1
|
Childress PJ, Nielsen JJ, Bemenderfer TB, Dadwal UC, Chakraborty N, Harris JS, Bethel M, Alvarez MB, Tucker A, Wessel AR, Millikan PD, Wilhite JH, Engle A, Brinker A, Rytlewski JD, Scofield DC, Griffin KS, Shelley WC, Manikowski KJ, Jackson KL, Miller SA, Cheng YH, Ghosh J, Mulcrone PL, Srour EF, Yoder MC, Natoli RM, Shively KD, Gautam A, Hammamieh R, Low SA, Low PS, McKinley TO, Anglen JO, Lowery JW, Chu TMG, Kacena MA. Thrombopoietic agents enhance bone healing in mice, rats, and pigs. J Bone Miner Res 2024; 40:125-139. [PMID: 39566068 DOI: 10.1093/jbmr/zjae191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/15/2024] [Accepted: 11/19/2024] [Indexed: 11/22/2024]
Abstract
Achieving bone union remains a significant clinical dilemma. The use of osteoinductive agents, specifically bone morphogenetic proteins (BMPs), has gained wide attention. However, multiple side effects, including increased incidence of cancer, have renewed interest in investigating alternatives that provide safer, yet effective bone regeneration. Here we demonstrate the robust bone healing capabilities of the main megakaryocyte (MK) growth factor, thrombopoietin (TPO), and second-generation TPO agents using multiple animal models, including mice, rats, and pigs. This bone healing activity is shown in two fracture models (critical-sized defect [CSD] and closed fracture) and with local or systemic administration. Our transcriptomic analyses, cellular studies, and protein arrays demonstrate that TPO enhances multiple cellular processes important to fracture healing, particularly angiogenesis, which is required for bone union. Finally, the therapeutic potential of thrombopoietic agents is high since they are used in the clinic for other indications (eg, thrombocytopenia) with established safety profiles and act upon a narrowly defined population of cells.
Collapse
Affiliation(s)
- Paul J Childress
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, United States
| | - Jeffery J Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, United States
| | - Thomas B Bemenderfer
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Ushashi C Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, United States
| | - Nabarun Chakraborty
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, United States
| | - Jonathan S Harris
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Monique Bethel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Marta B Alvarez
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, United States
| | - Aamir Tucker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Alexander R Wessel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Patrick D Millikan
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Jonathan H Wilhite
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Andrew Engle
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Alexander Brinker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Jeffrey D Rytlewski
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - David C Scofield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Kaitlyn S Griffin
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - W Christopher Shelley
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Kelli J Manikowski
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, 46222, United States
| | - Krista L Jackson
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, 46222, United States
| | - Stacy-Ann Miller
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, United States
| | - Ying-Hua Cheng
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Joydeep Ghosh
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Patrick L Mulcrone
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, United States
| | - Edward F Srour
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Roman M Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Karl D Shively
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Aarti Gautam
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, United States
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, United States
| | - Stewart A Low
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, United States
| | - Philip S Low
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, United States
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Jeffrey O Anglen
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN, 46222, United States
| | - Tien-Min G Chu
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, 46202, United States
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, United States
| |
Collapse
|
2
|
Nazzal MK, Battina HL, Tewari NP, Mostardo SL, Nagaraj RU, Zhou D, Awosanya OD, Majety SK, Samson S, Blosser RJ, Dadwal UC, Mulcrone PL, Kacena MA. The effects of young and aged, male and female megakaryocyte conditioned media on angiogenic properties of endothelial cells. Aging (Albany NY) 2024; 16:13181-13200. [PMID: 39578050 PMCID: PMC11719103 DOI: 10.18632/aging.206077] [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: 09/28/2023] [Accepted: 07/11/2024] [Indexed: 11/24/2024]
Abstract
With aging, the risk of fractures and compromised healing increases. Angiogenesis plays a significant role in bone healing and is impaired with aging. We have previously shown the impact of megakaryocytes (MKs) in regulating bone healing. Notably, MKs produce factors known to promote angiogenesis. We examined the effects of conditioned media (CM) generated from MKs derived from young (3-4-month-old) and aged (22-24-month-old), male and female C57BL/6J mice on bone marrow endothelial cell (BMEC) growth and function. Female MK CM, regardless of age, caused a >65% increase in BMEC proliferation and improved vessel formation by >115%. Likewise, young male MK CM increased vessel formation by 160%. Although aged male MK CM resulted in >150% increases in the formation of vascular nodes and meshes, 62% fewer vessels formed compared to young male MK CM treatment. Aged female MK CM improved migration by over 2500%. However, aged female and male MK CM caused less wound closure. MK CM treatments also significantly altered the expression of several genes including PDGFRβ, CXCR4, and CD36 relative to controls and between ages. Further testing of mechanisms responsible for age-associated differences may allow for novel strategies to improve MK-mediated angiogenesis and bone healing, particularly within the aging population.
Collapse
Affiliation(s)
- Murad K. Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hanisha L. Battina
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nikhil P. Tewari
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sarah L. Mostardo
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| | - Rohit U. Nagaraj
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Donghui Zhou
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Olatundun D. Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Saveda K. Majety
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sue Samson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rachel J. Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| | - Ushashi C. Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| | - Patrick L. Mulcrone
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| | - Melissa A. Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
3
|
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
|
4
|
Mohamad SF, El Koussa R, Ghosh J, Blosser R, Gunawan A, Layer J, Zhang C, Karnik S, Davé U, Kacena MA, Srour EF. Osteomacs promote maintenance of murine hematopoiesis through megakaryocyte-induced upregulation of Embigin and CD166. Stem Cell Reports 2024; 19:486-500. [PMID: 38458190 PMCID: PMC11096441 DOI: 10.1016/j.stemcr.2024.02.004] [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: 08/24/2022] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 03/10/2024] Open
Abstract
Maintenance of hematopoietic stem cell (HSC) function in the niche is an orchestrated event. Osteomacs (OM) are key cellular components of the niche. Previously, we documented that osteoblasts, OM, and megakaryocytes interact to promote hematopoiesis. Here, we further characterize OM and identify megakaryocyte-induced mediators that augment the role of OM in the niche. Single-cell mRNA-seq, mass spectrometry, and CyTOF examination of megakaryocyte-stimulated OM suggested that upregulation of CD166 and Embigin on OM augment their hematopoiesis maintenance function. CD166 knockout OM or shRNA-Embigin knockdown OM confirmed that the loss of these molecules significantly reduced the ability of OM to augment the osteoblast-mediated hematopoietic-enhancing activity. Recombinant CD166 and Embigin partially substituted for OM function, characterizing both proteins as critical mediators of OM hematopoietic function. Our data identify Embigin and CD166 as OM-regulated critical components of HSC function in the niche and potential participants in various in vitro manipulations of stem cells.
