1
|
Kalal AA, Mohapatra S. A Comprehensive Review Exploring the Role of Bone Morphogenetic Proteins [BMP]: Biological Mechanisms. Curr Issues Mol Biol 2025; 47:156. [PMID: 40136410 PMCID: PMC11941256 DOI: 10.3390/cimb47030156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/15/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
Bone morphogenetic proteins (BMPs) belong to the TGF-β family. They perform diverse roles in development, osteogenesis, and vasculogenesis. BMPs have crucial functions in embryonic development and regulate the specialization of various cell types. The dysregulation of BMP activity at various stages in signal transduction is associated with a diverse range of human diseases. It is not surprising that BMPs also have a role in tumor formation and control the progression of cancer through different phases. Nevertheless, their specific roles remain ambiguous and the findings regarding this have been inconsistent. The objective of this review is to highlight the important functions of BMP ligands, receptors, and signaling mediators and the subsequent effects on final cellular responses resulting from these signaling modalities. This review elucidates the dysregulation of BMPs identified in various cancer types, which serves as a predictive sign for favorable results in cancer therapy. Alterations in the BMP pathway can represent a crucial milestone in the genetic and molecular mechanisms that facilitate cancer formation. This review has shown that alterations in certain components of the BMP pathway are evident in various tumor forms, including breast, gastric, colorectal, and myeloma cancer. This review reinforces the conclusion that BMPs exert both beneficial and detrimental effects on cancer biology. Collectively, these findings indicate that BMPs serve multiple functions in cancer; therefore, directing therapeutic efforts to focus on BMP may be a highly effective method for treating several cancers.
Collapse
Affiliation(s)
| | - Satyajit Mohapatra
- SRM Centre for Clinical Trials and Research, SRM Medical College Hospital & Research Centre, SRM Institute of Science and Technology (SRMIST), Kattankulathur 603203, India;
| |
Collapse
|
2
|
Gobbo F, Martelli F, Di Virgilio A, Demaria E, Sarli G, Migliaccio AR. The Variation in the Traits Ameliorated by Inhibitors of JAK1/2, TGF-β, P-Selectin, and CXCR1/CXCR2 in the Gata1low Model Suggests That Myelofibrosis Should Be Treated by These Drugs in Combination. Int J Mol Sci 2024; 25:7703. [PMID: 39062946 PMCID: PMC11277099 DOI: 10.3390/ijms25147703] [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: 05/09/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Studies conducted on animal models have identified several therapeutic targets for myelofibrosis, the most severe of the myeloproliferative neoplasms. Unfortunately, many of the drugs which were effective in pre-clinical settings had modest efficacy when tested in the clinic. This discrepancy suggests that treatment for this disease requires combination therapies. To rationalize possible combinations, the efficacy in the Gata1low model of drugs currently used for these patients (the JAK1/2 inhibitor Ruxolitinib) was compared with that of drugs targeting other abnormalities, such as p27kip1 (Aplidin), TGF-β (SB431542, inhibiting ALK5 downstream to transforming growth factor beta (TGF-β) signaling and TGF-β trap AVID200), P-selectin (RB40.34), and CXCL1 (Reparixin, inhibiting the CXCL1 receptors CXCR1/2). The comparison was carried out by expressing the endpoints, which had either already been published or had been retrospectively obtained for this study, as the fold change of the values in the corresponding vehicles. In this model, only Ruxolitinib was found to decrease spleen size, only Aplidin and SB431542/AVID200 increased platelet counts, and with the exception of AVID200, all the inhibitors reduced fibrosis and microvessel density. The greatest effects were exerted by Reparixin, which also reduced TGF-β content. None of the drugs reduced osteopetrosis. These results suggest that future therapies for myelofibrosis should consider combining JAK1/2 inhibitors with drugs targeting hematopoietic stem cells (p27Kip1) or the pro-inflammatory milieu (TGF-β or CXCL1).
Collapse
Affiliation(s)
- Francesca Gobbo
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University, 40126 Bologna, Italy; (F.G.); (G.S.)
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.M.); (A.D.V.)
| | - Antonio Di Virgilio
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.M.); (A.D.V.)
| | - Elena Demaria
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University, 40126 Bologna, Italy;
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University, 40126 Bologna, Italy; (F.G.); (G.S.)
| | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, 73100 Lecce, Italy
| |
Collapse
|
3
|
Karnik SJ, Nazzal MK, Kacena MA, Bruzzaniti A. Megakaryocyte Secreted Factors Regulate Bone Marrow Niche Cells During Skeletal Homeostasis, Aging, and Disease. Calcif Tissue Int 2023; 113:83-95. [PMID: 37243755 PMCID: PMC11179715 DOI: 10.1007/s00223-023-01095-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/01/2023] [Indexed: 05/29/2023]
Abstract
The bone marrow microenvironment contains a diverse array of cell types under extensive regulatory control and provides for a novel and complex mechanism for bone regulation. Megakaryocytes (MKs) are one such cell type that potentially acts as a master regulator of the bone marrow microenvironment due to its effects on hematopoiesis, osteoblastogenesis, and osteoclastogenesis. While several of these processes are induced/inhibited through MK secreted factors, others are primarily regulated by direct cell-cell contact. Notably, the regulatory effects that MKs exert on these different cell populations has been found to change with aging and disease states. Overall, MKs are a critical component of the bone marrow that should be considered when examining regulation of the skeletal microenvironment. An increased understanding of the role of MKs in these physiological processes may provide insight into novel therapies that can be used to target specific pathways important in hematopoietic and skeletal disorders.
