly greater in female mice. This sex dimorphism is not dependent on gonadally derived sex hormones, and reciprocal cell transplantations indicate that apparent differences in osteogenic activity are intrinsic to the sex of the transplanted cells. By circumventing the absolute requirement for activin A using an agonist of mutant ACVR1, we show that the female-specific response to muscle injury or BMP2 implantation is dependent on activin A. These data identify sex as a critical variable in basic and pre-clinical studies of FOP.
Collapse
Affiliation(s)
- Lorraine N. Burdick
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT 06269, USA; (L.N.B.); (A.H.D.); (L.R.H.); (B.B.G.)
| | - Amanda H. DelVichio
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT 06269, USA; (L.N.B.); (A.H.D.); (L.R.H.); (B.B.G.)
| | - L. Russell Hanson
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT 06269, USA; (L.N.B.); (A.H.D.); (L.R.H.); (B.B.G.)
| | - Brenden B. Griffith
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT 06269, USA; (L.N.B.); (A.H.D.); (L.R.H.); (B.B.G.)
| | - Keith R. Bouchard
- Alexion Pharmaceuticals Inc., 100 College Street, New Haven, CT 06510, USA; (K.R.B.); (J.W.H.)
| | - Jeffrey W. Hunter
- Alexion Pharmaceuticals Inc., 100 College Street, New Haven, CT 06510, USA; (K.R.B.); (J.W.H.)
| | - David J. Goldhamer
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT 06269, USA; (L.N.B.); (A.H.D.); (L.R.H.); (B.B.G.)
| |
Collapse
|
12
|
Wang Y, Nguyen JH, de Ruiter RD, Mendell J, Srinivasan D, Davis JD, Eekhoff EMW. Garetosmab in Fibrodysplasia Ossificans Progressiva: Clinical Pharmacology Results from the Phase 2 LUMINA-1 Trial. J Clin Pharmacol 2024; 64:264-274. [PMID: 37694449 DOI: 10.1002/jcph.2344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
Here, we report the clinical pharmacology data from LUMINA-1 (NCT03188666), a Phase 2 trial that evaluated garetosmab (a monoclonal antibody against activin A) in patients with fibrodysplasia ossificans progressiva. Forty-four patients were randomly assigned to intravenous 10 mg/kg of garetosmab or placebo every 4 weeks in a double-blind 28-week treatment period, followed by a 28-week open-label treatment period with garetosmab, and subsequent open-label extension. Serum samples were obtained to assess pharmacokinetics (PK), immunogenicity, and bone morphogenetic protein 9 (BMP9). Comparative exposure-response analyses for efficacy and safety were performed with trough concentrations (Ctrough ) of garetosmab prior to dosing. Steady-state PK was reached 12-16 weeks after the first dose of garetosmab, with mean (standard deviation) Ctrough of 105 ± 30.8 mg/L. Immunogenicity assessments showed anti-garetosmab antibody formation in 1 patient (1/43; 2.3%); titers were low, and did not affect PK or clinical efficacy. Median concentrations of BMP9 in serum were approximately 40 pg/mL at baseline. There were no meaningful differences in PK or BMP9 concentration-time profiles between patients who did and did not experience epistaxis or death. The comparative exposure-response analyses demonstrated no association between Ctrough and efficacy or safety. PK findings were consistent with prior data in healthy volunteers and were typical for a monoclonal antibody administered at doses sufficient to saturate target-mediated clearance. There were no trends that suggested patients with higher serum exposures to garetosmab were more likely to experience a reduction in heterotopic ossification or adverse events. Garetosmab is being further evaluated in the Phase 3 OPTIMA trial.
Collapse
Affiliation(s)
- Yuhuan Wang
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Ruben D de Ruiter
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Vrije Universiteit, Amsterdam UMC Expert Center in Rare Bone Disease, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | | | | | | | - E Marelise W Eekhoff
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Vrije Universiteit, Amsterdam UMC Expert Center in Rare Bone Disease, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Szilágyi SS, Burdzinski W, Jatzlau J, Ehrlich M, Knaus P, Henis YI. The Activation of the Fibrodysplasia Ossificans Progressiva-Inducing ALK2-R206H Mutant Depends on the Distinct Homo-Oligomerization Patterns of ACVR2B and ACVR2A. Cells 2024; 13:221. [PMID: 38334613 PMCID: PMC10854824 DOI: 10.3390/cells13030221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Mutations in activin-like kinase 2 (ALK2), e.g., ALK2-R206H, induce aberrant signaling to SMAD1/5/8, leading to Fibrodysplasia Ossificans Progressiva (FOP). In spite of extensive studies, the underlying mechanism is still unclear. Here, we quantified the homomeric and heteromeric interactions of ACVR2A, ACVR2B, ALK2-WT, and ALK2-R206H by combining IgG-mediated immobilization of one receptor with fluorescence recovery after photobleaching (FRAP) measurements on the lateral diffusion of a co-expressed receptor. ACVR2B formed stable homomeric complexes that were enhanced by Activin A (ActA), while ACVR2A required ActA for homodimerization. ALK2-WT, but not ALK2-R206H, exhibited homomeric complexes unaffected by ActA. ACVR2B formed ActA-enhanced heterocomplexes with ALK2-R206H or ALK2-WT, while ACVR2A interacted mainly with ALK2-WT. The extent of the homomeric complex formation of ACVR2A or ACVR2B was reflected in their ability to induce the oligomerization of ALK2-R206H and ALK2-WT. Thus, ACVR2B, which forms dimers without ligand, induced ActA-independent ALK2-R206H clustering but required ActA for enhancing the oligomerization of the largely dimeric ALK2-WT. In contrast, ACVR2A, which undergoes homodimerization in response to ActA, required ActA to induce ALK2-R206H oligomerization. To investigate whether these interactions are translated into signaling, we studied signaling by the FOP-inducing hyperactive ALK2-R206H mutant, with ALK2-WT signaling as control. The activation of SMAD1/5/8 signaling in cells expressing ALK2-R206H alone or together with ACVR2A or ACVR2B was measured by blotting for pSMAD1/5/8 and by transcriptional activation assays using BRE-Luc reporter. In line with the biophysical studies, ACVR2B activated ALK2-R206H without ligand, while activation by ACVR2A was weaker and required ActA. We propose that the homodimerization of ACVR2B or ACVR2A dictates their ability to recruit ALK2-R206H into higher complexes, enabling the homomeric interactions of ALK2-R206H receptors and, subsequently, their activation.
Collapse
Affiliation(s)
- Szabina Szófia Szilágyi
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Wiktor Burdzinski
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jerome Jatzlau
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
| | - Yoav I. Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| |
Collapse
|
14
|
Wang H, Kaplan FS, Pignolo RJ. The HIF-1α and mTOR Pathways Amplify Heterotopic Ossification. Biomolecules 2024; 14:147. [PMID: 38397384 PMCID: PMC10887042 DOI: 10.3390/biom14020147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP; MIM# 135100) is an ultra-rare congenital disorder caused by gain-of-function point mutations in the Activin receptor A type I (ACVR1, also known as ALK2) gene. FOP is characterized by episodic heterotopic ossification (HO) in skeletal muscles, tendons, ligaments, or other soft tissues that progressively causes irreversible loss of mobility. FOP mutations cause mild ligand-independent constitutive activation as well as ligand-dependent bone morphogenetic protein (BMP) pathway hypersensitivity of mutant ACVR1. BMP signaling is also a key pathway for mediating acquired HO. However, HO is a highly complex biological process involving multiple interacting signaling pathways. Among them, the hypoxia-inducible factor (HIF) and mechanistic target of rapamycin (mTOR) pathways are intimately involved in both genetic and acquired HO formation. HIF-1α inhibition or mTOR inhibition reduces HO formation in mouse models of FOP or acquired HO in part by de-amplifying the BMP pathway signaling. Here, we review the recent progress on the mechanisms of the HIF-1α and mTOR pathways in the amplification of HO lesions and discuss the future directions and strategies to translate the targeting of HIF-1α and the mTOR pathways into clinical interventions for FOP and other forms of HO.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Medicine, Geriatric Medicine & Gerontology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Frederick S. Kaplan
- Department of Orthopaedic Surgery, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
- The Center for Research in FOP and Related Disorders, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert J. Pignolo
- Department of Medicine, Geriatric Medicine & Gerontology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Medicine, Divisions of Endocrinology, Hospital Internal Medicine, Rochester, MN 55905, USA
| |
Collapse
|
15
|
Srinivasan D, Arostegui M, Goebel EJ, Hart KN, Aykul S, Lees-Shepard JB, Idone V, Hatsell SJ, Economides AN. How Activin A Became a Therapeutic Target in Fibrodysplasia Ossificans Progressiva. Biomolecules 2024; 14:101. [PMID: 38254701 PMCID: PMC10813747 DOI: 10.3390/biom14010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder characterized by episodic yet cumulative heterotopic ossification (HO) of skeletal muscles, tendons, ligaments, and fascia. FOP arises from missense mutations in Activin Receptor type I (ACVR1), a type I bone morphogenetic protein (BMP) receptor. Although initial findings implicated constitutive activity of FOP-variant ACVR1 (ACVR1FOP) and/or hyperactivation by BMPs, it was later shown that HO in FOP requires activation of ACVR1FOP by Activin A. Inhibition of Activin A completely prevents HO in FOP mice, indicating that Activin A is an obligate driver of HO in FOP, and excluding a key role for BMPs in this process. This discovery led to the clinical development of garetosmab, an investigational antibody that blocks Activin A. In a phase 2 trial, garetosmab inhibited new heterotopic bone lesion formation in FOP patients. In contrast, antibodies to ACVR1 activate ACVR1FOP and promote HO in FOP mice. Beyond their potential clinical relevance, these findings have enhanced our understanding of FOP's pathophysiology, leading to the identification of fibroadipogenic progenitors as the cells that form HO, and the discovery of non-signaling complexes between Activin A and wild type ACVR1 and their role in tempering HO, and are also starting to inform biological processes beyond FOP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Aris N. Economides
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA; (D.S.); (M.A.); (E.J.G.); (K.N.H.); (S.A.); (J.B.L.-S.); (V.I.); (S.J.H.)