Collapse
Affiliation(s)
- Safa F Mohamad
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roy El Koussa
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joydeep Ghosh
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rachel Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Gunawan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Justin Layer
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sonali Karnik
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Utpal Davé
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Edward F Srour
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
5
|
Mohamad SF, Kacena MA. Isolation of Murine Neonatal and Adult Osteomacs to Examine Their Role in the Hematopoietic Niche. Methods Mol Biol 2024. [PMID: 38507212 DOI: 10.1007/7651_2024_535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Maintenance of hematopoietic stem cell (HSC) function is an orchestrated event between multiple cell types, and crosstalk between these cell types is an essential part of HSC regulation. Among the cell groups of the niche involved in this process are a group of bone-resident macrophages known as osteomacs (OM). Previously, it was demonstrated that OM and osteoblasts contained within neonatal calvarial cells are critical to maintain hematopoietic function. Additionally, interactions between neonatal calvarial cells and megakaryocytes further enhance this hematopoietic activity. In this chapter, we explore one such interaction involving OM and osteoblasts in the hematopoietic niche. We describe a protocol to isolate OM from both neonatal and adult mice, and subsequently use colony-forming assays to demonstrate their interaction with osteoblasts in maintaining HSC function.
Collapse
Affiliation(s)
- Safa F Mohamad
- Department of Hematology and Oncology, Boston Children's Hospital/Harvard School of Medicine, Boston, MA, USA.
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
6
|
Andreev D, Kachler K, Liu M, Chen Z, Krishnacoumar B, Ringer M, Frey S, Krönke G, Voehringer D, Schett G, Bozec A. Eosinophils preserve bone homeostasis by inhibiting excessive osteoclast formation and activity via eosinophil peroxidase. Nat Commun 2024; 15:1067. [PMID: 38316791 PMCID: PMC10844633 DOI: 10.1038/s41467-024-45261-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 01/18/2024] [Indexed: 02/07/2024] Open
Abstract
Eosinophils are involved in tissue homeostasis. Herein, we unveiled eosinophils as important regulators of bone homeostasis. Eosinophils are localized in proximity to bone-resorbing osteoclasts in the bone marrow. The absence of eosinophils in ΔdblGATA mice results in lower bone mass under steady-state conditions and amplified bone loss upon sex hormone deprivation and inflammatory arthritis. Conversely, increased numbers of eosinophils in IL-5 transgenic mice enhance bone mass under steady-state conditions and protect from hormone- and inflammation- mediated bone loss. Eosinophils strongly inhibit the differentiation and demineralization activity of osteoclasts and lead to profound changes in the transcriptional profile of osteoclasts. This osteoclast-suppressive effect of eosinophils is based on the release of eosinophil peroxidase causing impaired reactive oxygen species and mitogen-activated protein kinase induction in osteoclast precursors. In humans, the number and the activity of eosinophils correlates with bone mass in healthy participants and rheumatoid arthritis patients. Taken together, experimental and human data indicate a regulatory function of eosinophils on bone.
Collapse
Affiliation(s)
- Darja Andreev
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany.
| | - Katerina Kachler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Mengdan Liu
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Department of Rheumatology, Zhejiang University - School of Medicine, Hangzhou, China
| | - Zhu Chen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Department of Rheumatology and Immunology, Anhui Medical University Affiliated Provincial Hospital, Hefei, China
| | - Brenda Krishnacoumar
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Institute of Physiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mark Ringer
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Silke Frey
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
- Department of Rheumatology and Clinical Immunology, Charité University Medicine, Berlin, Germany
| | - David Voehringer
- Department of Infection Biology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany.
| |
Collapse
|
7
|
Hernández-Barrientos D, Pelayo R, Mayani H. The hematopoietic microenvironment: a network of niches for the development of all blood cell lineages. J Leukoc Biol 2023; 114:404-420. [PMID: 37386890 DOI: 10.1093/jleuko/qiad075] [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: 03/07/2023] [Revised: 05/25/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023] Open
Abstract
Blood cell formation (hematopoiesis) takes place mainly in the bone marrow, within the hematopoietic microenvironment, composed of a number of different cell types and their molecular products that together shape spatially organized and highly specialized microstructures called hematopoietic niches. From the earliest developmental stages and throughout the myeloid and lymphoid lineage differentiation pathways, hematopoietic niches play a crucial role in the preservation of cellular integrity and the regulation of proliferation and differentiation rates. Current evidence suggests that each blood cell lineage develops under specific, discrete niches that support committed progenitor and precursor cells and potentially cooperate with transcriptional programs determining the gradual lineage commitment and specification. This review aims to discuss recent advances on the cellular identity and structural organization of lymphoid, granulocytic, monocytic, megakaryocytic, and erythroid niches throughout the hematopoietic microenvironment and the mechanisms by which they interconnect and regulate viability, maintenance, maturation, and function of the developing blood cells.
Collapse
Affiliation(s)
- Daniel Hernández-Barrientos
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Av. Cuauhtemoc 330. Mexico City, 06720, Mexico
| | - Rosana Pelayo
- Onco-Immunology Laboratory, Eastern Biomedical Research Center, IMSS, Km 4.5 Atlixco-Metepec, 74360, Puebla, Mexico
| | - Hector Mayani
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Av. Cuauhtemoc 330. Mexico City, 06720, Mexico
| |
Collapse
|
8
|
Lai J, Li Y, Ran M, Huang Q, Huang F, Zhu L, Wu Y, Zou W, Xie X, Tang Y, Yang F, Wu A, Ge G, Wu J. Xanthotoxin, a novel inducer of platelet formation, promotes thrombocytopoiesis via IL-1R1 and MEK/ERK signaling. Biomed Pharmacother 2023; 163:114811. [PMID: 37156117 DOI: 10.1016/j.biopha.2023.114811] [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: 03/17/2023] [Revised: 04/20/2023] [Accepted: 04/30/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Thrombocytopenia is a common hematological disease caused by many factors. It usually complicates critical diseases and increases morbidity and mortality. The treatment of thrombocytopenia remains a great challenge in clinical practice, however, its treatment options are limited. In this study, the active monomer xanthotoxin (XAT) was screened out to explore its medicinal value and provide novel therapeutic strategies for the clinical treatment of thrombocytopenia. METHODS The effects of XAT on megakaryocyte differentiation and maturation were detected by flow cytometry, Giemsa and phalloidin staining. RNA-seq identified differentially expressed genes and enriched pathways. The signaling pathway and transcription factors were verified through WB and immunofluorescence staining. Tg (cd41: eGFP) transgenic zebrafish and mice with thrombocytopenia were used to evaluate the biological activity of XAT on platelet formation and the related hematopoietic organ index in vivo. RESULTS XAT promoted the differentiation and maturation of Meg-01 cells in vitro. Meanwhile, XAT could stimulate platelet formation in transgenic zebrafish and recover platelet production and function in irradiation-induced thrombocytopenia mice. Further RNA-seq prediction and WB verification revealed that XAT activates the IL-1R1 target and MEK/ERK signaling pathway, and upregulates the expression of transcription factors related to the hematopoietic lineage to promote megakaryocyte differentiation and platelet formation. CONCLUSION XAT accelerates megakaryocyte differentiation and maturation to promote platelet production and recovery through triggering IL-1R1 and activating the MEK/ERK signaling pathway, providing a new pharmacotherapy strategy for thrombocytopenia.