Collapse
Affiliation(s)
- Sonali J Karnik
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Murad K Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| | - Angela Bruzzaniti
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA.
| |
Collapse
|
4
|
Verachi P, Gobbo F, Martelli F, Falchi M, di Virgilio A, Sarli G, Wilke C, Bruederle A, Prahallad A, Arciprete F, Zingariello M, Migliaccio AR. Preclinical studies on the use of a P-selectin-blocking monoclonal antibody to halt progression of myelofibrosis in the Gata1 low mouse model. Exp Hematol 2023; 117:43-61. [PMID: 36191885 PMCID: PMC10450205 DOI: 10.1016/j.exphem.2022.09.004] [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: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 01/10/2023]
Abstract
The bone marrow (BM) and spleen from patients with myelofibrosis (MF), as well as those from the Gata1low mouse model of the disease contain increased number of abnormal megakaryocytes. These cells express high levels of the adhesion receptor P-selectin on their surface, which triggers a pathologic neutrophil emperipolesis, leading to increased bioavailability of transforming growth factor-β (TGF-β) in the microenvironment and disease progression. With age, Gata1low mice develop a phenotype similar to that of patients with MF, which is the most severe of the Philadelphia-negative myeloproliferative neoplasms. We previously demonstrated that Gata1low mice lacking the P-selectin gene do not develop MF. In the current study, we tested the hypothesis that pharmacologic inhibition of P-selectin may normalize the phenotype of Gata1low mice that have already developed MF. To test this hypothesis, we have investigated the phenotype expressed by aged Gata1low mice treated with the antimouse monoclonal antibody RB40.34, alone and also in combination with ruxolitinib. The results indicated that RB40.34 in combination with ruxolitinib normalizes the phenotype of Gata1low mice with limited toxicity by reducing fibrosis and the content of TGF-β and CXCL1 (two drivers of fibrosis in this model) in the BM and spleen and by restoring hematopoiesis in the BM and the architecture of the spleen. In conclusion, we provide preclinical evidence that treatment with an antibody against P-selectin in combination with ruxolitinib may be more effective than ruxolitinib alone to treat MF in patients.
Collapse
Affiliation(s)
- Paola Verachi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Francesca Gobbo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy; Department of Veterinary Medical Sciences, University of Bologna, Italy
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National Center for HIV/AIDS Research, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio di Virgilio
- Center for Animal Experimentation and Well-being, Istituto Superiore di Santà, Rome, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Italy
| | | | | | | | - Francesca Arciprete
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Anna Rita Migliaccio
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy; Altius Institute for Biomedical Sciences, Seattle, WA, USA.
| |
Collapse
|
5
|
Chifotides HT, Bose P, Verstovsek S. Momelotinib: an emerging treatment for myelofibrosis patients with anemia. J Hematol Oncol 2022; 15:7. [PMID: 35045875 PMCID: PMC8772195 DOI: 10.1186/s13045-021-01157-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022] Open
Abstract
The suite of marked anemia benefits that momelotinib has consistently conferred on myelofibrosis (MF) patients stem from its unique inhibitory activity on the BMP6/ACVR1/SMAD and IL-6/JAK/STAT3 pathways, resulting in decreased hepcidin (master iron regulator) expression, higher serum iron and hemoglobin levels, and restored erythropoiesis. Clinical data on momelotinib from the phase 2 and the two phase 3 SIMPLIFY trials consistently demonstrated high rates of sustained transfusion-independence. In a recent phase 2 translational study, 41% of the patients achieved transfusion independence for ≥ 12 weeks. In the phase 3 trials SIMPLIFY-1 and SIMPLIFY-2, 17% more JAK inhibitor-naïve patients and two-fold more JAK inhibitor-treated patients achieved or maintained transfusion independence with momelotinib versus ruxolitinib and best available therapy (89% ruxolitinib), respectively. Anemia is present in approximately a third of MF patients at diagnosis, eventually developing in nearly all patients. The need for red blood cell transfusions is an independent adverse risk factor for both overall survival and leukemic transformation. Presently, FDA-approved medications to address anemia are lacking. Momelotinib is one of the prime candidates to durably address the critical unmet needs of MF patients with moderate/severe anemia. Importantly, momelotinib may have overall survival benefits in frontline and second-line MF patients. MOMENTUM is an international registration-track phase 3 trial further assessing momelotinib’s unique constellation of anemia and other benefits in second-line MF patients; the results of the MOMENTUM trial are keenly awaited and may lead to regulatory approval of momelotinib.
Collapse
|
6
|
Stavnichuk M, Komarova SV. Megakaryocyte-driven changes in bone health: lessons from mouse models of myelofibrosis and related disorders. Am J Physiol Cell Physiol 2021; 322:C177-C184. [PMID: 34910601 DOI: 10.1152/ajpcell.00328.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Over the years, numerous studies demonstrated reciprocal communications between processes of bone marrow hematopoiesis and bone remodeling. Megakaryocytes, rare bone marrow cells responsible for platelet production, were demonstrated to be involved in bone homeostasis. Myelofibrosis, characterized by an increase in pleomorphic megakaryocytes in the bone marrow, commonly leads to the development of osteosclerosis. In vivo, an increase in megakaryocyte number was shown to result in osteosclerosis in GATA-1low, NF-E2-/-, TPOhigh, Mpllf/f;PF4cre, Lnk-/-, Mpig6b-/-, Mpig6bfl/fl;Gp1ba-Cr+/KI, Pt-vWD mouse models. In vitro, megakaryocytes stimulate osteoblast proliferation and have variable effects on osteoclast proliferation and activity through soluble factors and direct cell-cell communications. Intriguingly, new studies revealed that the ability of megakaryocytes to communicate with bone cells is affected by the age and sex of animals. This mini-review summarises changes seen in bone architecture and bone cell function in mouse models with an elevated number of megakaryocytes and the effects megakaryocytes have on osteoblasts and osteoclasts in vitro, and discusses potential molecular players that can mediate these effects.
Collapse
Affiliation(s)
- Mariya Stavnichuk
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Bone marrow microenvironment of MPN cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 34756245 DOI: 10.1016/bs.ircmb.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
In this chapter, we will discuss the current knowledge concerning the alterations of the cellular components in the bone marrow niche in Myeloproliferative Neoplasms (MPNs), highlighting the central role of the megakaryocytes in MPN progression, and the extracellular matrix components characterizing the fibrotic bone marrow.
Collapse
|
8
|
Spampinato M, Giallongo C, Romano A, Longhitano L, La Spina E, Avola R, Scandura G, Dulcamare I, Bramanti V, Di Rosa M, Vicario N, Parenti R, Li Volti G, Tibullo D, Palumbo GA. Focus on Osteosclerotic Progression in Primary Myelofibrosis. Biomolecules 2021. [PMID: 33477816 DOI: 10.3390/biom11010122.pmid:33477816;pmcid:pmc7832894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hematopoietic stem-cell-derived clonal proliferation, leading to bone marrow (BM) fibrosis. Hematopoiesis alterations are closely associated with modifications of the BM microenvironment, characterized by defective interactions between vascular and endosteal niches. As such, neoangiogenesis, megakaryocytes hyperplasia and extensive bone marrow fibrosis, followed by osteosclerosis and bone damage, are the most relevant consequences of PMF. Moreover, bone tissue deposition, together with progressive fibrosis, represents crucial mechanisms of disabilities in patients. Although the underlying mechanisms of bone damage observed in PMF are still unclear, the involvement of cytokines, growth factors and bone marrow microenvironment resident cells have been linked to disease progression. Herein, we focused on the role of megakaryocytes and their alterations, associated with cytokines and chemokines release, in modulating functions of most of the bone marrow cell populations and in creating a complex network where impaired signaling strongly contributes to progression and disabilities.