| |
Collapse
|
16
|
Akhurst RJ. From shape-shifting embryonic cells to oncology: The fascinating history of epithelial mesenchymal transition. Semin Cancer Biol 2023; 96:100-114. [PMID: 37852342 PMCID: PMC10883734 DOI: 10.1016/j.semcancer.2023.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/29/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023]
Abstract
Epithelial-to-mesenchymal transition or transformation (EMT) is a cell shape-changing process that is utilized repeatedly throughout embryogenesis and is critical to the attainment of a precise body plan. In the adult, EMT is observed under both normal and pathological conditions, such as during normal wounding healing, during development of certain fibrotic states and vascular anomalies, as well as in some cancers when malignant cells progress to become more aggressive, invasive, and metastatic. Epithelia derived from any of the three embryonic germ layers can undergo EMT, including those derived from mesoderm, such as endothelial cells (sometimes termed Endo-MT) and those derived from endoderm such as fetal liver stroma. At the cellular level, EMT is defined as the transformation of epithelial cells towards a mesenchymal phenotype and is marked by attenuation of expression of epithelial markers and de novo expression of mesenchymal markers. This process is induced by extracellular factors and can be reversible, resulting in mesenchymal-to-epithelial transformation (MET). It is now clear that a cell can simultaneously express properties of both epithelia and mesenchyme, and that such transitional cell-types drive tumor cell heterogeneity, an important aspect of cancer progression, development of a stem-like cell state, and drug resistance. Here we review some of the earliest studies demonstrating the existence of EMT during embryogenesis and discuss the discovery of the extracellular factors and intracellular signaling pathways that contribute to this process, with components of the TGFβ signaling superfamily playing a prominent role. We mention early controversies surrounding in vivo EMT during embryonic development and in adult diseased states, and the maturation of the field to a stage wherein targeting EMT to control disease states is an aspirational goal.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and UCSF Helen Diller Family Comprehensive Cancer Center, USA
| |
Collapse
|
17
|
Vestal KA, Kattamuri C, Koyiloth M, Ongaro L, Howard JA, Deaton A, Ticau S, Dubey A, Bernard DJ, Thompson TB. Activin E is a TGFβ ligand that signals specifically through activin receptor-like kinase 7. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559288. [PMID: 37808681 PMCID: PMC10557571 DOI: 10.1101/2023.09.25.559288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Activins are one of the three distinct subclasses within the greater Transforming Growth Factor β (TGFβ) superfamily. First discovered for their critical roles in reproductive biology, activins have since been shown to alter cellular differentiation and proliferation. At present, members of the activin subclass include activin A (ActA), ActB, ActC, ActE, and the more distant members myostatin and GDF11. While the biological roles and signaling mechanisms of most activins class members have been well-studied, the signaling potential of ActE has remained largely unknown. Here, we characterized the signaling capacity of homodimeric ActE. Molecular modeling of the ligand:receptor complexes showed that ActC and ActE shared high similarity in both the type I and type II receptor binding epitopes. ActE signaled specifically through ALK7, utilized the canonical activin type II receptors, ActRIIA and ActRIIB, and was resistant to the extracellular antagonists follistatin and WFIKKN. In mature murine adipocytes, ActE invoked a SMAD2/3 response via ALK7, similar to ActC. Collectively, our results establish ActE as an ALK7 ligand, thereby providing a link between genetic and in vivo studies of ActE as a regulator of adipose tissue.
Collapse
Affiliation(s)
- Kylie A Vestal
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Chandramohan Kattamuri
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Muhasin Koyiloth
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Luisina Ongaro
- Department of Pharmacology and Therapeutics, Centre for Research in Reproduction and Development, McGill University, Montreal, Quebec, Canada
| | - James A Howard
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH 45267, USA
| | | | | | | | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, Centre for Research in Reproduction and Development, McGill University, Montreal, Quebec, Canada
| | - Thomas B Thompson
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
18
|
Yang YS, Lin C, Ma H, Xie J, Kaplan FS, Gao G, Shim JH. AAV-Mediated Targeting of the Activin A-ACVR1 R206H Signaling in Fibrodysplasia Ossificans Progressiva. Biomolecules 2023; 13:1364. [PMID: 37759764 PMCID: PMC10526456 DOI: 10.3390/biom13091364] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare genetic disorder characterized by progressive disabling heterotopic ossification (HO) at extra-skeletal sites. Here, we developed adeno-associated virus (AAV)-based gene therapy that suppresses trauma-induced HO in FOP mice harboring a heterozygous allele of human ACVR1R206H (Acvr1R206H/+) while limiting the expression in non-skeletal organs such as the brain, heart, lung, liver, and kidney. AAV gene therapy carrying the combination of codon-optimized human ACVR1 (ACVR1opt) and artificial miRNAs targeting Activin A and its receptor ACVR1R206H ablated the aberrant activation of BMP-Smad1/5 signaling and the osteogenic differentiation of Acvr1R206H/+ skeletal progenitors. The local delivery of AAV gene therapy to HO-causing cells in the skeletal muscle resulted in a significant decrease in endochondral bone formation in Acvr1R206H/+ mice. These mice showed little to no expression in a major AAV-targeted organ, the liver, due to liver-abundant miR-122-mediated repression. Thus, AAV gene therapy is a promising therapeutic strategy to explore in suppressing HO in FOP.
Collapse
Affiliation(s)
- Yeon-Suk Yang
- Department of Medicine, Division of Rheumatology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (Y.-S.Y.); (C.L.)
| | - Chujiao Lin
- Department of Medicine, Division of Rheumatology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (Y.-S.Y.); (C.L.)
| | - Hong Ma
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (H.M.); (J.X.)
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
- Viral Vector Core, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (H.M.); (J.X.)
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
- Viral Vector Core, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Frederick S. Kaplan
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- The Center for Research in FOP and Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (H.M.); (J.X.)
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
- Viral Vector Core, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Jae-Hyuck Shim
- Department of Medicine, Division of Rheumatology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (Y.-S.Y.); (C.L.)
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (H.M.); (J.X.)
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
19
|
Koelsch N, Mirshahi F, Aqbi HF, Saneshaw M, Idowu MO, Olex AL, Sanyal AJ, Manjili MH. The crosstalking immune cells network creates a collective function beyond the function of each cellular constituent during the progression of hepatocellular carcinoma. Sci Rep 2023; 13:12630. [PMID: 37537225 PMCID: PMC10400568 DOI: 10.1038/s41598-023-39020-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023] Open
Abstract
Abundance of data on the role of inflammatory immune responses in the progression or inhibition of hepatocellular carcinoma (HCC) has failed to offer a curative immunotherapy for HCC. This is largely because of focusing on detailed specific cell types and missing the collective function of the hepatic immune system. To discover the collective immune function, we take systems immunology approach by performing high-throughput analysis of snRNAseq data collected from the liver of DIAMOND mice during the progression of nonalcoholic fatty liver disease (NAFLD) to HCC. We report that mutual signaling interactions of the hepatic immune cells in a dominant-subdominant manner, as well as their interaction with structural cells shape the immunological pattern manifesting a collective function beyond the function of the cellular constituents. Such pattern discovery approach recognized direct role of the innate immune cells in the progression of NASH and HCC. These data suggest that discovery of the immune pattern not only detects the immunological mechanism of HCC in spite of dynamic changes in immune cells during the course of disease but also offers immune modulatory interventions for the treatment of NAFLD and HCC.
Collapse
Affiliation(s)
- Nicholas Koelsch
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
| | - Faridoddin Mirshahi
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA, 23298, USA
| | - Hussein F Aqbi
- College of Science, Mustansiriyah University, P.O. Box 14022, Baghdad, Iraq
| | - Mulugeta Saneshaw
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA, 23298, USA
| | - Michael O Idowu
- Department of Pathology, VCU School of Medicine, Richmond, VA, 23298, USA
- Department of Microbiology & Immunology, VCU Massey Cancer Center, 401 College Street, Box 980035, Richmond, VA, 23298, USA
| | - Amy L Olex
- C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Arun J Sanyal
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA, 23298, USA.
- Department of Microbiology & Immunology, VCU Massey Cancer Center, 401 College Street, Box 980035, Richmond, VA, 23298, USA.