Collapse
Affiliation(s)
- Jia Lai
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Yueyue Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mei Ran
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qianqian Huang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Linjie Zhu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Yuesong Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wenjun Zou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiang Xie
- School of Basic Medical Sciences, Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Fei Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Anguo Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Kim WR, Park EG, Lee HE, Park SJ, Huh JW, Kim JN, Kim HS. Hsa-miR-422a Originated from Short Interspersed Nuclear Element Increases ARID5B Expression by Collaborating with NF-E2. Mol Cells 2022; 45:465-478. [PMID: 35444070 PMCID: PMC9260135 DOI: 10.14348/molcells.2022.2158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/13/2021] [Accepted: 12/27/2021] [Indexed: 12/03/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate the expression of target messenger RNA (mRNA) complementary to the 3' untranslated region (UTR) at the post-transcriptional level. Hsa-miR-422a, which is commonly known as miRNA derived from transposable element (MDTE), was derived from short interspersed nuclear element (SINE). Through expression analysis, hsa-miR-422a was found to be highly expressed in both the small intestine and liver of crab-eating monkey. AT-Rich Interaction Domain 5 B (ARID5B) was selected as the target gene of hsa-miR-422a, which has two binding sites in both the exon and 3'UTR of ARID5B. To identify the interaction between hsa-miR-422a and ARID5B, a dual luciferase assay was conducted in HepG2 cell line. The luciferase activity of cells treated with the hsa-miR-422a mimic was upregulated and inversely downregulated when both the hsa-miR-422a mimic and inhibitor were administered. Nuclear factor erythroid-2 (NF-E2) was selected as the core transcription factor (TF) via feed forward loop analysis. The luciferase expression was downregulated when both the hsa-miR-422a mimic and siRNA of NF-E2 were treated, compared to the treatment of the hsa-miR-422a mimic alone. The present study suggests that hsa-miR-422a derived from SINE could bind to the exon region as well as the 3'UTR of ARID5B. Additionally, hsa-miR-422a was found to share binding sites in ARID5Bwith several TFs, including NF-E2. The hsa-miR-422a might thus interact with TF to regulate the expression of ARID5B, as demonstrated experimentally. Altogether, hsa-miR-422a acts as a super enhancer miRNA of ARID5Bby collaborating with TF and NF-E2.
Collapse
Affiliation(s)
- Woo Ryung Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
- Institute of Systems Biology, Pusan National University, Busan 46241, Korea
| | - Eun Gyung Park
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
- Institute of Systems Biology, Pusan National University, Busan 46241, Korea
| | - Hee-Eun Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28199, Korea
| | - Sang-Je Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28199, Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28199, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Jeong Nam Kim
- Department of Microbiology, College of Natural Sciences, Pusan National University, Busan 46241, Korea
| | - Heui-Soo Kim
- Institute of Systems Biology, Pusan National University, Busan 46241, Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Korea
| |
Collapse
|
12
|
Karagianni A, Matsuura S, Gerstenfeld LC, Ravid K. Inhibition of Osteoblast Differentiation by JAK2V617F Megakaryocytes Derived From Male Mice With Primary Myelofibrosis. Front Oncol 2022; 12:929498. [PMID: 35880162 PMCID: PMC9307716 DOI: 10.3389/fonc.2022.929498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/09/2022] [Indexed: 11/16/2022] Open
Abstract
Past studies described interactions between normal megakaryocytes, the platelet precursors, and bone cell precursors in the bone marrow. This relationship has also been studied in context of various mutations associated with increased number of megakaryocytes. The current study is the first to examine the effects of megakaryocytes from transgenic mice carrying the most common mutation that causes primary myelofibrosis (PMF) in humans (JAK2V617F) on bone cell differentiation. Organ level assessments of mice using micro-computed tomography showed decreased bone volume in JAK2V617F males, compared to matching controls. Tissue level histology revealed increased deposition of osteoid (bone matrix prior mineralization) in these mutated mice, suggesting an effect on osteoblast differentiation. Mechanistic studies using a megakaryocyte-osteoblast co-culture system, showed that both wild type or JAK2V617F megakaryocytes derived from male mice inhibited osteoblast differentiation, but JAK2V617F cells exerted a more significant inhibitory effect. A mouse mRNA osteogenesis array showed increased expression of Noggin, Chordin, Alpha-2-HS-glycoprotein, Collagen type IV alpha 1 and Collagen type XIV alpha 1 (mostly known to inhibit bone differentiation), and decreased expression of alkaline phosphatase, Vascular cell adhesion molecule 1, Sclerostin, Distal-less homeobox 5 and Collagen type III alpha 1 (associated with osteogenesis) in JAK2V617F megakaryocytes, compared to controls. This suggested that the mutation re-programs megakaryocytes to express a cluster of genes, which together could orchestrate greater suppression of osteogenesis in male mice. These findings provide mechanistic insight into the effect of JAK2V617F mutation on bone, encouraging future examination of patients with this or other PMF-inducing mutations.