Collapse
Affiliation(s)
- Mariarita Spampinato
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, 95123 Catania, Italy
| | - Alessandra Romano
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, 95123 Catania, Italy
| | - Lucia Longhitano
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Enrico La Spina
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, 95123 Catania, Italy
| | - Roberto Avola
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Grazia Scandura
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, 95123 Catania, Italy
| | - Ilaria Dulcamare
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, 95123 Catania, Italy
| | - Vincenzo Bramanti
- Division of Clinical Pathology, "Giovanni Paolo II" Hospital-A.S.P. Ragusa, 97100 Ragusa, Italy
| | - Michelino Di Rosa
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Nunzio Vicario
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giovanni Li Volti
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Daniele Tibullo
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giuseppe A Palumbo
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, 95123 Catania, Italy
| |
Collapse
|
9
|
Spampinato M, Giallongo C, Romano A, Longhitano L, La Spina E, Avola R, Scandura G, Dulcamare I, Bramanti V, Di Rosa M, Vicario N, Parenti R, Li Volti G, Tibullo D, Palumbo GA. Focus on Osteosclerotic Progression in Primary Myelofibrosis. Biomolecules 2021; 11:biom11010122. [PMID: 33477816 PMCID: PMC7832894 DOI: 10.3390/biom11010122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/22/2022] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hematopoietic stem-cell-derived clonal proliferation, leading to bone marrow (BM) fibrosis. Hematopoiesis alterations are closely associated with modifications of the BM microenvironment, characterized by defective interactions between vascular and endosteal niches. As such, neoangiogenesis, megakaryocytes hyperplasia and extensive bone marrow fibrosis, followed by osteosclerosis and bone damage, are the most relevant consequences of PMF. Moreover, bone tissue deposition, together with progressive fibrosis, represents crucial mechanisms of disabilities in patients. Although the underlying mechanisms of bone damage observed in PMF are still unclear, the involvement of cytokines, growth factors and bone marrow microenvironment resident cells have been linked to disease progression. Herein, we focused on the role of megakaryocytes and their alterations, associated with cytokines and chemokines release, in modulating functions of most of the bone marrow cell populations and in creating a complex network where impaired signaling strongly contributes to progression and disabilities.
Collapse
Affiliation(s)
- Mariarita Spampinato
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Alessandra Romano
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (A.R.); (E.L.S.); (G.S.); (I.D.)
| | - Lucia Longhitano
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
| | - Enrico La Spina
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (A.R.); (E.L.S.); (G.S.); (I.D.)
| | - Roberto Avola
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
| | - Grazia Scandura
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (A.R.); (E.L.S.); (G.S.); (I.D.)
| | - Ilaria Dulcamare
- Department of General Surgery and Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy; (A.R.); (E.L.S.); (G.S.); (I.D.)
| | - Vincenzo Bramanti
- Division of Clinical Pathology, “Giovanni Paolo II” Hospital–A.S.P. Ragusa, 97100 Ragusa, Italy;
| | - Michelino Di Rosa
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Nunzio Vicario
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (N.V.); (R.P.)
| | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (N.V.); (R.P.)
| | - Giovanni Li Volti
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
- Correspondence: (G.L.V.); (G.A.P.)
| | - Daniele Tibullo
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.S.); (L.L.); (R.A.); (D.T.)
| | - Giuseppe A. Palumbo
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
- Correspondence: (G.L.V.); (G.A.P.)
| |
Collapse
|
10
|
Zingariello M, Martelli F, Verachi P, Bardelli C, Gobbo F, Mazzarini M, Migliaccio AR. Novel targets to cure primary myelofibrosis from studies on Gata1 low mice. IUBMB Life 2019; 72:131-141. [PMID: 31749302 DOI: 10.1002/iub.2198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/24/2019] [Indexed: 01/06/2023]
Abstract
In 2002, we discovered that mice carrying the hypomorphic Gata1low mutation that reduces expression of the transcription factor GATA1 in megakaryocytes (Gata1low mice) develop myelofibrosis, a phenotype that recapitulates the features of primary myelofibrosis (PMF), the most severe of the Philadelphia-negative myeloproliferative neoplasms (MPNs). At that time, this discovery had a great impact on the field because mutations driving the development of PMF had yet to be discovered. Later studies identified that PMF, as the others MPNs, is associated with mutations activating the thrombopoietin/JAK2 axis raising great hope that JAK inhibitors may be effective to treat the disease. Unfortunately, ruxolitinib, the JAK1/2 inhibitor approved by FDA and EMEA for PMF, ameliorates symptoms but does not improve the natural course of the disease, and the cure of PMF is still an unmet clinical need. Although GATA1 is not mutated in PMF, reduced GATA1 content in megakaryocytes as a consequence of ribosomal deficiency is a hallmark of myelofibrosis (both in humans and mouse models) and, in fact, a driving event in the disease. Conversely, mice carrying the hypomorphic Gata1low mutation express an activated TPO/JAK2 pathway and partially respond to JAK inhibitors in a fashion similar to PMF patients (reduction of spleen size but limited improvement of the natural history of the disease). These observations cross-validated Gata1low mice as a bona fide animal model for PMF and prompted the use of this model to identify abnormalities that might be targeted to cure the disease. We will summarize here data generated in Gata1low mice indicating that the TGF-β/P-selectin axis is abnormal in PMF and represents a novel target for its treatment.