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
- Department of Microbiology & Immunology, VCU Massey Cancer Center, 401 College Street, Box 980035, Richmond, VA, 23298, USA.
| |
Collapse
|
20
|
Katagiri T, Tsukamoto S, Kuratani M, Tsuji S, Nakamura K, Ohte S, Kawaguchi Y, Takaishi K. A blocking monoclonal antibody reveals dimerization of intracellular domains of ALK2 associated with genetic disorders. Nat Commun 2023; 14:2960. [PMID: 37231012 PMCID: PMC10212922 DOI: 10.1038/s41467-023-38746-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Mutations in activin receptor-like kinase 2 (ALK2) can cause the pathological osteogenic signaling seen in some patients with fibrodysplasia ossificans progressiva and other conditions such as diffuse intrinsic pontine glioma. Here, we report that intracellular domain of wild-type ALK2 readily dimerizes in response to BMP7 binding to drive osteogenic signaling. This osteogenic signaling is pathologically triggered by heterotetramers of type II receptor kinases and ALK2 mutant forms, which form intracellular domain dimers in response to activin A binding. We develop a blocking monoclonal antibody, Rm0443, that can suppress ALK2 signaling. We solve the crystal structure of the ALK2 extracellular domain complex with a Fab fragment of Rm0443 and show that Rm0443 induces dimerization of ALK2 extracellular domains in a back-to-back orientation on the cell membrane by binding the residues H64 and F63 on opposite faces of the ligand-binding site. Rm0443 could prevent heterotopic ossification in a mouse model of fibrodysplasia ossificans progressiva that carries the human R206H pathogenic mutant.
Collapse
Affiliation(s)
- Takenobu Katagiri
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan.
- Project of Clinical and Basic Research for FOP, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan.
| | - Sho Tsukamoto
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
- Project of Clinical and Basic Research for FOP, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Mai Kuratani
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Shinnosuke Tsuji
- Specialty Medicine Research Laboratories I, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kensuke Nakamura
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Satoshi Ohte
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
- Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Yoshiro Kawaguchi
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kiyosumi Takaishi
- Specialty Medicine Research Laboratories I, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| |
Collapse
|
21
|
Schoenmaker T, Zwaak J, Loos BG, Volckmann R, Koster J, Eekhoff EMW, de Vries TJ. Transcriptomic Differences Underlying the Activin-A Induced Large Osteoclast Formation in Both Healthy Control and Fibrodysplasia Ossificans Progressiva Osteoclasts. Int J Mol Sci 2023; 24:ijms24076822. [PMID: 37047804 PMCID: PMC10095588 DOI: 10.3390/ijms24076822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
Fibrodysplasia Ossificans Progressiva (FOP) is a very rare genetic disease characterized by progressive heterotopic ossification (HO) of soft tissues, leading to immobility and premature death. FOP is caused by a mutation in the Activin receptor Type 1 (ACVR1) gene, resulting in altered responsiveness to Activin-A. We recently revealed that Activin-A induces fewer, but larger and more active, osteoclasts regardless of the presence of the mutated ACVR1 receptor. The underlying mechanism of Activin-A-induced changes in osteoclastogenesis at the gene expression level remains unknown. Transcriptomic changes induced by Activin-A during osteoclast formation from healthy controls and patient-derived CD14-positive monocytes were studied using RNA sequencing. CD14-positive monocytes from six FOP patients and six age- and sex-matched healthy controls were differentiated into osteoclasts in the absence or presence of Activin-A. RNA samples were isolated after 14 days of culturing and analyzed by RNA sequencing. Non-supervised principal component analysis (PCA) showed that samples from the same culture conditions (e.g., without or with Activin-A) tended to cluster, indicating that the variability induced by Activin-A treatment was larger than the variability between the control and FOP samples. RNA sequencing analysis revealed 1480 differentially expressed genes induced by Activin-A in healthy control and FOP osteoclasts with p(adj) < 0.01 and a Log2 fold change of ≥±2. Pathway and gene ontology enrichment analysis revealed several significantly enriched pathways for genes upregulated by Activin-A that could be linked to the differentiation or function of osteoclasts, cell fusion or inflammation. Our data showed that Activin-A has a substantial effect on gene expression during osteoclast formation and that this effect occurred regardless of the presence of the mutated ACVR1 receptor causing FOP.
Collapse
Affiliation(s)
- Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
| | - Joy Zwaak
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
| | - Bruno G. Loos
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
| | - Richard Volckmann
- Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jan Koster
- Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - E. Marelise W. Eekhoff
- Department of Internal Medicine Section Endocrinology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Rare Bone Disease Center Amsterdam, Bone Center, 1081 HV Amsterdam, The Netherlands
| | - Teun J. de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
| |
Collapse
|
22
|
Liu H, Hallauer Hastings M, Kitchen R, Xiao C, Baldovino Guerra JR, Kuznetsov A, Rosenzweig A. Beneficial Effects of Moderate Hepatic Activin A Expression on Metabolic Pathways, Inflammation, and Atherosclerosis. Arterioscler Thromb Vasc Biol 2023; 43:330-349. [PMID: 36453275 DOI: 10.1161/atvbaha.122.318138] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Atherosclerosis is an inflammatory vascular disease marked by hyperlipidemia and hematopoietic stem cell expansion. Activin A, a member of the Activin/GDF/TGFβ/BMP (growth/differentiation factor/transforming growth factor beta/bone morphogenetic protein) family is broadly expressed and increases in human atherosclerosis, but its functional effects in vivo in this context remain unclear. METHODS We studied LDLR-/- mice on a Western diet for 12 weeks and used adeno-associated viral vectors with a liver-specific TBG (thyroxine-binding globulin) promoter to express Activin A or GFP (control). Atherosclerotic lesions were analyzed by oil red staining. Blood lipid profiling was performed by high-performance liquid chromatography, and immune cells were evaluated by flow cytometry. Liver RNA-sequencing was performed to explore the underlying mechanisms. RESULTS Activin A expression decreased in both livers and aortae from LDLR-/- mice fed a Western diet compared with standard laboratory diet. Adenoassociated virus-TBG-Activin A increased Activin A hepatic expression ≈10-fold at 12 weeks; P<0.001) and circulating Activin A levels ≈2000 pg/ml versus ≈50 pg/ml; P<0.001, compared with controls). Hepatic Activin A expression decreased plasma total and LDL (low-density lipoprotein) cholesterol ≈60% and ≈40%, respectively), reduced inflammatory cells in aortae and proliferating hematopoietic stem cells in bone marrow, and reduced atherosclerotic lesion and necrotic core area in aortae. Activin A also attenuated liver steatosis and expression of the lipogenesis genes, Srebp1 and Srebp2. RNA sequencing revealed Activin A not only blocked expression of genes involved in hepatic de novo lipogenesis but also fatty acid uptake and liver inflammation. In addition, Activin A expressed in the liver also reduced white fat tissue accumulation, decreased adipocyte size, and improved glucose tolerance. CONCLUSIONS Our studies reveal hepatic Activin A expression reduces inflammation, hematopoietic stem cell expansion, liver steatosis, circulating cholesterol, and fat accumulation, which likely all contribute to the observed protection against atherosclerosis. The reduced Activin A observed in LDLR-/- mice on a Western diet seems maladaptive and deleterious for atherogenesis.
Collapse
Affiliation(s)
- Huan Liu
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | | | - Robert Kitchen
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | - Chunyang Xiao
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | | | - Alexandra Kuznetsov
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | - Anthony Rosenzweig
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| |
Collapse
|
23
|
Quist-Løkken I, Andersson-Rusch C, Kastnes MH, Kolos JM, Jatzlau J, Hella H, Olsen OE, Sundan A, Knaus P, Hausch F, Holien T. FKBP12 is a major regulator of ALK2 activity in multiple myeloma cells. Cell Commun Signal 2023; 21:25. [PMID: 36717825 PMCID: PMC9885706 DOI: 10.1186/s12964-022-01033-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/28/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The immunophilin FKBP12 binds to TGF-β family type I receptors, including the BMP type I receptor ALK2. FKBP12 keeps the type I receptor in an inactive state and controls signaling activity. Removal of FKBP12 with drugs such as the FKBP-ligand FK506 enhances BMP activity in various cell types. In multiple myeloma cells, activation of SMAD1/5/8 leads to apoptosis. We hypothesized that removing FKBP12 from ALK2 in myeloma cells would potentiate BMP-induced ALK2-SMAD1/5/8 activity and in consequence cell death. METHODS Multiple myeloma cell lines were treated with FK506, or other FKBP-binding compounds, combined with different BMPs before analyzing SMAD1/5/8 activity and cell viability. SMAD1/5/8 activity was also investigated using a reporter cell line, INA-6 BRE-luc. To characterize the functional signaling receptor complex, we genetically manipulated receptor expression by siRNA, shRNA and CRISPR/Cas9 technology. RESULTS FK506 potentiated BMP-induced SMAD1/5/8 activation and apoptosis in multiple myeloma cell lines. By using FKBP-binding compounds with different affinity profiles, and siRNA targeting FKBP12, we show that the FK506 effect is mediated by binding to FKBP12. Ligands that typically signal via ALK3 in myeloma cells, BMP2, BMP4, and BMP10, did not induce apoptosis in cells lacking ALK3. Notably, BMP10 competed with BMP6 and BMP9 and antagonized their activity via ALK2. However, upon addition of FK506, we saw a surprising shift in specificity, as the ALK3 ligands gained the ability to signal via ALK2 and induce apoptosis. This indicates that the receptor complex can switch from an inactive non-signaling complex (NSC) to an active one by adding FK506. This gain of activity was also seen in other cell types, indicating that the observed effects have broader relevance. BMP2, BMP4 and BMP10 depended on BMPR2 as type II receptor to signal, which contrasts with BMP6 and BMP9, that activate ALK2 more potently when BMPR2 is knocked down. CONCLUSIONS In summary, our data suggest that FKBP12 is a major regulator of ALK2 activity in multiple myeloma cells, partly by switching an NSC into an active signaling complex. FKBP12 targeting compounds devoid of immunosuppressing activity could have potential in novel treatment strategies aiming at reducing multiple myeloma tumor load. Video Abstract.