Collapse
Affiliation(s)
- Aikaterini Karagianni
- Department of Internal Medicine, University of Crete, School of Medicine, Heraklion, Greece
- Department of Medicine, Whitaker Cardiovascular Research Institute, Boston University School of Medicine, Boston, MA, United States
| | - Shinobu Matsuura
- Department of Medicine, Whitaker Cardiovascular Research Institute, Boston University School of Medicine, Boston, MA, United States
| | - Louis C. Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, United States
| | - Katya Ravid
- Department of Medicine, Whitaker Cardiovascular Research Institute, Boston University School of Medicine, Boston, MA, United States
- *Correspondence: Katya Ravid,
| |
Collapse
|
13
|
Circulating platelet concentration is associated with bone mineral density in women. Arch Osteoporos 2022; 17:44. [PMID: 35257290 DOI: 10.1007/s11657-022-01089-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/26/2022] [Indexed: 02/03/2023]
Abstract
In this cross-sectional study, enrollment included 818 female adults undergoing bone mineral density (BMD) assessment during the health examination. Subjects with osteoporosis had the lowest circulating platelet concentrations. The circulating platelet concentration was positively correlated with BMD. A high platelet concentration had independently low odds of osteoporosis. PURPOSE Platelets play an important role in bone metabolism. However, the association between circulating platelet counts and bone mineral density (BMD) has been inconsistently reported. We aimed to investigate the relationship between platelet counts and osteoporosis in Chinese women. METHODS In this cross-sectional study, a total of 818 female adults who underwent BMD assessment during the health examination were enrolled. Blood cell counts and biochemistry data were recorded. RESULTS Subjects with osteoporosis had the lowest platelet counts (238 ± 59 × 109/L) compared with subjects with osteopenia (256 ± 64 × 109/L) and a normal BMD (269 ± 76 × 109/L, P < 0.001). The circulating platelet concentration was positively correlated with the BMD of the lumbar spine (r = 0.195, P < 0.001), left hip (r = 0.145, P < 0.001), and right hip (r = 0.149, P < 0.001). According to the receiver operating characteristic curve, the cutoff platelet concentration for differentiating osteoporosis was 260 × 109/L. A high platelet concentration had significantly low odds of osteoporosis after adjusting for other covariates (odds ratio = 0.574, 95% confidence interval: 0.346‒0.953, P = 0.032). CONCLUSION The circulating platelet concentration was significantly correlated with BMD in Chinese women.
Collapse
|
14
|
G6b-B regulates an essential step in megakaryocyte maturation. Blood Adv 2022; 6:3155-3161. [PMID: 35134123 PMCID: PMC9131916 DOI: 10.1182/bloodadvances.2021006151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/20/2022] [Indexed: 12/05/2022] Open
Abstract
Loss of G6b-B leads to an unexpected megakaryocyte development defect resulting in severe macrothrombocytopenia. G6b-B–deficient mice display reduced levels of MK-specific transcripts, surface receptors, GATA-1, and thrombopoietin signaling.
G6b-B is a megakaryocyte lineage-specific immunoreceptor tyrosine-based inhibition motif–containing receptor, essential for platelet homeostasis. Mice with a genomic deletion of the entire Mpig6b locus develop severe macrothrombocytopenia and myelofibrosis, which is reflected in humans with null mutations in MPIG6B. The current model proposes that megakaryocytes lacking G6b-B develop normally, whereas proplatelet release is hampered, but the underlying molecular mechanism remains unclear. We report on a spontaneous recessive single nucleotide mutation in C57BL/6 mice, localized within the intronic region of the Mpig6b locus that abolishes G6b-B expression and reproduces macrothrombocytopenia, myelofibrosis, and osteosclerosis. As the mutation is based on a single-nucleotide exchange, Mpig6bmut mice represent an ideal model to study the role of G6b-B. Megakaryocytes from these mice were smaller, displayed a less-developed demarcation membrane system, and had a reduced expression of receptors. RNA sequencing revealed a striking global reduction in the level of megakaryocyte-specific transcripts, in conjunction with decreased protein levels of the transcription factor GATA-1 and impaired thrombopoietin signaling. The reduced number of mature MKs in the bone marrow was corroborated on a newly developed Mpig6b-null mouse strain. Our findings highlight an unexpected essential role of G6b-B in the early differentiation within the megakaryocytic lineage.
Collapse
|
15
|
Khatib-Massalha E, Méndez-Ferrer S. Megakaryocyte Diversity in Ontogeny, Functions and Cell-Cell Interactions. Front Oncol 2022; 12:840044. [PMID: 35186768 PMCID: PMC8854253 DOI: 10.3389/fonc.2022.840044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cells (HSCs) rely on local interactions in the bone marrow (BM) microenvironment with stromal cells and other hematopoietic cells that facilitate their survival and proliferation, and also regulate their functions. HSCs and multipotent progenitor cells differentiate into lineage-specific progenitors that generate all blood and immune cells. Megakaryocytes (Mks) are hematopoietic cells responsible for producing blood platelets, which are essential for normal hemostasis and blood coagulation. Although the most prominent function of Mks is platelet production (thrombopoiesis), other increasingly recognized functions include HSC maintenance and host immune response. However, whether and how these diverse programs are executed by different Mk subpopulations remains poorly understood. This Perspective summarizes our current understanding of diversity in ontogeny, functions and cell-cell interactions. Cumulative evidence suggests that BM microenvironment dysfunction, partly caused by mutated Mks, can induce or alter the progression of a variety of hematologic malignancies, including myeloproliferative neoplasms (MPNs) and other disorders associated with tissue scarring (fibrosis). Therefore, as an example of the heterogeneous functions of Mks in malignant hematopoiesis, we will discuss the role of Mks in the onset and progression of BM fibrosis. In this regard, abnormal interactions between of Mks and other immune cells might directly contribute to fibrotic diseases. Overall, further understanding of megakaryopoiesis and how Mks interact with HSCs and immune cells has potential clinical implications for stem cell transplantation and other therapies for hematologic malignancies, as well as for treatments to stimulate platelet production and prevent thrombocytopenia.
Collapse
Affiliation(s)
- Eman Khatib-Massalha
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Simón Méndez-Ferrer
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Instituto de Biomedicina de Sevilla-IBiS, Hospitales Universitarios Virgen del Rocío y Macarena/Spanish National Research Council (CSIC)/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
16
|
Yang X, Chitalia SV, Matsuura S, Ravid K. Integrins and their role in megakaryocyte development and function. Exp Hematol 2022; 106:31-39. [PMID: 34910941 PMCID: PMC8795491 DOI: 10.1016/j.exphem.2021.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 02/03/2023]
Abstract
Mature megakaryocytes, the platelet precursors, originate from hematopoietic stem cell progenitors, which, once committed to this lineage, undergo endomitosis leading to polyploidization. The process entails repeated rounds of DNA replication without cell division, yielding polyploid cells. Supporting the cell's developmental process and various cellular functions are integrin receptors, a conduit of communication between the extracellular environment and the cell actin cytoskeleton. Integrins are heterodimers of α and β subunits, where different combinations of the known 18 α and 8 β subunits confer specificity to the receptor. Integrin ligands range from extracellular matrices through soluble ligands, infectious agents, and counterreceptors, to cells. In this review, we describe the different integrins expressed on bone marrow megakaryocytes and their attributed roles in lineage development and cellular functions, including adhesion, spreading, proplatelet formation, and functional interaction with other cells. Pathologies associated with dysregulated megakaryocyte integrin expression are also reviewed.