Collapse
Affiliation(s)
- Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | | | - Paola Verachi
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Claudio Bardelli
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Francesca Gobbo
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Maria Mazzarini
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy.,Myeloproliferative Neoplasms Research Consortium, New York, New York
| |
Collapse
|
11
|
Guo L, Wang R, Zhang K, Yuan J, Wang J, Wang X, Ma J, Wu C. A PINCH-1-Smurf1 signaling axis mediates mechano-regulation of BMPR2 and stem cell differentiation. J Cell Biol 2019; 218:3773-3794. [PMID: 31578224 PMCID: PMC6829670 DOI: 10.1083/jcb.201902022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/30/2019] [Accepted: 08/08/2019] [Indexed: 12/13/2022] Open
Abstract
Mechanical cues from extracellular matrix exert strong effects on stem cell differentiation. This study finds that a signaling axis consisting of PINCH-1, Smurf1, and BMPR2 senses mechanical signals from extracellular matrix and regulates BMP signaling and mesenchymal stem cell differentiation. Mechano-environment plays multiple critical roles in the control of mesenchymal stem cell (MSC) fate decision, but the underlying signaling mechanisms remain undefined. We report here a signaling axis consisting of PINCH-1, SMAD specific E3 ubiquitin protein ligase 1 (Smurf1), and bone morphogenetic protein type 2 receptor (BMPR2) that links mechano-environment to MSC fate decision. PINCH-1 interacts with Smurf1, which inhibits the latter from interacting with BMPR2 and consequently suppresses BMPR2 degradation, resulting in augmented BMP signaling and MSC osteogenic differentiation (OD). Extracellular matrix (ECM) stiffening increases PINCH-1 level and consequently activates this signaling axis. Depletion of PINCH-1 blocks stiff ECM-induced BMP signaling and OD, whereas overexpression of PINCH-1 overrides signals from soft ECM and promotes OD. Finally, perturbation of either Smurf1 or BMPR2 expression is sufficient to block the effects of PINCH-1 on BMP signaling and MSC fate decision. Our findings delineate a key signaling mechanism through which mechano-environment controls BMPR2 level and MSC fate decision.
Collapse
Affiliation(s)
- Ling Guo
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Rong Wang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Kuo Zhang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jifan Yuan
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jiaxin Wang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xiaoxia Wang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jianfei Ma
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chuanyue Wu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
12
|
Megakaryocyte Contribution to Bone Marrow Fibrosis: many Arrows in the Quiver. Mediterr J Hematol Infect Dis 2018; 10:e2018068. [PMID: 30416700 PMCID: PMC6223581 DOI: 10.4084/mjhid.2018.068] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/23/2018] [Indexed: 01/14/2023] Open
Abstract
In Primary Myelofibrosis (PMF), megakaryocyte dysplasia/hyperplasia determines the release of inflammatory cytokines that, in turn, stimulate stromal cells and induce bone marrow fibrosis. The pathogenic mechanism and the cells responsible for progression to bone marrow fibrosis in PMF are not completely understood. This review article aims to provide an overview of the crucial role of megakaryocytes in myelofibrosis by discussing the role and the altered secretion of megakaryocyte-derived soluble factors, enzymes and extracellular matrices that are known to induce bone marrow fibrosis.
Collapse
|
13
|
Yang N, Park S, Cho MS, Lee M, Hong KS, Mun YC, Seong CM, Huh HJ, Huh J. GATA1 Expression in BCR/ABL1-negative Myeloproliferative Neoplasms. Ann Lab Med 2018; 38:296-305. [PMID: 29611379 PMCID: PMC5895858 DOI: 10.3343/alm.2018.38.4.296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/19/2017] [Accepted: 02/20/2018] [Indexed: 12/21/2022] Open
Abstract
Background This study aimed to determine GATA1 expression levels to better characterize subgroups in BCR/ABL1-negative myeloproliferative neoplasms (MPNs). Methods This study enrolled 49 patients diagnosed as having BCR/ABL1-negative MPN on the basis of the 2016 World Health Organization classification : nine polycythemia vera (PV), 17 essential thrombocythemia (ET), 12 prefibrotic primary myelofibrosis (prePMF), and 11 overt primary myelofibrosis (PMF). Relevant clinical and laboratory data were retrieved from the medical records. The molecular analysis of CALR and MPL mutations and quantification of JAK2 V617F allele burden were performed. GATA1 expression was assessed by an immunohistochemical assay on bone marrow biopsy. GATA1 expression was analyzed serially in 18 patients. Results GATA1 expression decreased significantly in PMF compared with that in other subtypes, while no statistical difference was identified between ET and prePMF. GATA1 expression did not differ according to the mutation profiles or the allele burden of JAK2 V617F, but it decreased significantly in patients with overt fibrosis or leukemic transformation. Conclusions Our results suggest that GATA1 expression is significantly low in PMF and decreases with progressive fibrosis and possibly with leukemic transformation, although our attempt to accurately distinguish between subgroups using GATA1 immunohistochemical approach did not achieve statistical significance. A large patient cohort with long term follow-up is required to evaluate the prognostic value of GATA1 expression.
Collapse
Affiliation(s)
- Naery Yang
- Department of Laboratory Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Sholhui Park
- Department of Laboratory Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Min Sun Cho
- Department of Pathology, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Miae Lee
- Department of Laboratory Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Ki Sook Hong
- Department of Laboratory Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Yeung Chul Mun
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Chu Myong Seong
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Hee Jin Huh
- Department of Laboratory Medicine, Dongguk University Ilsan Hospital, Goyang, Korea.
| | - Jungwon Huh
- Department of Laboratory Medicine, College of Medicine, Ewha Womans University, Seoul, Korea.
| |
Collapse
|
14
|
Profile of fibrosis-related gene transcripts and megakaryocytic changes in the bone marrow of myelodysplastic syndromes with fibrosis. Ann Hematol 2018; 97:2099-2106. [DOI: 10.1007/s00277-018-3411-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/21/2018] [Indexed: 11/26/2022]
|
15
|
Kim MJ, Park SY, Chang HR, Jung EY, Munkhjargal A, Lim JS, Lee MS, Kim Y. Clinical significance linked to functional defects in bone morphogenetic protein type 2 receptor, BMPR2. BMB Rep 2018; 50:308-317. [PMID: 28391780 PMCID: PMC5498141 DOI: 10.5483/bmbrep.2017.50.6.059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Indexed: 12/18/2022] Open
Abstract
Bone morphogenetic protein type 2 receptor (BMPR2) is one of the transforming growth factor-β (TGF-β) superfamily receptors, performing diverse roles during embryonic development, vasculogenesis, and osteogenesis. Human BMPR2 consists of 1,038 amino acids, and contains functionally conserved extracellular, transmembrane, kinase, and C-terminal cytoplasmic domains. Bone morphogenetic proteins (BMPs) engage the tetrameric complex, composed of BMPR2 and its corresponding type 1 receptors, which initiates SMAD proteins-mediated signal transduction leading to the expression of target genes implicated in the development or differentiation of the embryo, organs and bones. In particular, genetic alterations of BMPR2 gene are associated with several clinical disorders, including representative pulmonary arterial hypertension, cancers, and metabolic diseases, thus demonstrating the physiological importance of BMPR2. In this mini review, we summarize recent findings regarding the molecular basis of BMPR2 functions in BMP signaling, and the versatile roles of BMPR2. In addition, various aspects of experimentally validated pathogenic mutations of BMPR2 and the linked human diseases will also be discussed, which are important in clinical settings for diagnostics and treatment.