Collapse
Affiliation(s)
- Ingrid Quist-Løkken
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Clara Andersson-Rusch
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Martin Haugrud Kastnes
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Centre of Molecular Inflammation Research, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Jürgen Markus Kolos
- grid.6546.10000 0001 0940 1669Department of Chemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jerome Jatzlau
- grid.14095.390000 0000 9116 4836Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Hanne Hella
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Oddrun Elise Olsen
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| | - Anders Sundan
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Centre of Molecular Inflammation Research, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Hematology, St. Olav’s University Hospital, Trondheim, Norway
| | - Petra Knaus
- grid.14095.390000 0000 9116 4836Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Felix Hausch
- grid.6546.10000 0001 0940 1669Department of Chemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Toril Holien
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology - NTNU, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Immunology and Transfusion Medicine, St. Olav’s University Hospital, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Hematology, St. Olav’s University Hospital, Trondheim, Norway ,grid.5947.f0000 0001 1516 2393Department of Biomedical Laboratory Science, Norwegian University of Science and Technology - NTNU, Trondheim, Norway
| |
Collapse
|
24
|
Minato Y, Nakano-Doi A, Maeda S, Nakagomi T, Yagi H. A Bone Morphogenetic Protein Signaling Inhibitor, LDN193189, Converts Ischemia-Induced Multipotent Stem Cells into Neural Stem/Progenitor Cell-Like Cells. Stem Cells Dev 2022; 31:756-765. [PMID: 36053672 DOI: 10.1089/scd.2022.0139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stem cell therapy is used to restore neurological function in stroke patients. We have previously reported that ischemia-induced multipotent stem cells (iSCs), which are likely derived from brain pericytes, develop in poststroke human and mouse brains. Although we have demonstrated that iSCs can differentiate into neural lineage cells, the factors responsible for inducing this differentiation remain unclear. In this study, we found that LDN193189, a bone morphogenetic protein (BMP) inhibitor, caused irreversible changes in the shape of iSCs. In addition, compared with iSCs incubated without LDN193189, the iSCs incubated with LDN193189 (LDN-iSCs) showed upregulated expression of neural lineage-related genes and proteins, including those expressed in neural stem/progenitor cells (NSPCs), and downregulated expression of mesenchymal and pericytic-related genes and proteins. Moreover, microarray analysis revealed that LDN-iSCs and NSPCs had similar gene expression profiles. Furthermore, LDN-iSCs differentiated into electrophysiologically functional neurons. These results indicate that LDN193189 induces NSPC-like cells from iSCs, suggesting that bioactive molecules regulating BMP signaling are potential targets for promoting neurogenesis from iSCs in the pathological brain, such as during ischemic stroke. We believe that our findings will bring us one step closer to the clinical application of iSCs.
Collapse
Affiliation(s)
- Yusuke Minato
- Department of Anatomy and Cell Biology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Japan.,Department of Therapeutic Progress in Brain Diseases, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Seishi Maeda
- Department of Anatomy and Cell Biology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Japan.,Department of Therapeutic Progress in Brain Diseases, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Hideshi Yagi
- Department of Anatomy and Cell Biology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Japan
| |
Collapse
|
25
|
MacDonnell S, Megna J, Ruan Q, Zhu O, Halasz G, Jasewicz D, Powers K, E H, del Pilar Molina-Portela M, Jin X, Zhang D, Torello J, Feric NT, Graziano MP, Shekhar A, Dunn ME, Glass D, Morton L. Activin A directly impairs human cardiomyocyte contractile function indicating a potential role in heart failure development. Front Cardiovasc Med 2022; 9:1038114. [PMID: 36440002 PMCID: PMC9685658 DOI: 10.3389/fcvm.2022.1038114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/10/2022] [Indexed: 09/27/2023] Open
Abstract
Activin A has been linked to cardiac dysfunction in aging and disease, with elevated circulating levels found in patients with hypertension, atherosclerosis, and heart failure. Here, we investigated whether Activin A directly impairs cardiomyocyte (CM) contractile function and kinetics utilizing cell, tissue, and animal models. Hydrodynamic gene delivery-mediated overexpression of Activin A in wild-type mice was sufficient to impair cardiac function, and resulted in increased cardiac stress markers (N-terminal pro-atrial natriuretic peptide) and cardiac atrophy. In human-induced pluripotent stem cell-derived (hiPSC) CMs, Activin A caused increased phosphorylation of SMAD2/3 and significantly upregulated SERPINE1 and FSTL3 (markers of SMAD2/3 activation and activin signaling, respectively). Activin A signaling in hiPSC-CMs resulted in impaired contractility, prolonged relaxation kinetics, and spontaneous beating in a dose-dependent manner. To identify the cardiac cellular source of Activin A, inflammatory cytokines were applied to human cardiac fibroblasts. Interleukin -1β induced a strong upregulation of Activin A. Mechanistically, we observed that Activin A-treated hiPSC-CMs exhibited impaired diastolic calcium handling with reduced expression of calcium regulatory genes (SERCA2, RYR2, CACNB2). Importantly, when Activin A was inhibited with an anti-Activin A antibody, maladaptive calcium handling and CM contractile dysfunction were abrogated. Therefore, inflammatory cytokines may play a key role by acting on cardiac fibroblasts, causing local upregulation of Activin A that directly acts on CMs to impair contractility. These findings demonstrate that Activin A acts directly on CMs, which may contribute to the cardiac dysfunction seen in aging populations and in patients with heart failure.
Collapse
Affiliation(s)
| | - Jake Megna
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Qin Ruan
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Olivia Zhu
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Gabor Halasz
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Dan Jasewicz
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Kristi Powers
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Hock E
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | | | - Ximei Jin
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Dongqin Zhang
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | | | - Nicole T. Feric
- TARA Biosystems Inc., Alexandria Center for Life Sciences, New York, NY, United States
| | - Michael P. Graziano
- TARA Biosystems Inc., Alexandria Center for Life Sciences, New York, NY, United States
| | | | | | - David Glass
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Lori Morton
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| |
Collapse
|
26
|
Yamamoto M, Stoessel SJ, Yamamoto S, Goldhamer DJ. Overexpression of Wild-Type ACVR1 in Fibrodysplasia Ossificans Progressiva Mice Rescues Perinatal Lethality and Inhibits Heterotopic Ossification. J Bone Miner Res 2022; 37:2077-2093. [PMID: 35637634 PMCID: PMC9708949 DOI: 10.1002/jbmr.4617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/22/2022] [Accepted: 05/28/2022] [Indexed: 11/07/2022]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a devastating disease of progressive heterotopic bone formation for which effective treatments are currently unavailable. FOP is caused by dominant gain-of-function mutations in the receptor ACVR1 (also known as ALK2), which render the receptor inappropriately responsive to activin ligands. In previous studies, we developed a genetic mouse model of FOP that recapitulates most clinical aspects of the disease. In this model, genetic loss of the wild-type Acvr1 allele profoundly exacerbated heterotopic ossification, suggesting the hypothesis that the stoichiometry of wild-type and mutant receptors dictates disease severity. Here, we tested this model by producing FOP mice that conditionally overexpress human wild-type ACVR1. Injury-induced heterotopic ossification (HO) was completely blocked in FOP mice when expression of both the mutant and wild-type receptor were targeted to Tie2-positive cells, which includes fibro/adipogenic progenitors (FAPs). Perinatal lethality of Acvr1R206H/+ mice was rescued by constitutive ACVR1 overexpression, and these mice survived to adulthood at predicted Mendelian frequencies. Constitutive overexpression of ACVR1 also provided protection from spontaneous abnormal skeletogenesis, and the incidence and severity of injury-induced HO in these mice was dramatically reduced. Analysis of pSMAD1/5/8 signaling both in cultured cells and in vivo indicates that ACVR1 overexpression functions cell-autonomously by reducing osteogenic signaling in response to activin A. We propose that ACVR1 overexpression inhibits HO by decreasing the abundance of ACVR1(R206H)-containing signaling complexes at the cell surface while increasing the representation of activin-A-bound non-signaling complexes comprised of wild-type ACVR1. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Masakazu Yamamoto
- Department of Molecular and Cell BiologyUniversity of Connecticut Stem Cell Institute, University of ConnecticutStorrsCTUSA
| | - Sean J Stoessel
- Department of Molecular and Cell BiologyUniversity of Connecticut Stem Cell Institute, University of ConnecticutStorrsCTUSA
| | - Shoko Yamamoto
- Department of Molecular and Cell BiologyUniversity of Connecticut Stem Cell Institute, University of ConnecticutStorrsCTUSA
| | - David J Goldhamer
- Department of Molecular and Cell BiologyUniversity of Connecticut Stem Cell Institute, University of ConnecticutStorrsCTUSA
| |
Collapse
|
27
|
Ruan Q, Lin X, Wang L, Wang N, Zhao Y, Wang H, Tian FY, Hu N, Li Y, Zhao B. An engineered (CAGA)12-EGFP cell-based biosensor for high-content and accurate detection of active TGF-β. Biosens Bioelectron 2022; 220:114884. [DOI: 10.1016/j.bios.2022.114884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/10/2022]
|
28
|
Forlino A. Shedding Light on Bone Morphogenetic Protein (BMP) Signaling Modifiers to Modulate Fibrodysplasia Ossificans Progressiva Severity. J Bone Miner Res 2022; 37:2055-2057. [PMID: 36368771 DOI: 10.1002/jbmr.4730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 11/13/2022]
Affiliation(s)
- Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| |
Collapse
|
29
|
Yang YS, Kim JM, Xie J, Chaugule S, Lin C, Ma H, Hsiao E, Hong J, Chun H, Shore EM, Kaplan FS, Gao G, Shim JH. Suppression of heterotopic ossification in fibrodysplasia ossificans progressiva using AAV gene delivery. Nat Commun 2022; 13:6175. [PMID: 36258013 PMCID: PMC9579182 DOI: 10.1038/s41467-022-33956-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
Heterotopic ossification is the most disabling feature of fibrodysplasia ossificans progressiva, an ultra-rare genetic disorder for which there is currently no prevention or treatment. Most patients with this disease harbor a heterozygous activating mutation (c.617 G > A;p.R206H) in ACVR1. Here, we identify recombinant AAV9 as the most effective serotype for transduction of the major cells-of-origin of heterotopic ossification. We use AAV9 delivery for gene replacement by expression of codon-optimized human ACVR1, ACVR1R206H allele-specific silencing by AAV-compatible artificial miRNA and a combination of gene replacement and silencing. In mouse skeletal cells harboring a conditional knock-in allele of human mutant ACVR1 and in patient-derived induced pluripotent stem cells, AAV gene therapy ablated aberrant Activin A signaling and chondrogenic and osteogenic differentiation. In Acvr1(R206H) knock-in mice treated locally in early adulthood or systemically at birth, trauma-induced endochondral bone formation was markedly reduced, while inflammation and fibroproliferative responses remained largely intact in the injured muscle. Remarkably, spontaneous heterotopic ossification also substantially decreased in in Acvr1(R206H) knock-in mice treated systemically at birth or in early adulthood. Collectively, we develop promising gene therapeutics that can prevent disabling heterotopic ossification in mice, supporting clinical translation to patients with fibrodysplasia ossificans progressiva.