Collapse
Affiliation(s)
- Xiaosheng Yang
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, 02118
| | - Shlok V. Chitalia
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, 02118
| | - Shinobu Matsuura
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, 02118
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, 02118,To whom correspondence should be addressed: Katya Ravid, Boston University School of Medicine, 700 Albany St, W-6, Boston, MA 02118, Tel: (617)358-8042,
| |
Collapse
|
17
|
Zingariello M, Verachi P, Gobbo F, Martelli F, Falchi M, Mazzarini M, Valeri M, Sarli G, Marinaccio C, Melo-Cardenas J, Crispino JD, Migliaccio AR. Resident Self-Tissue of Proinflammatory Cytokines Rather than Their Systemic Levels Correlates with Development of Myelofibrosis in Gata1low Mice. Biomolecules 2022; 12:biom12020234. [PMID: 35204735 PMCID: PMC8961549 DOI: 10.3390/biom12020234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Serum levels of inflammatory cytokines are currently investigated as prognosis markers in myelofibrosis, the most severe Philadelphia-negative myeloproliferative neoplasm. We tested this hypothesis in the Gata1low model of myelofibrosis. Gata1low mice, and age-matched wild-type littermates, were analyzed before and after disease onset. We assessed cytokine serum levels by Luminex-bead-assay and ELISA, frequency and cytokine content of stromal cells by flow cytometry, and immunohistochemistry and bone marrow (BM) localization of GFP-tagged hematopoietic stem cells (HSC) by confocal microscopy. Differences in serum levels of 32 inflammatory-cytokines between prefibrotic and fibrotic Gata1low mice and their wild-type littermates were modest. However, BM from fibrotic Gata1low mice contained higher levels of lipocalin-2, CXCL1, and TGF-β1 than wild-type BM. Although frequencies of endothelial cells, mesenchymal cells, osteoblasts, and megakaryocytes were higher than normal in Gata1low BM, the cells which expressed these cytokines the most were malignant megakaryocytes. This increased bioavailability of proinflammatory cytokines was associated with altered HSC localization: Gata1low HSC were localized in the femur diaphysis in areas surrounded by microvessels, neo-bones, and megakaryocytes, while wild-type HSC were localized in the femur epiphysis around adipocytes. In conclusion, bioavailability of inflammatory cytokines in BM, rather than blood levels, possibly by reshaping the HSC niche, correlates with myelofibrosis in Gata1low mice.
Collapse
Affiliation(s)
| | - Paola Verachi
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Francesca Gobbo
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Mario Falchi
- National Center HIV/AIDS Research, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Mazzarini
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Mauro Valeri
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | | | - Johanna Melo-Cardenas
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
- Center for Integrated Biomedical Research, Campus Bio-Medico, 00128 Rome, Italy
- Correspondence:
| |
Collapse
|
18
|
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
|
19
|
Liu X, Tao J, Yao Y, Yang P, Wang J, Yu M, Hou J, Zhang Y, Gui L. Resveratrol induces proliferation in preosteoblast cell MC3T3-E1 via GATA-1 activating autophagy. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1495-1504. [PMID: 34637502 DOI: 10.1093/abbs/gmab135] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Indexed: 01/08/2023] Open
Abstract
Resveratrol (RSV) could promote osteogenic activity, but its clinical application has been hampered in view of its poor bioavailability. Therefore, it is desirable to identify with certainty the molecular target of its bone mass boosting function, which is crucial to the design of an effective therapeutic strategy for the optimal treatment of osteoporosis. Emerging evidence has indicated that GATA-1, an important transcription factor in megakaryocyte and erythrocyte differentiation, can directly activate autophagy in erythrocytes, alluding to its impact on bone metabolism. In light of this, we sought to determine whether GATA-1 would be a putative target by which RSV would act on osteoblast proliferation and, if so, to explore the underlying mechanism involved in the process. We examined the cell viability, colony formation, cell cyclin expression, autophagy level, and the expression levels of GATA-1 and adenosine 5'-monophosphate (AMP)-activated protein kinase α (AMPKα) in osteoblastic cell strain MC3T3-E1. The results showed that RSV promoted the proliferation process in MC3T3-E1 coupled with increased expression of GATA-1 and phosphorylated AMPKα and activated autophagy. When GATA-1 was interfered with siRNA, both autophagy and proliferation were decreased. Administration of the agonist of phosphorylated AMPKα1 (Thr172) promoted the translocation of GATA-1 into the nucleus. Based on the above results, we concluded that RSV induces the proliferation of MC3T3-E1 by increasing GATA-1 expression, which thence activates autophagy; and of note, AMPKα is one of the upstream regulators of GATA-1.