Collapse
Affiliation(s)
- Myung-Jin Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Seon Young Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Hae Ryung Chang
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Eun Young Jung
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Anudari Munkhjargal
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Jong-Seok Lim
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Myeong-Sok Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Yonghwan Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| |
Collapse
|
16
|
Ceglia I, Dueck AC, Masiello F, Martelli F, He W, Federici G, Petricoin EF, Zeuner A, Iancu-Rubin C, Weinberg R, Hoffman R, Mascarenhas J, Migliaccio AR. Preclinical rationale for TGF-β inhibition as a therapeutic target for the treatment of myelofibrosis. Exp Hematol 2016; 44:1138-1155.e4. [PMID: 27592389 PMCID: PMC5778911 DOI: 10.1016/j.exphem.2016.08.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 08/16/2016] [Accepted: 08/19/2016] [Indexed: 01/20/2023]
Abstract
To assess the role of abnormal transforming growth factor-beta (TGF-β) signaling in the pathogenesis of primary myelofibrosis (PMF), the effects of the TGF-β receptor-1 kinase inhibitor SB431542 on ex vivo expansion of hematopoietic cells in cultures from patients with JAK2V617+-polycythemia vera (PV) or PMF (JAK2V617F+, CALRpQ365f+, or unknown) and from normal sources (adult blood, AB, or cord blood, CB) were compared. In cultures of normal sources, SB431542 significantly increased by 2.5-fold the number of progenitor cells generated by days 1-2 (CD34+) and 6 (colony-forming cells) (CB) and that of precursor cells, mostly immature erythroblasts, by days 14-17 (AB and CB). In cultures of JAK2V617F+-PV, SB431542 increased by twofold the numbers of progenitor cells by day 10 and had no effect on that of precursors cells by days 12-17 (∼fourfold increase in all cases). In contrast, SB431542 had no effect on the number of either progenitor or precursor cells in cultures of JAK2V617F+ and CALR pQ365fs+ PMF. These ontogenetic- and disease-specific effects were associated with variegation in the ability of SB431542 to induce CD34+ cells from AB (increased), CB (decreased), or PV and PMF (unaffected) into cycle and erythroblasts in proliferation (increased for AB and PV and unaffected for CB and PMF). Differences in expansion of erythroblasts from AB, CB, and PV were associated with differences in activation of TGF-β signaling (SHCY317, SMAD2S245/250/255, and SMAD1S/S/SMAD5S/S/SMAD8S/S) detectable in these cells by phosphoproteomic profiling. In conclusion, treatment with TGF-β receptor-1 kinase inhibitors may reactivate normal hematopoiesis in PMF patients, providing a proliferative advantage over the unresponsive malignant clone.
Collapse
Affiliation(s)
- Ilaria Ceglia
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Francesca Masiello
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Fabrizio Martelli
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Wu He
- Flow Cytometry Shared Resource Facility, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Federici
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy; Regina Elena National Cancer Institute, Rome, Italy
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Ann Zeuner
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Camelia Iancu-Rubin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Rita Migliaccio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy.
| |
Collapse
|
17
|
Genome-wide association study of platelet aggregation in African Americans. BMC Genet 2015; 16:58. [PMID: 26024889 PMCID: PMC4448541 DOI: 10.1186/s12863-015-0217-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/13/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND We have previously shown that platelet aggregation has higher heritability in African Americans than European Americans. However, a genome-wide association study (GWAS) of platelet aggregation in African Americans has not been reported. We measured platelet aggregation in response to arachidonic acid, ADP, collagen, or epinephrine by optical aggregometry. The discovery cohort was 825 African Americans from the GeneSTAR study. Two replication cohorts were used: 119 African Americans from the Platelet Genes and Physiology Study and 1221 European Americans from GeneSTAR. Genotyping was conducted with Illumina 1 M arrays. For each cohort, age- and sex-adjusted linear mixed models were used to test for association between each SNP and each phenotype under an additive model. RESULTS Six SNPs were significantly associated with platelet aggregation (P<5×10(-8)) in the discovery sample. Of these, three SNPs in three different loci were confirmed: 1) rs12041331, in PEAR1 (platelet endothelial aggregation receptor 1), replicated in both African and European Americans for collagen- and epinephrine-induced aggregation, and in European Americans for ADP-induced aggregation; 2) rs11202221, in BMPR1A (bone morphogenetic protein receptor type1A), replicated in African Americans for ADP-induced aggregation; and 3) rs6566765 replicated in European Americans for ADP-induced aggregation. The rs11202221 and rs6566765 associations with agonist-induced platelet aggregation are novel. CONCLUSIONS In this first GWAS of agonist-induced platelet aggregation in African Americans, we discovered and replicated, novel associations of two variants with ADP-induced aggregation, and confirmed the association of a PEAR1 variant with multi-agonist-induced aggregation. Further study of these genes may provide novel insights into platelet biology.
Collapse
|
18
|
Krause DS, Scadden DT, Preffer FI. The hematopoietic stem cell niche--home for friend and foe? CYTOMETRY. PART B, CLINICAL CYTOMETRY 2013; 84:7-20. [PMID: 23281119 PMCID: PMC3691061 DOI: 10.1002/cyto.b.21066] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 11/16/2012] [Accepted: 11/21/2012] [Indexed: 12/22/2022]
Abstract
The hematopoietic stem cell (HSC) niche is involved in the maintainance and regulation of quiescence, self-renewal and differentiation of hematopoietic stem cells and the fate of their progeny in mammals dealing with the daily stresses to the hematopoietic system. From the discovery that perturbations of the HSC niche can lead to hematopoietic disorders, we have now arrived at the prospect that the HSC niche may play a role in hematological malignancies and that this HSC niche may be a target for therapy. This review attempts to capture the discoveries of the last few years regarding the normal and malignant hematopoietic stem cell niche and possible ways to target this niche.