Collapse
Affiliation(s)
- Yeon-Suk Yang
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA
| | - Jung-Min Kim
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA
| | - Jun Xie
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Viral Vector Core, UMass Chan Medical School, Worcester, MA, USA
| | - Sachin Chaugule
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA
| | - Chujiao Lin
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA
| | - Hong Ma
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Viral Vector Core, UMass Chan Medical School, Worcester, MA, USA
| | - Edward Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine; the Institute for Human Genetics; the Program in Craniofacial Biology; and the Eli and Edyth Broad Institute of Regeneration Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Jaehyoung Hong
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hyonho Chun
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eileen M Shore
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- The Center for Research in FOP and Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Frederick S Kaplan
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- The Center for Research in FOP and Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA.
- Viral Vector Core, UMass Chan Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA.
| | - Jae-Hyuck Shim
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA.
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
30
|
Nagar G, Mittal P, Gupta SRR, Pahuja M, Sanger M, Mishra R, Singh A, Singh IK. Multi-omics therapeutic perspective on ACVR1 gene: from genetic alterations to potential targeting. Brief Funct Genomics 2022; 22:123-142. [PMID: 36003055 DOI: 10.1093/bfgp/elac026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/04/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Activin A receptor type I (ACVR1), a transmembrane serine/threonine kinase, belongs to the transforming growth factor-β superfamily, which signals via phosphorylating the downstream effectors and SMAD transcription factors. Its central role in several biological processes and intracellular signaling is well known. Genetic variation in ACVR1 has been associated with a rare disease, fibrodysplasia ossificans progressive, and its somatic alteration is reported in rare cancer diffuse intrinsic pontine glioma. Furthermore, altered expression or variation of ACVR1 is associated with multiple pathologies such as polycystic ovary syndrome, congenital heart defects, diffuse idiopathic skeletal hyperostosis, posterior fossa ependymoma and other malignancies. Recent advancements have witnessed ACVR1 as a potential pharmacological target, and divergent promising approaches for its therapeutic targeting have been explored. This review highlights the structural and functional characteristics of receptor ACVR1, associated signaling pathways, genetic variants in several diseases and cancers, protein-protein interaction, gene expression, regulatory miRNA prediction and potential therapeutic targeting approaches. The comprehensive knowledge will offer new horizons and insights into future strategies harnessing its therapeutic potential.
Collapse
|
31
|
Hwang CD, Pagani CA, Nunez JH, Cherief M, Qin Q, Gomez-Salazar M, Kadaikal B, Kang H, Chowdary AR, Patel N, James AW, Levi B. Contemporary perspectives on heterotopic ossification. JCI Insight 2022; 7:158996. [PMID: 35866484 PMCID: PMC9431693 DOI: 10.1172/jci.insight.158996] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Heterotopic ossification (HO) is the formation of ectopic bone that is primarily genetically driven (fibrodysplasia ossificans progressiva [FOP]) or acquired in the setting of trauma (tHO). HO has undergone intense investigation, especially over the last 50 years, as awareness has increased around improving clinical technologies and incidence, such as with ongoing wartime conflicts. Current treatments for tHO and FOP remain prophylactic and include NSAIDs and glucocorticoids, respectively, whereas other proposed therapeutic modalities exhibit prohibitive risk profiles. Contemporary studies have elucidated mechanisms behind tHO and FOP and have described new distinct niches independent of inflammation that regulate ectopic bone formation. These investigations have propagated a paradigm shift in the approach to treatment and management of a historically difficult surgical problem, with ongoing clinical trials and promising new targets.
Collapse
Affiliation(s)
- Charles D Hwang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Massachusetts General Hospital, Harvard University, Boston, Massachusetts, USA
| | - Chase A Pagani
- Department of Surgery, Center for Organogenesis Research and Trauma, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Johanna H Nunez
- Department of Surgery, Center for Organogenesis Research and Trauma, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Balram Kadaikal
- Department of Surgery, Center for Organogenesis Research and Trauma, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Heeseog Kang
- Department of Surgery, Center for Organogenesis Research and Trauma, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ashish R Chowdary
- Department of Surgery, Center for Organogenesis Research and Trauma, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nicole Patel
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Benjamin Levi
- Department of Surgery, Center for Organogenesis Research and Trauma, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
32
|
Pathophysiology and Emerging Molecular Therapeutic Targets in Heterotopic Ossification. Int J Mol Sci 2022; 23:ijms23136983. [PMID: 35805978 PMCID: PMC9266941 DOI: 10.3390/ijms23136983] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 12/23/2022] Open
Abstract
The term heterotopic ossification (HO) describes bone formation in tissues where bone is normally not present. Musculoskeletal trauma induces signalling events that in turn trigger cells, probably of mesenchymal origin, to differentiate into bone. The aetiology of HO includes extremely rare but severe, generalised and fatal monogenic forms of the disease; and as a common complex disorder in response to musculoskeletal, neurological or burn trauma. The resulting bone forms through a combination of endochondral and intramembranous ossification, depending on the aetiology, initiating stimulus and affected tissue. Given the heterogeneity of the disease, many cell types and biological pathways have been studied in efforts to find effective therapeutic strategies for the disorder. Cells of mesenchymal, haematopoietic and neuroectodermal lineages have all been implicated in the pathogenesis of HO, and the emerging dominant signalling pathways are thought to occur through the bone morphogenetic proteins (BMP), mammalian target of rapamycin (mTOR), and retinoic acid receptor pathways. Increased understanding of these disease mechanisms has resulted in the emergence of several novel investigational therapeutic avenues, including palovarotene and other retinoic acid receptor agonists and activin A inhibitors that target both canonical and non-canonical signalling downstream of the BMP type 1 receptor. In this article we aim to illustrate the key cellular and molecular mechanisms involved in the pathogenesis of HO and outline recent advances in emerging molecular therapies to treat and prevent HO that have had early success in the monogenic disease and are currently being explored in the common complex forms of HO.
Collapse
|
33
|
Goebel EJ, Ongaro L, Kappes EC, Vestal K, Belcheva E, Castonguay R, Kumar R, Bernard DJ, Thompson TB. The orphan ligand, activin C, signals through activin receptor-like kinase 7. eLife 2022; 11:78197. [PMID: 35736809 PMCID: PMC9224996 DOI: 10.7554/elife.78197] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 06/09/2022] [Indexed: 12/11/2022] Open
Abstract
Activin ligands are formed from two disulfide-linked inhibin β (Inhβ) subunit chains. They exist as homodimeric proteins, as in the case of activin A (ActA; InhβA/InhβA) or activin C (ActC; InhβC/InhβC), or as heterodimers, as with activin AC (ActAC; InhβA:InhβC). While the biological functions of ActA and activin B (ActB) have been well characterized, little is known about the biological functions of ActC or ActAC. One thought is that the InhβC chain functions to interfere with ActA production by forming less active ActAC heterodimers. Here, we assessed and characterized the signaling capacity of ligands containing the InhβC chain. ActC and ActAC activated SMAD2/3-dependent signaling via the type I receptor, activin receptor-like kinase 7 (ALK7). Relative to ActA and ActB, ActC exhibited lower affinity for the cognate activin type II receptors and was resistant to neutralization by the extracellular antagonist, follistatin. In mature murine adipocytes, which exhibit high ALK7 expression, ActC elicited a SMAD2/3 response similar to ActB, which can also signal via ALK7. Collectively, these results establish that ActC and ActAC are active ligands that exhibit a distinct signaling receptor and antagonist profile compared to other activins.