Collapse
Affiliation(s)
- Xiang Liu
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Jun Tao
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Yueyi Yao
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Ping Yang
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Juhui Wang
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Mali Yu
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Jianhong Hou
- Department of Orthopaedics, The Third People’s Hospital of Yunnan Province, Kunming 650101, China
| | - Ying Zhang
- Faculty of Nursing, Kunming Medical University, Kunming 650500, China
| | - Li Gui
- Department of Endocrinology, The Third People’s Hospital of Yunnan Province, Kunming 650101, China
| |
Collapse
|
20
|
Aghali A. Craniofacial Bone Tissue Engineering: Current Approaches and Potential Therapy. Cells 2021; 10:cells10112993. [PMID: 34831216 PMCID: PMC8616509 DOI: 10.3390/cells10112993] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 01/10/2023] Open
Abstract
Craniofacial bone defects can result from various disorders, including congenital malformations, tumor resection, infection, severe trauma, and accidents. Successfully regenerating cranial defects is an integral step to restore craniofacial function. However, challenges managing and controlling new bone tissue formation remain. Current advances in tissue engineering and regenerative medicine use innovative techniques to address these challenges. The use of biomaterials, stromal cells, and growth factors have demonstrated promising outcomes in vitro and in vivo. Natural and synthetic bone grafts combined with Mesenchymal Stromal Cells (MSCs) and growth factors have shown encouraging results in regenerating critical-size cranial defects. One of prevalent growth factors is Bone Morphogenetic Protein-2 (BMP-2). BMP-2 is defined as a gold standard growth factor that enhances new bone formation in vitro and in vivo. Recently, emerging evidence suggested that Megakaryocytes (MKs), induced by Thrombopoietin (TPO), show an increase in osteoblast proliferation in vitro and bone mass in vivo. Furthermore, a co-culture study shows mature MKs enhance MSC survival rate while maintaining their phenotype. Therefore, MKs can provide an insight as a potential therapy offering a safe and effective approach to regenerating critical-size cranial defects.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA;
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47908, USA
| |
Collapse
|
21
|
Martelli F, Verachi P, Zingariello M, Mazzarini M, Vannucchi AM, Lonetti A, Bacci B, Sarli G, Migliaccio AR. hGATA1 Under the Control of a μLCR/β-Globin Promoter Rescues the Erythroid but Not the Megakaryocytic Phenotype Induced by the Gata1 low Mutation in Mice. Front Genet 2021; 12:720552. [PMID: 34707640 PMCID: PMC8542976 DOI: 10.3389/fgene.2021.720552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/24/2021] [Indexed: 11/13/2022] Open
Abstract
The phenotype of mice carrying the Gata1low mutation that decreases expression of Gata1 in erythroid cells and megakaryocytes, includes anemia, thrombocytopenia, hematopoietic failure in bone marrow and development of extramedullary hematopoiesis in spleen. With age, these mice develop myelofibrosis, a disease sustained by alterations in stem/progenitor cells and megakaryocytes. This study analyzed the capacity of hGATA1 driven by a μLCR/β-globin promoter to rescue the phenotype induced by the Gata1low mutation in mice. Double hGATA1/Gata1low/0 mice were viable at birth with hematocrits greater than those of their Gata1low/0 littermates but platelet counts remained lower than normal. hGATA1 mRNA was expressed by progenitor and erythroid cells from double mutant mice but not by megakaryocytes analyzed in parallel. The erythroid cells from hGATA1/Gata1low/0 mice expressed greater levels of GATA1 protein and of α- and β-globin mRNA than cells from Gata1low/0 littermates and a reduced number of them was in apoptosis. By contrast, hGATA1/Gata1low/0 megakaryocytes expressed barely detectable levels of GATA1 and their expression of acetylcholinesterase, Von Willebrand factor and platelet factor 4 as well as their morphology remained altered. In comparison with Gata1+/0 littermates, Gata1low/0 mice contained significantly lower total and progenitor cell numbers in bone marrow while the number of these cells in spleen was greater than normal. The presence of hGATA1 greatly increased the total cell number in the bone marrow of Gata1low/0 mice and, although did not affect the total cell number of the spleen which remained greater than normal, it reduced the frequency of progenitor cells in this organ. The ability of hGATA1 to rescue the hematopoietic functions of the bone marrow of the double mutants was confirmed by the observation that these mice survive well splenectomy and did not develop myelofibrosis with age. These results indicate that hGATA1 under the control of µLCR/β-globin promoter is expressed in adult progenitors and erythroid cells but not in megakaryocytes rescuing the erythroid but not the megakaryocyte defect induced by the Gata1low/0 mutation.
Collapse
Affiliation(s)
- Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Verachi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Maria Mazzarini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro M Vannucchi
- Department of Clinical and Experimental Medicine, Center of Research and Innovation of Myeloproliferative neoplasms (CRIMM), AOU Careggi, University of Florence, Florence, Italy
| | - Annalisa Lonetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Barbara Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Myeloproliferative Neoplasm Research Consortium, New York, NY, United States.,Department of Medicine and Surgery, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
22
|
Myeloproliferative Disorders and its Effect on Bone Homeostasis: The Role of Megakaryocytes. Blood 2021; 139:3127-3137. [PMID: 34428274 DOI: 10.1182/blood.2021011480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/11/2021] [Indexed: 11/20/2022] Open
Abstract
Myeloproliferative Neoplasms (MPNs) are a heterogeneous group of chronic hematological diseases that arise from the clonal expansion of abnormal hematopoietic stem cells, of which Polycythemia Vera (PV), Essential Thrombocythemia (ET), and Primary Myelofibrosis (PMF) have been extensively reviewed in context of clonal expansion, fibrosis and other phenotypes. Here, we review current knowledge on the influence of different forms of MPN on bone health. Studies implicated various degrees of effect of different forms of MPN on bone density, and on osteoblast proliferation and differentiation, using murine models and human data. The majority of studies show that bone volume is generally increased in PMF patients, whereas it is slightly decreased or not altered in ET and PV patients, although possible differences between male and female phenotypes were not fully explored in most MPN forms. Osteosclerosis seen in PMF patients is a serious complication that can lead to bone marrow failure, and the loss of bone reported in some ET and PV patients can lead to osteoporotic fractures. Some MPN forms are associated with increased number of megakaryocytes (MKs), and several of the MK-associated factors in MPN are known to affect bone development. Here, we review known mechanisms involved in these processes, with focus on the role of MKs and secreted factors. Understanding MPN-associated changes in bone health could improve early intervention and treatment of this side effect of the pathology.