Collapse
Affiliation(s)
- Daniela S Krause
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | | | | |
Collapse
|
19
|
Liu H, Zhang R, Chen D, Oyajobi BO, Zhao M. Functional redundancy of type II BMP receptor and type IIB activin receptor in BMP2-induced osteoblast differentiation. J Cell Physiol 2012; 227:952-63. [PMID: 21503889 DOI: 10.1002/jcp.22802] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Signaling pathways for bone morphogenetic proteins (BMPs) are important in osteoblast differentiation. Although the precise function of type I BMP receptors in mediating BMP signaling for osteoblast differentiation and bone formation has been characterized previously, the role of type II BMP receptors in osteoblasts is to be well clarified. In this study, we investigated the role of type II BMP receptor (BMPR-II) and type IIB activin receptor (ActR-IIB) in BMP2-induced osteoblast differentiation. While osteoblastic 2T3 cells expressed BMPR-II and ActR-IIB, loss-of-function studies, using dominant negative receptors and siRNAs, showed that BMPR-II and ActR-IIB compensated each other functionally in mediating BMP2 signaling and BMP2-induced osteoblast differentiation. This was evidenced by two findings. First, unless there was loss of function of both type II receptors, isolated disruption of either BMPR-II or ActR-IIB did not remove BMP2 activity. Second, in cells with loss of function of both receptors, restoration of function of either BMPR-II or ActR-IIB by transfection of the wild-type forms, restored BMP2 activity. These findings suggest a functional redundancy between BMPR-II and ActR-IIB in osteoblast differentiation. Results from experiments to test the effects of transforming growth factor β (TGF-β), activin, and fibroblast growth factor (FGF) on osteoblast proliferation and differentiation suggest that inhibition of receptor signaling by double-blockage of BMPR-II and ActR-IIB is BMP-signaling specific. The observed functional redundancy of type II BMP receptors in osteoblasts is novel information about the BMP signaling pathway essential for initiating osteoblast differentiation.
Collapse
Affiliation(s)
- Hongbin Liu
- Department of Biostatistics & Bioinformatics, Tulane University, New Orleans, Louisiana 70112, USA
| | | | | | | | | |
Collapse
|
20
|
Alanne MH, Siljamäki E, Peltonen S, Väänänen K, Windle JJ, Parada LF, Määttä JA, Peltonen J. Phenotypic characterization of transgenic mice harboring Nf1+/− or Nf1−/− osteoclasts in otherwise Nf1+/+ background. J Cell Biochem 2012; 113:2136-46. [DOI: 10.1002/jcb.24088] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
21
|
Verrucci M, Pancrazzi A, Aracil M, Martelli F, Guglielmelli P, Zingariello M, Ghinassi B, D'Amore E, Jimeno J, Vannucchi AM, Migliaccio AR. CXCR4-independent rescue of the myeloproliferative defect of the Gata1low myelofibrosis mouse model by Aplidin. J Cell Physiol 2010; 225:490-9. [PMID: 20458749 DOI: 10.1002/jcp.22228] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The discovery of JAK2 mutations in Philadelphia-negative myeloproliferative neoplasms has prompted investigators to evaluate mutation-targeted treatments to restore hematopoietic cell functions in these diseases. However, the results of the first clinical trials with JAK2 inhibitors are not as promising as expected, prompting a search for additional drugable targets to treat these disorders. In this paper, we used the hypomorphic Gata1(low) mouse model of primary myelofibrosis (PMF), the most severe of these neoplasms, to test the hypothesis that defective marrow hemopoiesis and development of extramedullary hematopoiesis in myelofibrosis is due to insufficient p27(Kip1) activity and is treatable by Aplidin, a cyclic depsipeptide that activates p27(Kip1) in several cancer cells. Aplidin restored expression of Gata1 and p27(Kip1) in Gata1(low) hematopoietic cells, proliferation of marrow progenitor cells in vitro and maturation of megakaryocytes in vivo (reducing TGF-beta/VEGF levels released in the microenvironment by immature Gata1(low) megakaryocytes). Microvessel density, fibrosis, bone growth, and marrow cellularity were normal in Aplidin-treated mice and extramedullary hematopoiesis did not develop in liver although CXCR4 expression in Gata1(low) progenitor cells remained low. These results indicate that Aplidin effectively alters the natural history of myelofibrosis in Gata1(low) mice and suggest this drug as candidate for clinical evaluation in PMF.
Collapse
Affiliation(s)
- Maria Verrucci
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yu YY, Lieu S, Lu C, Miclau T, Marcucio RS, Colnot C. Immunolocalization of BMPs, BMP antagonists, receptors, and effectors during fracture repair. Bone 2010; 46:841-51. [PMID: 19913648 DOI: 10.1016/j.bone.2009.11.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 11/04/2009] [Accepted: 11/04/2009] [Indexed: 01/20/2023]
Abstract
Bone morphogenetic proteins (BMPs) are potent bone inducers used clinically to enhance fracture repair. BMPs have been shown to be produced in the fracture callus; however, the comparative expression of BMPs and BMP signaling components has only been partially examined at the cellular level. The aim of the present study was to establish a detailed spatiotemporal localization of BMPs and BMP signaling components in mouse models of stabilized and nonstabilized fractures. During healing of nonstabilized fractures, which occurs via endochondral ossification, BMP2, 3, 4, 5, and 8, noggin, BMPRIA, BMPRII, and pSmad 1/5/8 were immunolocalized in the activated periosteum as early as 3 days after fracture. BMP2, 4, 5, 6, 7, and 8 and noggin were also found in isolated inflammatory cells within granulation tissue during the early stages of repair, but not BMP receptors and effectors. During the soft callus phase of repair, all BMPs and BMP signaling components were detected in chondrocytes with various intensities of staining depending on the stage of chondrocyte differentiation and their location in the callus. The strongest staining was observed in hypertrophic chondrocytes with decreased intensity during the hard callus phase of repair. All BMPs and components of the BMP pathway were detected in osteoblasts and osteocytes within new bone, with strongest intensity of immunoreaction reported during the early soft callus phase followed by decreasing intensity during the hard callus phase of repair. Most components of the BMP pathway were also detected in endothelial cells associated with new bone. In stabilized fractures that heal strictly via intramembranous ossification, BMPs and BMP antagonists were detected in isolated inflammatory cells and BMP signaling components were not detectable in osteoblasts or osteocytes within new bone. In conclusion, the BMP signaling pathway is primarily activated during fracture healing via endochondral ossification, suggesting that this pathway may influence the mode of healing during the recruitment of skeletal progenitors.