Collapse
Affiliation(s)
- Erich J Goebel
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, United States
| | - Luisina Ongaro
- Department of Pharmacology and Therapeutics, Centre for Research in Reproduction and Development, McGill University, Montreal, Canada
| | - Emily C Kappes
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, United States
| | - Kylie Vestal
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, United States
| | | | | | | | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, Centre for Research in Reproduction and Development, McGill University, Montreal, Canada
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, United States
| |
Collapse
|
34
|
Collins MT. Twists in the fibrodysplasia ossificans progressiva story challenge and expand our understanding of BMP biology. J Clin Invest 2022; 132:160773. [PMID: 35703179 PMCID: PMC9197510 DOI: 10.1172/jci160773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an ultrarare, debilitating disease in which heterotopic bone is formed in certain soft tissues. A gain-of-function variant in the cytoplasmic domain of the activin A receptor type I (ACVR1) exists in all patients with FOP. Strikingly, these FOP-causing variants imbue a neofunction to ACVR1 — the ability to recognize activin A as an agonist with bone morphogenic protein–like signaling that leads to heterotopic ossification (HO). These findings are supported by the efficacy of anti–activin A antibodies in preventing HO in FOP mice. This surprising story continues in companion papers in this issue of the JCI. Aykul et al. and Lees-Shepard et al. independently found that antibodies against ACVR1, which were being developed as potential therapeutics for FOP, instead caused HO in FOP mice. While this unexpected finding may be the clinical final act for such antibodies, it provides another twist in the unique and evolving FOP story.
Collapse
|
35
|
Horst B, Pradhan S, Chaudhary R, Listik E, Quintero-Macias L, Choi AS, Southard M, Liu Y, Whitaker R, Hempel N, Berchuck A, Nixon AB, Lee NY, Henis YI, Mythreye K. Hypoxia-induced inhibin promotes tumor growth and vascular permeability in ovarian cancers. Commun Biol 2022; 5:536. [PMID: 35654828 PMCID: PMC9163327 DOI: 10.1038/s42003-022-03495-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/17/2022] [Indexed: 12/21/2022] Open
Abstract
Hypoxia, a driver of tumor growth and metastasis, regulates angiogenic pathways that are targets for vessel normalization and ovarian cancer management. However, toxicities and resistance to anti-angiogenics can limit their use making identification of new targets vital. Inhibin, a heteromeric TGFβ ligand, is a contextual regulator of tumor progression acting as an early tumor suppressor, yet also an established biomarker for ovarian cancers. Here, we find that hypoxia increases inhibin levels in ovarian cancer cell lines, xenograft tumors, and patients. Inhibin is regulated primarily through HIF-1, shifting the balance under hypoxia from activins to inhibins. Hypoxia regulated inhibin promotes tumor growth, endothelial cell invasion and permeability. Targeting inhibin in vivo through knockdown and anti-inhibin strategies robustly reduces permeability in vivo and alters the balance of pro and anti-angiogenic mechanisms resulting in vascular normalization. Mechanistically, inhibin regulates permeability by increasing VE-cadherin internalization via ACVRL1 and CD105, a receptor complex that we find to be stabilized directly by inhibin. Our findings demonstrate direct roles for inhibins in vascular normalization via TGF-β receptors providing new insights into the therapeutic significance of inhibins as a strategy to normalize the tumor vasculature in ovarian cancer. Hypoxia increases levels of the heteromeric TGFβ ligand inhibin in ovarian cancer and inhibin promotes tumor growth, endothelial cell invasion and permeability.
Collapse
|
36
|
Type II BMP and activin receptors BMPR2 and ACVR2A share a conserved mode of growth factor recognition. J Biol Chem 2022; 298:102076. [PMID: 35643319 PMCID: PMC9234707 DOI: 10.1016/j.jbc.2022.102076] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
BMPR2 is a type II Transforming Growth Factor (TGF)-β family receptor that is fundamentally associated with pulmonary arterial hypertension (PAH) in humans. BMPR2 shares functional similarities with the type II activin receptors ACVR2A and ACVR2B, as it interacts with an overlapping group of TGF-β family growth factors (GFs). However, how BMPR2 recognizes GFs remains poorly understood. Here, we solved crystal structures of BMPR2 in complex with the GF activin B and of ACVR2A in complex with the related GF activin A. We show that both BMPR2 and ACVR2A bind GFs with nearly identical geometry using a conserved hydrophobic hot spot, while differences in contacting residues are predominantly found in loop areas. Upon further exploration of the GF-binding spectrum of the two receptors, we found that although many GFs bind both receptors, the high-affinity BMPR2 GFs comprise BMP15, BMP10, and Nodal, whereas those of ACVR2A are activin A, activin B, and GDF11. Lastly, we evaluated GF-binding domain BMPR2 variants found in human PAH patients. We demonstrate that mutations within the GF-binding interface resulted in loss of GF binding, while mutations in loop areas allowed BMPR2 to retain the ability to bind cognate GFs with high affinity. In conclusion, the in vitro activities of BMPR2 variants and the crystal structures reported here indicate biochemically relevant complexes that explain how some GF-binding domain variants can lead to PAH.
Collapse
|
37
|
Aykul S, Huang L, Wang L, Das NM, Reisman S, Ray Y, Zhang Q, Rothman NJ, Nannuru KC, Kamat V, Brydges S, Troncone L, Johnsen L, Yu PB, Fazio S, Lees-Shepard J, Schutz K, Murphy AJ, Economides AN, Idone V, Hatsell SJ. ACVR1 antibodies exacerbate heterotopic ossification in fibrodysplasia ossificans progressiva (FOP) by activating FOP-mutant ACVR1. J Clin Invest 2022; 132:153792. [PMID: 35511419 PMCID: PMC9197526 DOI: 10.1172/jci153792] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 04/28/2022] [Indexed: 11/21/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder whose most debilitating pathology is progressive and cumulative heterotopic ossification (HO) of skeletal muscles, ligaments, tendons, and fascia. FOP is caused by mutations in the type I BMP receptor gene ACVR1, which enable ACVR1 to utilize its natural antagonist, activin A, as an agonistic ligand. The physiological relevance of this property is underscored by the fact that HO in FOP is exquisitely dependent on activation of FOP-mutant ACVR1 by activin A, an effect countered by inhibition of anti–activin A via monoclonal antibody treatment. Hence, we surmised that anti-ACVR1 antibodies that block activation of ACVR1 by ligands should also inhibit HO in FOP and provide an additional therapeutic option for this condition. Therefore, we generated anti-ACVR1 monoclonal antibodies that block ACVR1’s activation by its ligands. Surprisingly, in vivo, these anti-ACVR1 antibodies stimulated HO and activated signaling of FOP-mutant ACVR1. This property was restricted to FOP-mutant ACVR1 and resulted from anti-ACVR1 antibody–mediated dimerization of ACVR1. Conversely, wild-type ACVR1 was inhibited by anti-ACVR1 antibodies. These results uncover an additional property of FOP-mutant ACVR1 and indicate that anti-ACVR1 antibodies should not be considered as therapeutics for FOP.
Collapse
Affiliation(s)
- Senem Aykul
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Lily Huang
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Lili Wang
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Nanditha M Das
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Sandra Reisman
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Yonaton Ray
- Department of Therapeutic Proteins, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Qian Zhang
- Department of Aging/Age-Related Disorders, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Nyanza J Rothman
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Kalyan C Nannuru
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Vishal Kamat
- Department of Therapeutic Proteins, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Susannah Brydges
- Velocigene, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Luca Troncone
- Department of Medicine, Harvard Medical School, Boston, United States of America
| | - Laura Johnsen
- Research Program Management, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Paul B Yu
- Department of Medicine, Harvard Medical School, Boston, United States of America
| | - Sergio Fazio
- Research Council, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - John Lees-Shepard
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Kevin Schutz
- Antibody Engineering, Adimab, Lebanon, United States of America
| | - Andrew J Murphy
- Regeneron Pharmaceuticals, Inc., Tarryrtown, United States of America
| | - Aris N Economides
- Genome Engineering Technologies, and Skeletal Diseases, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Vincent Idone
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Sarah J Hatsell
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| |
Collapse
|
38
|
Szilágyi SS, Amsalem-Zafran AR, Shapira KE, Ehrlich M, Henis YI. Competition between type I activin and BMP receptors for binding to ACVR2A regulates signaling to distinct Smad pathways. BMC Biol 2022; 20:50. [PMID: 35177083 PMCID: PMC8855587 DOI: 10.1186/s12915-022-01252-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 02/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background Activins and bone morphogenetic proteins (BMPs) play critical, sometimes opposing roles, in multiple physiological and pathological processes and diseases. They signal to distinct Smad branches; activins signal mainly to Smad2/3, while BMPs activate mainly Smad1/5/8. This gives rise to the possibility that competition between the different type I receptors through which activin and BMP signal for common type II receptors can provide a mechanism for fine-tuning the cellular response to activin/BMP stimuli. Among the transforming growth factor-β superfamily type II receptors, ACVR2A/B are highly promiscuous, due to their ability to interact with different type I receptors (e.g., ALK4 vs. ALK2/3/6) and with their respective ligands [activin A (ActA) vs. BMP9/2]. However, studies on complex formation between these full-length receptors situated at the plasma membrane, and especially on the potential competition between the different activin and BMP type I receptors for a common activin type II receptor, were lacking. Results We employed a combination of IgG-mediated patching-immobilization of several type I receptors in the absence or presence of ligands with fluorescence recovery after photobleaching (FRAP) measurements on the lateral diffusion of an activin type II receptor, ACVR2A, to demonstrate the principle of competition between type I receptors for ACVR2. Our results show that ACVR2A can form stable heteromeric complexes with ALK4 (an activin type I receptor), as well as with several BMP type I receptors (ALK2/3/6). Of note, ALK4 and the BMP type I receptors competed for binding ACVR2A. To assess the implications of this competition for signaling output, we first validated that in our cell model system (U2OS cells), ACVR2/ALK4 transduce ActA signaling to Smad2/3, while BMP9 signaling to Smad1/5/8 employ ACVR2/ALK2 or ACVR2/ALK3. By combining ligand stimulation with overexpression of a competing type I receptor, we showed that differential complex formation of distinct type I receptors with a common type II receptor balances the signaling to the two Smad branches. Conclusions Different type I receptors that signal to distinct Smad pathways (Smad2/3 vs. Smad1/5/8) compete for binding to common activin type II receptors. This provides a novel mechanism to balance signaling between Smad2/3 and Smad1/5/8. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01252-z.