Collapse
|
23
|
Lee SH, Ihn HJ, Park EK, Kim JE. S100 Calcium-Binding Protein P Secreted from Megakaryocytes Promotes Osteoclast Maturation. Int J Mol Sci 2021; 22:ijms22116129. [PMID: 34200172 PMCID: PMC8201154 DOI: 10.3390/ijms22116129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/26/2022] Open
Abstract
Megakaryocytes (MKs) differentiate from hematopoietic stem cells and produce platelets at the final stage of differentiation. MKs directly interact with bone cells during bone remodeling. However, whether MKs are involved in regulating bone metabolism through indirect regulatory effects on bone cells is unclear. Here, we observed increased osteoclast differentiation of bone marrow-derived macrophages (BMMs) cultured in MK-cultured conditioned medium (MK CM), suggesting that this medium contains factors secreted from MKs that affect osteoclastogenesis. To identify the MK-secreted factor, DNA microarray analysis of the human leukemia cell line K562 and MKs was performed, and S100 calcium-binding protein P (S100P) was selected as a candidate gene affecting osteoclast differentiation. S100P was more highly expressed in MKs than in K562 cells, and showed higher levels in MK CM than in K562-cultured conditioned medium. In BMMs cultured in the presence of recombinant human S100P protein, osteoclast differentiation was promoted and marker gene expression was increased. The resorption area was significantly larger in S100P protein-treated osteoclasts, demonstrating enhanced resorption activity. Overall, S100P secreted from MKs promotes osteoclast differentiation and resorption activity, suggesting that MKs indirectly regulate osteoclast differentiation and activity through the paracrine action of S100P.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, Kyungpook National University, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea;
| | - Hye Jung Ihn
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea;
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, IHBR, Kyungpook National University, Daegu 41944, Korea;
| | - Jung-Eun Kim
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, Kyungpook National University, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea;
- Correspondence: ; Tel.: +82-53-420-4949
| |
Collapse
|
24
|
Stavnichuk M, Tauer JT, Nagy Z, Mazharian A, Welman M, Lordkipanidzé M, Senis YA, Komarova SV. Severity of Megakaryocyte-Driven Osteosclerosis in Mpig6b-Deficient Mice Is Sex-Linked. J Bone Miner Res 2021; 36:803-813. [PMID: 33434328 DOI: 10.1002/jbmr.4245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/16/2020] [Accepted: 12/29/2020] [Indexed: 01/15/2023]
Abstract
Patients with chronic myelofibrosis often suffer from osteosclerosis, which is associated with bone pain and may lead to bone marrow failure. The pathogenesis of myelofibrosis is linked to aberrant megakaryocyte development and function. Null and loss-of-function mutations in MPIG6B, which codes for the inhibitory heparan sulfate receptor G6b-B, result in severe macrothrombocytopenia, large megakaryocyte clusters, and focal primary myelofibrosis in mice and humans. We investigated the development of osteosclerosis in Mpig6b null (Mpig6b-/- ) mice. Although male and female Mpig6b-/- mice presented with elevated bone marrow megakaryocyte number and macrothrombocytopenia, female Mpig6b-/- mice developed progressive splenomegaly starting at 8 weeks of age. Micro-computed tomography (μCT) of femurs showed that female Mpig6b-/- mice had increased cortical thickness and reduced bone marrow area starting at 8 weeks of age and developed occlusion of the medullary cavity by trabeculae by 16 weeks of age. In contrast, male Mpig6b-/- mice developed only a small number of trabeculae in the medullary cavity at the proximal diaphysis and demonstrated a temporary decrease in bone volume fraction and trabecular thickness at 16 weeks. Ovariectomy of 10-week-old female Mpig6b-/- mice prevented the development of medullary cavity osteosclerosis, whereas orchiectomy of male Mpig6b-/- mice did not exacerbate their disease. Importantly, ovariectomized female Mpig6b-/- mice also demonstrated improvement in spleen weight compared to sham-operated Mpig6b-/- mice, establishing estrogen as a contributing factor to the severity of the megakaryocyte-driven osteosclerosis. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Mariya Stavnichuk
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada.,Shriners Hospital for Children-Canada, Montreal, QC, Canada
| | - Josephine T Tauer
- Shriners Hospital for Children-Canada, Montreal, QC, Canada.,Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Zoltan Nagy
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Alexandra Mazharian
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale (INSERM), Etablissement Français du Sang Grand Est, Unité Mixte de Recherche (UMR)-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Mélanie Welman
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC, Canada.,Faculty of Pharmacy, University of Montreal, Montreal, QC, Canada
| | - Yotis A Senis
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale (INSERM), Etablissement Français du Sang Grand Est, Unité Mixte de Recherche (UMR)-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Svetlana V Komarova
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada.,Shriners Hospital for Children-Canada, Montreal, QC, Canada.,Faculty of Dentistry, McGill University, Montreal, QC, Canada
| |
Collapse
|
25
|
Zamarioli A, Campbell ZR, Maupin KA, Childress PJ, Ximenez JPB, Adam G, Chakraborty N, Gautam A, Hammamieh R, Kacena MA. Analysis of the effects of spaceflight and local administration of thrombopoietin to a femoral defect injury on distal skeletal sites. NPJ Microgravity 2021; 7:12. [PMID: 33772025 PMCID: PMC7997973 DOI: 10.1038/s41526-021-00140-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 02/26/2021] [Indexed: 02/01/2023] Open
Abstract
With increased human presence in space, bone loss and fractures will occur. Thrombopoietin (TPO) is a recently patented bone healing agent. Here, we investigated the systemic effects of TPO on mice subjected to spaceflight and sustaining a bone fracture. Forty, 9-week-old, male, C57BL/6 J were divided into 4 groups: (1) Saline+Earth; (2) TPO + Earth; (3) Saline+Flight; and (4) TPO + Flight (n = 10/group). Saline- and TPO-treated mice underwent a femoral defect surgery, and 20 mice were housed in space ("Flight") and 20 mice on Earth for approximately 4 weeks. With the exception of the calvarium and incisor, positive changes were observed in TPO-treated, spaceflight bones, suggesting TPO may improve osteogenesis in the absence of mechanical loading. Thus, TPO, may serve as a new bone healing agent, and may also improve some skeletal properties of astronauts, which might be extrapolated for patients on Earth with restraint mobilization and/or are incapable of bearing weight on their bones.