Collapse
Affiliation(s)
- Yan Yiu Yu
- Department of Orthopaedic Surgery, University of California, San Francisco General Hospital, San Francisco, CA 94110, USA
| | | | | | | | | | | |
Collapse
|
23
|
Marconett CN, Sundar SN, Poindexter KM, Stueve TR, Bjeldanes LF, Firestone GL. Indole-3-carbinol triggers aryl hydrocarbon receptor-dependent estrogen receptor (ER)alpha protein degradation in breast cancer cells disrupting an ERalpha-GATA3 transcriptional cross-regulatory loop. Mol Biol Cell 2010; 21:1166-77. [PMID: 20130088 PMCID: PMC2847521 DOI: 10.1091/mbc.e09-08-0689] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We have established in human breast cancer cells that indole-3-carbinol (I3C), a promising anti-cancer phytochemical from Brassica vegetables, ablates ERα expression by stimulating the Rbx-1 E3 ligase mediated degradation of ERα protein and disruption of a cross-regulatory positive feedback loop involving ERα and the GATA3 transcription factor. Estrogen receptor (ER)α is a critical target of therapeutic strategies to control the proliferation of hormone-dependent breast cancers. Preferred clinical options have significant adverse side effects that can lead to treatment resistance due to the persistence of active estrogen receptors. We have established the cellular mechanism by which indole-3-carbinol (I3C), a promising anticancer phytochemical from Brassica vegetables, ablates ERα expression, and we have uncovered a critical role for the GATA3 transcription factor in this indole-regulated cascade. I3C-dependent activation of the aryl hydrocarbon receptor (AhR) initiates Rbx-1 E3 ligase-mediated ubiquitination and proteasomal degradation of ERα protein. I3C inhibits endogenous binding of ERα with the 3′-enhancer region of GATA3 and disrupts endogenous GATA3 interactions with the ERα promoter, leading to a loss of GATA3 and ERα expression. Ectopic expression of GATA3 has no effect on I3C-induced ERα protein degradation but does prevent I3C inhibition of ERα promoter activity, demonstrating the importance of GATA3 in this I3C-triggered cascade. Our preclinical results implicate I3C as a novel anticancer agent in human cancers that coexpress ERα, GATA3, and AhR, a combination found in a large percentage of breast cancers but not in other critical ERα target tissues essential to patient health.
Collapse
Affiliation(s)
- Crystal N Marconett
- Department of Molecular and Cell Biology, University of California-Berkeley, CA 94720, USA
| | | | | | | | | | | |
Collapse
|
24
|
Martelli F, Verrucci M, Migliaccio G, Zingariello M, Rana RA, Vannucchi AM, Migliaccio AR. Removal of the spleen in mice alters the cytokine expression profile of the marrow micro-environment and increases bone formation. Ann N Y Acad Sci 2009; 1176:77-86. [PMID: 19796235 DOI: 10.1111/j.1749-6632.2009.04968.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Splenectomized mice express progressively increased numbers of platelets in the blood and reduced numbers of megakaryocytes in the marrow with age. The megakaryocytes in the marrow of these animals express reduced levels of Gata1, a transcription factor necessary for their maturation. In addition, the marrow from these animals expresses greater levels of cytokines (TGF-beta, PDGF-alpha, and VEGF) known to be produced at high levels by megakaryocytes expressing reduced levels of Gata1. This high level of cytokine expression is in turn associated with active osteoblast proliferation localized to areas of the femur, where megakaryocytes expressing reduced Gata1 levels are also found. These results confirm the role of megakaryocytes as regulator of bone formation in mice and suggest that a cross-talk between the spleen and marrow may regulate the total numbers of hemopoietic niches present in an animal.
Collapse
Affiliation(s)
- Fabrizio Martelli
- Department of Hematology, Oncology, and Molecular Medicine, Istituto Superiore Sanità, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Gata1 expression driven by the alternative HS2 enhancer in the spleen rescues the hematopoietic failure induced by the hypomorphic Gata1low mutation. Blood 2009; 114:2107-20. [PMID: 19571316 DOI: 10.1182/blood-2009-03-211680] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rigorously defined reconstitution assays developed in recent years have allowed recognition of the delicate relationship that exists between hematopoietic stem cells and their niches. This balance ensures that hematopoiesis occurs in the marrow under steady-state conditions. However, during development, recovery from hematopoietic stress and in myeloproliferative disorders, hematopoiesis occurs in extramedullary sites whose microenvironments are still poorly defined. The hypomorphic Gata1(low) mutation deletes the regulatory sequences of the gene necessary for its expression in hematopoietic cells generated in the marrow. By analyzing the mechanism that rescues hematopoiesis in mice carrying this mutation, we provide evidence that extramedullary microenvironments sustain maturation of stem cells that would be otherwise incapable of maturing in the marrow.
Collapse
|
26
|
Zheng C, Yang R, Han Z, Zhou B, Liang L, Lu M. TPO-independent megakaryocytopoiesis. Crit Rev Oncol Hematol 2008; 65:212-22. [PMID: 18093840 DOI: 10.1016/j.critrevonc.2007.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2006] [Revised: 06/19/2007] [Accepted: 11/08/2007] [Indexed: 12/25/2022] Open
Abstract
Megakaryocytopoiesis is a continuous developmental process of platelet production. In this process, a complex network of hemopoietic growth factors are involved, among which TPO (thrombopoietin) is the most thoroughly investigated regulator of MKs (megakaryocytes). In addition to TPO, other regulators also have non-negligible effects on megakaryocytopoiesis. The majority of their effects are independent of TPO signaling. To date, TPO-independent megakaryocytopoiesis forms a regulatory system that includes four signals and (an) unknown signaling pathway(s). These four pathways are the gp 130 (glycoprotein 130)-dependent signaling pathway, the Notch pathway, NMDA (N-methyl-d-aspartate) receptor-mediated signaling, and the SDF-1 (stromal cell-derived factor-1)/FGF-4 (fibroblast growth factor-4) paradigm. Understanding of the TPO-independent regulatory system is important because the system may offer additional opportunities to understand the developmental process and the mechanisms of disorders characterized by abnormal MK and platelet production, such as thrombocytopenia and thrombocythemia, and to advance the development of therapeutics.