Collapse
Affiliation(s)
- Szabina Szófia Szilágyi
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Ayelet R Amsalem-Zafran
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Keren E Shapira
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
39
|
Goebel EJ, Kattamuri C, Gipson GR, Krishnan L, Chavez M, Czepnik M, Maguire MC, Grenha R, Håkansson M, Logan DT, Grinberg AV, Sako D, Castonguay R, Kumar R, Thompson TB. Structures of activin ligand traps using natural sets of type I and type II TGFβ receptors. iScience 2022; 25:103590. [PMID: 35005539 PMCID: PMC8718839 DOI: 10.1016/j.isci.2021.103590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/18/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
The 30+ unique ligands of the TGFβ family signal by forming complexes using different combinations of type I and type II receptors. Therapeutically, the extracellular domain of a single receptor fused to an Fc molecule can effectively neutralize subsets of ligands. Increased ligand specificity can be accomplished by using the extracellular domains of both the type I and type II receptor to mimic the naturally occurring signaling complex. Here, we report the structure of one "type II-type I-Fc" fusion, ActRIIB-Alk4-Fc, in complex with two TGFβ family ligands, ActA, and GDF11, providing a snapshot of this therapeutic platform. The study reveals that extensive contacts are formed by both receptors, replicating the ternary signaling complex, despite the inherent low affinity of Alk4. Our study shows that low-affinity type I interactions support altered ligand specificity and can be visualized at the molecular level using this platform.
Collapse
Affiliation(s)
- Erich J. Goebel
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, 231 Albert Sabin Way ML 0524, Cincinnati, OH 45267, USA
| | - Chandramohan Kattamuri
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, 231 Albert Sabin Way ML 0524, Cincinnati, OH 45267, USA
| | - Gregory R. Gipson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, 231 Albert Sabin Way ML 0524, Cincinnati, OH 45267, USA
| | | | | | - Magdalena Czepnik
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, 231 Albert Sabin Way ML 0524, Cincinnati, OH 45267, USA
| | | | - Rosa Grenha
- Acceleron Pharma, Inc., Cambridge, MA 02139, USA
| | - Maria Håkansson
- SARomics Biostructures AB, Medicon Village, Scheeletorget 1, 223 63, Lund, Sweden
| | - Derek T. Logan
- SARomics Biostructures AB, Medicon Village, Scheeletorget 1, 223 63, Lund, Sweden
| | | | - Dianne Sako
- Acceleron Pharma, Inc., Cambridge, MA 02139, USA
| | | | | | - Thomas B. Thompson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, 231 Albert Sabin Way ML 0524, Cincinnati, OH 45267, USA
| |
Collapse
|
40
|
Song W, Dai WJ, Zhang MH, Wang H, Yang XZ. Comprehensive Analysis of the Expression of TGF- β Signaling Regulators and Prognosis in Human Esophageal Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:1812227. [PMID: 34725559 PMCID: PMC8557076 DOI: 10.1155/2021/1812227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023]
Abstract
More and more evidences show that TGF-β has a crucial role in tumor initiation and development. However, the mechanism of the TGF-β signal regulator in esophageal cancer (EC) is still unclear. Here, we use a variety of bioinformatics methods to analyze the expression and survival data of TGF-β signal regulators in patients with EC. We extracted the expression of the S-TGF-β signal regulator from The Cancer Genome Atlas (TCGA). The cBioPortal database was used to assess the frequency of genetic variation. The TGF-β signal regulator is expressed in EC and normal tissues. The objective is to use the Kaplan-Meier plotter database to investigate the prognostic value of TGF-β signal regulators in cancer patients. The DAVID and clusterProfiler software package were used for functional enrichment analysis. We found that patients with TGF-β signaling mutations have shorter overall survival, disease-free survival, disease-specific survival, platinum overall survival, and platinum-free progression survival. We found that compared with the noncancerous tissues of patients with EC, ZFYVE9, BMPR1B, TGFB3, TGFBRAP1, ACVRL1, TGFBR2, SMAD4, SMAD7, ACVR2A, BMPR1, and SMAD9 were significantly downregulated in tumor tissues, while ACVR1 and Smad1 were significantly upregulated in tumor samples. Univariate survival analysis showed that ACVR1, TGFBR3, TGFBRAP1, BMPR1A, SMAD4, and TGFBR2 were positively correlated with overall survival (OS) prolongation. In addition, TGF-β signal transduction regulators could be divided into two classes. Subclass 1 was involved in regulating cell adhesion, PI3K-Akt signaling, and Rap1 signaling. Subclass 2 was related to regulating angiogenesis and PI3K signaling. In short, all members of TGF-β signal regulators can be used as biomarkers to predict the prognosis of patients with EC.
Collapse
Affiliation(s)
- Wei Song
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, China
| | - Wei-Jie Dai
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, China
| | - Meng-hui Zhang
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, China
| | - Han Wang
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, China
| | - Xiao-Zhong Yang
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an 223300, China
| |
Collapse
|
41
|
Functionally diverse heteromeric traps for ligands of the transforming growth factor-β superfamily. Sci Rep 2021; 11:18341. [PMID: 34526551 PMCID: PMC8443706 DOI: 10.1038/s41598-021-97203-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/18/2021] [Indexed: 01/19/2023] Open
Abstract
Ligands of the transforming growth factor-β (TGF-β) 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-β 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-β ligand sets or be directed therapeutically to human diseases arising from dysregulated TGF-β superfamily signaling.
Collapse
|
42
|
Ramachandran A, Mehić M, Wasim L, Malinova D, Gori I, Blaszczyk BK, Carvalho DM, Shore EM, Jones C, Hyvönen M, Tolar P, Hill CS. Pathogenic ACVR1 R206H activation by Activin A-induced receptor clustering and autophosphorylation. EMBO J 2021; 40:e106317. [PMID: 34003511 PMCID: PMC8280795 DOI: 10.15252/embj.2020106317] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 11/23/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) and diffuse intrinsic pontine glioma (DIPG) are debilitating diseases that share causal mutations in ACVR1, a TGF-β family type I receptor. ACVR1R206H is a frequent mutation in both diseases. Pathogenic signaling via the SMAD1/5 pathway is mediated by Activin A, but how the mutation triggers aberrant signaling is not known. We show that ACVR1 is essential for Activin A-mediated SMAD1/5 phosphorylation and is activated by two distinct mechanisms. Wild-type ACVR1 is activated by the Activin type I receptors, ACVR1B/C. In contrast, ACVR1R206H activation does not require upstream kinases, but is predominantly activated via Activin A-dependent receptor clustering, which induces its auto-activation. We use optogenetics and live-imaging approaches to demonstrate Activin A-induced receptor clustering and show it requires the type II receptors ACVR2A/B. Our data provide molecular mechanistic insight into the pathogenesis of FOP and DIPG by linking the causal activating genetic mutation to disrupted signaling.
Collapse
Affiliation(s)
- Anassuya Ramachandran
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | - Merima Mehić
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| | - Laabiah Wasim
- Immune Receptor Activation LaboratoryThe Francis Crick InstituteLondonUK
| | | | - Ilaria Gori
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| | | | - Diana M Carvalho
- Division of Molecular PathologyThe Institute of Cancer ResearchSuttonUK
| | - Eileen M Shore
- Departments of Orthopaedic Surgery and GeneticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Chris Jones
- Division of Molecular PathologyThe Institute of Cancer ResearchSuttonUK
| | - Marko Hyvönen
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Pavel Tolar
- Immune Receptor Activation LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
Division of Infection and ImmunityInstitute of Immunity and TransplantationUniversity CollegeLondonUK
| | - Caroline S Hill
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
43
|
Accumulated Knowledge of Activin Receptor-Like Kinase 2 (ALK2)/Activin A Receptor, Type 1 (ACVR1) as a Target for Human Disorders. Biomedicines 2021; 9:biomedicines9070736. [PMID: 34206903 PMCID: PMC8301367 DOI: 10.3390/biomedicines9070736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/08/2021] [Accepted: 06/23/2021] [Indexed: 12/17/2022] Open
Abstract
Activin receptor-like kinase 2 (ALK2), also known as Activin A receptor type 1 (ACVR1), is a transmembrane kinase receptor for members of the transforming growth factor-β family. Wild-type ALK2/ACVR1 transduces osteogenic signaling in response to ligand binding. Fifteen years ago, a gain-of-function mutation in the ALK2/ACVR1 gene was detected in patients with the genetic disorder fibro-dysplasia ossificans progressiva, which is characterized by heterotopic ossification in soft tissues. Additional disorders, such as diffuse intrinsic pontin glioma, diffuse idiopathic skeletal hyperostosis, primary focal hyperhidrosis, and congenital heart defects, have also been found to be associated with ALK2/ACVR1. These findings further expand in vitro and in vivo model system research and promote our understanding of the molecular mechanisms of the pathogenesis and development of novel therapeutics and diagnosis for disorders associated with ALK2/ACVR1. Through aggressive efforts, some of the disorders associated with ALK2/ACVR1 will be overcome in the near future.