Collapse
Affiliation(s)
- Cuiling Zheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, PR China
| | | | | | | | | | | |
Collapse
|
27
|
Bock O, Höftmann J, Theophile K, Hussein K, Wiese B, Schlué J, Kreipe H. Bone morphogenetic proteins are overexpressed in the bone marrow of primary myelofibrosis and are apparently induced by fibrogenic cytokines. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:951-60. [PMID: 18349123 DOI: 10.2353/ajpath.2008.071030] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasia characterized by progressive deposition of extracellular matrix components in the bone marrow. The involvement of members of the bone morphogenetic protein (BMP) family in aberrant bone marrow matrix homeostasis in PMF has not yet been investigated. Therefore, we analyzed expression of BMP1, an activator of latent transforming growth factor beta-1 (TGFbeta-1) and processor of collagen precursors, and other BMPs in bone marrow from PMF patients and controls (n = 95). Expression of BMP1, BMP6, BMP7, and BMP-receptor 2 was significantly increased in advanced stages of myelofibrosis compared with controls (P < or = 0.01), and enhanced levels of BMP6 expression were already evident in prefibrotic stages of PMF. Immunohistochemistry showed that bone marrow stromal cells and megakaryocytes were the major cellular sources of BMP1 protein. Because TGFbeta-1 and basic fibroblast growth factor have been shown to be important in the development of myelofibrosis, we studied the induction of BMPs by these cytokines in cultured fibroblasts. Fibroblasts treated with TGFbeta-1 showed a pronounced up-regulation of BMP6, suggesting that stromal cells may be susceptible to BMP activation by cytokines with a proven role in the pathogenesis of PMF. We conclude that BMP family members may play an important role in the pathogenesis of myelofibrosis in PMF and are apparently induced by cytokines such as TGFbeta-1.
Collapse
Affiliation(s)
- Oliver Bock
- Assistant Professor, Institute of Pathology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
28
|
Suva LJ, Hartman E, Dilley JD, Russell S, Akel NS, Skinner RA, Hogue WR, Budde U, Varughese KI, Kanaji T, Ware J. Platelet dysfunction and a high bone mass phenotype in a murine model of platelet-type von Willebrand disease. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:430-9. [PMID: 18187573 DOI: 10.2353/ajpath.2008.070417] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The platelet glycoprotein Ib-IX receptor binds surface-bound von Willebrand factor and supports platelet adhesion to damaged vascular surfaces. A limited number of mutations within the glycoprotein Ib-IX complex have been described that permit a structurally altered receptor to interact with soluble von Willebrand factor, and this is the molecular basis of platelet-type von Willebrand disease. We have developed and characterized a mouse model of platelet-type von Willebrand disease (G233V) and have confirmed a platelet phenotype mimicking the human disorder. The mice have a dramatic increase in splenic megakaryocytes and splenomegaly. Recent studies have demonstrated that hematopoetic cells can influence the differentiation of osteogenic cells. Thus, we examined the skeletal phenotype of mice expressing the G233V variant complex. At 6 months of age, G233V mice exhibit a high bone mass phenotype with an approximate doubling of trabecular bone volume in both the tibia and femur. Serum measures of bone resorption were significantly decreased in G233V animals. With decreased bone resorption, cortical thickness was increased, medullary area decreased, and consequently, the mechanical strength of the femur was significantly increased. Using ex vivo bone marrow cultures, osteoclast-specific staining in the G233V mutant marrow was diminished, whereas osteoblastogenesis was unaffected. These studies provide new insights into the relationship between the regulation of megakaryocytopoiesis and bone mass.
Collapse
Affiliation(s)
- Larry J Suva
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Perry MJ, Redding KA, Alexander WS, Tobias JH. Mice rendered severely deficient in megakaryocytes through targeted gene deletion of the thrombopoietin receptor c-Mpl have a normal skeletal phenotype. Calcif Tissue Int 2007; 81:224-31. [PMID: 17674074 DOI: 10.1007/s00223-007-9051-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2007] [Accepted: 06/11/2007] [Indexed: 01/27/2023]
Abstract
To explore whether a functional relationship exists between megakaryocytes and the cellular processes responsible for bone formation, we examined if Mpl ( -/- ) mice, which are severely megakaryocyte-deficient through c-Mpl gene deletion, have an abnormal skeletal phenotype compared to Mpl ( +/- ) and wild-type littermates. We also analyzed whether the osteogenic response to high-dose estrogen treatment is altered in Mpl ( -/- ) mice. Megakaryocyte numbers and skeletal indices were compared between Mpl ( -/- ) mice and littermate Mpl ( +/- ) and wild-type 12-week-old mice (six per group). Dual-energy X-ray absorbtiometry of whole body, excised tibias, and femurs was performed. Histomorphometric analyses of the proximal metaphysis and mid-diaphysis were carried out on longitudinal and transverse sections, respectively. Histomorphometry was performed on the proximal tibial metaphysis of four Mpl ( -/- ) and four wild-type mice following high-dose estrogen treatment (0.5 mg/animal/week) for 4 weeks. Mpl ( -/- ) mice had 10% the megakaryocyte number of Mpl ( +/- ) and wild-type littermates. Bone mineral density values in Mpl ( -/- ) mice were identical to those in Mpl ( +/- ) and wild-type mice for whole body, femur, and tibia. Histomorphometric analysis demonstrated that cancellous and cortical tibial bone parameters were similar across all genotypes. The osteogenic response to estrogen treatment was indistinguishable between Mpl ( -/- )and wild-type mice. We found that mice severely deficient in megakaryocytes have a normal skeletal phenotype. Additionally, the deficiency did not diminish the osteogenic marrow response to high-dose estrogen treatment. These results represent the first in vivo evidence that severe megakaryocyte deficiency does not affect bone formation, suggesting that this process is not dependent on normal megakaryocyte number.
Collapse
Affiliation(s)
- Mark J Perry
- Anatomy and Clinical Sciences North Bristol, University of Bristol, Southwell Street, Bristol, BS2 8EJ, UK.
| | | | | | | |
Collapse
|