Collapse
|
44
|
Ventura F, Williams E, Ikeya M, Bullock AN, ten Dijke P, Goumans MJ, Sanchez-Duffhues G. Challenges and Opportunities for Drug Repositioning in Fibrodysplasia Ossificans Progressiva. Biomedicines 2021; 9:biomedicines9020213. [PMID: 33669809 PMCID: PMC7922784 DOI: 10.3390/biomedicines9020213] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 01/05/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an ultrarare congenital disease that progresses through intermittent episodes of bone formation at ectopic sites. FOP patients carry heterozygous gene point mutations in activin A receptor type I ACVR1, encoding the bone morphogenetic protein (BMP) type I serine/threonine kinase receptor ALK2, termed activin receptor-like kinase (ALK)2. The mutant ALK2 displays neofunctional responses to activin, a closely related BMP cytokine that normally inhibits regular bone formation. Moreover, the mutant ALK2 becomes hypersensitive to BMPs. Both these activities contribute to enhanced ALK2 signalling and endochondral bone formation in connective tissue. Being a receptor with an extracellular ligand-binding domain and intrinsic intracellular kinase activity, the mutant ALK2 is a druggable target. Although there is no approved cure for FOP yet, a number of clinical trials have been recently initiated, aiming to identify a safe and effective treatment for FOP. Among other targeted approaches, several repurposed drugs have shown promising results. In this review, we describe the molecular mechanisms underlying ALK2 mutation-induced aberrant signalling and ectopic bone formation. In addition, we recapitulate existing in vitro models to screen for novel compounds with a potential application in FOP. We summarize existing therapeutic alternatives and focus on repositioned drugs in FOP, at preclinical and clinical stages.
Collapse
Affiliation(s)
- Francesc Ventura
- Department de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L’Hospitalet de Llobregat, 08907 Barcelona, Spain;
| | - Eleanor Williams
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK; (E.W.); (A.N.B.)
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan;
| | - Alex N. Bullock
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK; (E.W.); (A.N.B.)
| | - Peter ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands;
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Cardiovascular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands;
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology, Cardiovascular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands;
- Correspondence:
| |
Collapse
|
45
|
Hildebrandt S, Kampfrath B, Fischer K, Hildebrand L, Haupt J, Stachelscheid H, Knaus P. ActivinA Induced SMAD1/5 Signaling in an iPSC Derived EC Model of Fibrodysplasia Ossificans Progressiva (FOP) Can Be Rescued by the Drug Candidate Saracatinib. Stem Cell Rev Rep 2021; 17:1039-1052. [PMID: 33410098 PMCID: PMC8166717 DOI: 10.1007/s12015-020-10103-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/20/2022]
Abstract
Balanced signal transduction is crucial in tissue patterning, particularly in the vasculature. Heterotopic ossification (HO) is tightly linked to vascularization with increased vessel number in hereditary forms of HO, such as Fibrodysplasia ossificans progressiva (FOP). FOP is caused by mutations in the BMP type I receptor ACVR1 leading to aberrant SMAD1/5 signaling in response to ActivinA. Whether observed vascular phenotype in human FOP lesions is connected to aberrant ActivinA signaling is unknown. Blocking of ActivinA prevents HO in FOP mice indicating a central role of the ligand in FOP. Here, we established a new FOP endothelial cell model generated from induced pluripotent stem cells (iECs) to study ActivinA signaling. FOP iECs recapitulate pathogenic ActivinA/SMAD1/5 signaling. Whole transcriptome analysis identified ActivinA mediated activation of the BMP/NOTCH pathway exclusively in FOP iECs, which was rescued to WT transcriptional levels by the drug candidate Saracatinib. We propose that ActivinA causes transcriptional pre-patterning of the FOP endothelium, which might contribute to differential vascularity in FOP lesions compared to non-hereditary HO. ![]()
Collapse
Affiliation(s)
- Susanne Hildebrandt
- Institute of Chemistry/Biochemistry, Thielallee 63, Freie Universität Berlin, 14195, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité, Universitätsmedizin Berlin, Föhrer Str. 15, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Branka Kampfrath
- Institute of Chemistry/Biochemistry, Thielallee 63, Freie Universität Berlin, 14195, Berlin, Germany
| | - Kristin Fischer
- Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- BIH Stem Cell Core Facility, Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| | - Laura Hildebrand
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité, Universitätsmedizin Berlin, Föhrer Str. 15, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Julia Haupt
- Institute of Chemistry/Biochemistry, Thielallee 63, Freie Universität Berlin, 14195, Berlin, Germany
| | - Harald Stachelscheid
- Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- BIH Stem Cell Core Facility, Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| | - Petra Knaus
- Institute of Chemistry/Biochemistry, Thielallee 63, Freie Universität Berlin, 14195, Berlin, Germany.
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité, Universitätsmedizin Berlin, Föhrer Str. 15, 13353, Berlin, Germany.
| |
Collapse
|
46
|
Gipson GR, Goebel EJ, Hart KN, Kappes EC, Kattamuri C, McCoy JC, Thompson TB. Structural perspective of BMP ligands and signaling. Bone 2020; 140:115549. [PMID: 32730927 PMCID: PMC7502536 DOI: 10.1016/j.bone.2020.115549] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022]
Abstract
The Bone Morphogenetic Proteins (BMPs) are the largest class signaling molecules within the greater Transforming Growth Factor Beta (TGFβ) family, and are responsible for a wide array of biological functions, including dorsal-ventral patterning, skeletal development and maintenance, as well as cell homeostasis. As such, dysregulation of BMPs results in a number of diseases, including fibrodysplasia ossificans progressiva (FOP) and pulmonary arterial hypertension (PAH). Therefore, understanding BMP signaling and regulation at the molecular level is essential for targeted therapeutic intervention. This review discusses the recent advances in the structural and biochemical characterization of BMPs, from canonical ligand-receptor interactions to co-receptors and antagonists. This work aims to highlight how BMPs differ from other members of the TGFβ family, and how that information can be used to further advance the field. Lastly, this review discusses several gaps in the current understanding of BMP structures, with the aim that discussion of these gaps will lead to advancements in the field.
Collapse
Affiliation(s)
- Gregory R Gipson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Erich J Goebel
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Kaitlin N Hart
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Emily C Kappes
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Chandramohan Kattamuri
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Jason C McCoy
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA.
| |
Collapse
|
47
|
Martinez-Hackert E, Sundan A, Holien T. Receptor binding competition: A paradigm for regulating TGF-β family action. Cytokine Growth Factor Rev 2020; 57:39-54. [PMID: 33087301 DOI: 10.1016/j.cytogfr.2020.09.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023]
Abstract
The transforming growth factor (TGF)-β family is a group of structurally related, multifunctional growth factors, or ligands that are crucially involved in the development, regulation, and maintenance of animal tissues. In humans, the family counts over 33 members. These secreted ligands typically form multimeric complexes with two type I and two type II receptors to activate one of two distinct signal transduction branches. A striking feature of the family is its promiscuity, i.e., many ligands bind the same receptors and compete with each other for binding to these receptors. Although several explanations for this feature have been considered, its functional significance has remained puzzling. However, several recent reports have promoted the idea that ligand-receptor binding promiscuity and competition are critical features of the TGF-β family that provide an essential regulating function. Namely, they allow a cell to read and process multi-ligand inputs. This capability may be necessary for producing subtle, distinctive, or adaptive responses and, possibly, for facilitating developmental plasticity. Here, we review the molecular basis for ligand competition, with emphasis on molecular structures and binding affinities. We give an overview of methods that were used to establish experimentally ligand competition. Finally, we discuss how the concept of ligand competition may be fundamentally tied to human physiology, disease, and therapy.
Collapse
Affiliation(s)
- Erik Martinez-Hackert
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA.
| | - Anders Sundan
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway; Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Toril Holien
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway; Department of Hematology, St. Olav's University Hospital, 7030, Trondheim, Norway.
| |
Collapse
|
48
|
Aykul S, Corpina RA, Goebel EJ, Cunanan CJ, Dimitriou A, Kim HJ, Zhang Q, Rafique A, Leidich R, Wang X, McClain J, Jimenez J, Nannuru KC, Rothman NJ, Lees-Shepard JB, Martinez-Hackert E, Murphy AJ, Thompson TB, Economides AN, Idone V. Activin A forms a non-signaling complex with ACVR1 and type II Activin/BMP receptors via its finger 2 tip loop. eLife 2020; 9:54582. [PMID: 32515349 PMCID: PMC7326492 DOI: 10.7554/elife.54582] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/08/2020] [Indexed: 01/18/2023] Open
Abstract
Activin A functions in BMP signaling in two ways: it either engages ACVR1B to activate Smad2/3 signaling or binds ACVR1 to form a non-signaling complex (NSC). Although the former property has been studied extensively, the roles of the NSC remain unexplored. The genetic disorder fibrodysplasia ossificans progressiva (FOP) provides a unique window into ACVR1/Activin A signaling because in that disease Activin can either signal through FOP-mutant ACVR1 or form NSCs with wild-type ACVR1. To explore the role of the NSC, we generated 'agonist-only' Activin A muteins that activate ACVR1B but cannot form the NSC with ACVR1. Using one of these muteins, we demonstrate that failure to form the NSC in FOP results in more severe disease pathology. These results provide the first evidence for a biological role for the NSC in vivo and pave the way for further exploration of the NSC's physiological role in corresponding knock-in mice.
Collapse
Affiliation(s)
- Senem Aykul
- Regeneron Pharmaceuticals, Tarrytown, United States
| | | | | | | | | | | | - Qian Zhang
- Regeneron Pharmaceuticals, Tarrytown, United States
| | | | | | - Xin Wang
- Regeneron Pharmaceuticals, Tarrytown, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|