Collapse
Affiliation(s)
- Ariane Zamarioli
- grid.257413.60000 0001 2287 3919Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN USA ,Department of Orthopaedics and Anaesthesiology, Ribeirão Preto Medical School, Ribeirão Preto, SP Brazil
| | - Zachery R. Campbell
- grid.257413.60000 0001 2287 3919Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN USA ,Marian University College of Osteopathic Medicine, Indianapolis, IN USA
| | - Kevin A. Maupin
- grid.257413.60000 0001 2287 3919Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN USA
| | - Paul J. Childress
- grid.257413.60000 0001 2287 3919Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN USA
| | - Joao P. B. Ximenez
- Laboratory of Molecular Biology, Blood Center of Ribeirão Preto, Medical School, Ribeirão Pre, SP Brazil
| | - Gremah Adam
- grid.257413.60000 0001 2287 3919Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN USA
| | - Nabarun Chakraborty
- grid.507680.c0000 0001 2230 3166Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD USA ,grid.507680.c0000 0001 2230 3166Geneva Foundation, Walter Reed Army Institute of Research, Silver Spring, MD USA
| | - Aarti Gautam
- grid.507680.c0000 0001 2230 3166Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD USA
| | - Rasha Hammamieh
- grid.507680.c0000 0001 2230 3166Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD USA
| | - Melissa A. Kacena
- grid.257413.60000 0001 2287 3919Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN USA ,grid.280828.80000 0000 9681 3540Richard L. Roudebush VA Medical Center, Indianapolis, IN USA
| |
Collapse
|
26
|
Kanagasabapathy D, Blosser RJ, Maupin KA, Hong JM, Alvarez M, Ghosh J, Mohamad SF, Aguilar-Perez A, Srour EF, Kacena MA, Bruzzaniti A. Megakaryocytes promote osteoclastogenesis in aging. Aging (Albany NY) 2020; 12:15121-15133. [PMID: 32634116 PMCID: PMC7425434 DOI: 10.18632/aging.103595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 06/13/2020] [Indexed: 01/26/2023]
Abstract
Megakaryocytes (MKs) support bone formation by stimulating osteoblasts (OBs) and inhibiting osteoclasts (OCs). Aging results in higher bone resorption, leading to bone loss. Whereas previous studies showed the effects of aging on MK-mediated bone formation, the effects of aging on MK-mediated OC formation is poorly understood. Here we examined the effect of thrombopoietin (TPO) and MK-derived conditioned media (CM) from young (3-4 months) and aged (22-25 months) mice on OC precursors. Our findings showed that aging significantly increased OC formation in vitro. Moreover, the expression of the TPO receptor, Mpl, and circulating TPO levels were elevated in the bone marrow cavity. We previously showed that MKs from young mice secrete factors that inhibit OC differentiation. However, rather than inhibiting OC development, we found that MKs from aged mice promote OC formation. Interestingly, these age-related changes in MK functionality were only observed using female MKs, potentially implicating the sex steroid, estrogen, in signaling. Further, RANKL expression was highly elevated in aged MKs suggesting MK-derived RANKL signaling may promote osteoclastogenesis in aging. Taken together, these data suggest that modulation in TPO-Mpl expression in bone marrow and age-related changes in the MK secretome promote osteoclastogenesis to impact skeletal aging.
Collapse
Affiliation(s)
- Deepa Kanagasabapathy
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rachel J Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kevin A Maupin
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jung Min Hong
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN 46202, USA
| | - Marta Alvarez
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Joydeep Ghosh
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Safa F Mohamad
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexandra Aguilar-Perez
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN 46202, USA
| | - Edward F Srour
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Angela Bruzzaniti
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN 46202, USA
| |
Collapse
|
27
|
Lee YS, Kwak MK, Moon SA, Choi YJ, Baek JE, Park SY, Kim BJ, Lee SH, Koh JM. Regulation of bone metabolism by megakaryocytes in a paracrine manner. Sci Rep 2020; 10:2277. [PMID: 32042021 PMCID: PMC7010738 DOI: 10.1038/s41598-020-59250-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 01/23/2020] [Indexed: 11/10/2022] Open
Abstract
Megakaryocytes (MKs) play key roles in regulating bone metabolism. To test the roles of MK-secreted factors, we investigated whether MK and promegakaryocyte (pro-MK) conditioned media (CM) may affect bone formation and resorption. K562 cell lines were differentiated into mature MKs. Mouse bone marrow macrophages were differentiated into mature osteoclasts, and MC3T3-E1 cells were used for osteoblastic experiments. Bone formation was determined by a calvaria bone formation assay in vivo. Micro-CT analyses were performed in the femurs of ovariectomized female C57B/L6 and Balb/c nude mice after intravenous injections of MK or pro-MK CM. MK CM significantly reduced in vitro bone resorption, largely due to suppressed osteoclastic resorption activity. Compared with pro-MK CM, MK CM suppressed osteoblastic differentiation, but stimulated its proliferation, resulting in stimulation of calvaria bone formation. In ovariectomized mice, treatment with MK CM for 4 weeks significantly increased trabecular bone mass parameters, such as bone volume fraction and trabecular thickness, in nude mice, but not in C57B/L6 mice. In conclusion, MKs may secrete anti-resorptive and anabolic factors that affect bone tissue, providing a novel insight linking MKs and bone cells in a paracrine manner. New therapeutic agents against metabolic bone diseases may be developed from MK-secreted factors.
Collapse
Affiliation(s)
- Young-Sun Lee
- Asan Institute for Life Sciences, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea
| | - Mi Kyung Kwak
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, 7, Keunjaebong-gil, Hwaseong-Si, Gyeonggi-Do, 445-907, Korea
| | - Sung-Ah Moon
- Asan Institute for Life Sciences, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea
| | - Young Jin Choi
- Asan Institute for Life Sciences, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea
| | - Ji Eun Baek
- Asan Institute for Life Sciences, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea
| | - Suk Young Park
- Department of Orthopedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea
| | - Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43 gil, Songpa-gu, Seoul, 05505, Korea.
| |
Collapse
|
28
|
Tang Y, Hu M, Xu Y, Chen F, Chen S, Chen M, Qi Y, Shen M, Wang C, Lu Y, Zhang Z, Zeng H, Quan Y, Wang F, Su Y, Zeng D, Wang S, Wang J. Megakaryocytes promote bone formation through coupling osteogenesis with angiogenesis by secreting TGF-β1. Am J Cancer Res 2020; 10:2229-2242. [PMID: 32104505 PMCID: PMC7019172 DOI: 10.7150/thno.40559] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022] Open
Abstract
Rationale: The hematopoietic system and skeletal system have a close relationship, and megakaryocytes (MKs) may be involved in maintaining bone homeostasis. However, the exact role and underlying mechanism of MKs in bone formation during steady-state and stress conditions are still unclear. Methods: We first evaluated the bone phenotype with MKs deficiency in bone marrow by using c-Mpl-deficient mice and MKs-conditionally deleted mice. Then, osteoblasts (OBs) proliferation and differentiation and CD31hiEmcnhi tube formation were assessed. The expression of growth factors related to bone formation in MKs was detected by RNA-sequencing and enzyme-linked immunosorbent assays (ELISAs). Mice with specific depletion of TGF-β1 in MKs were used to further verify the effect of MKs on osteogenesis and angiogenesis. Finally, MKs treatment of irradiation-induced bone injury was tested in a mouse model. Results: We found that MKs deficiency significantly impaired bone formation. Further investigations revealed that MKs could promote OBs proliferation and differentiation, as well as CD31hiEmcnhi vessels formation, by secreting high levels of TGF-β1. Consistent with these findings, mice with specific depletion of TGF-β1 in MKs displayed significantly decreased bone mass and strength. Importantly, treatment with MKs or thrombopoietin (TPO) substantially attenuated radioactive bone injury in mice by directly or indirectly increasing the level of TGF-β1 in bone marrow. MKs-derived TGF-β1 was also involved in suppressing apoptosis and promoting DNA damage repair in OBs after irradiation exposure. Conclusions: Our findings demonstrate that MKs contribute to bone formation through coupling osteogenesis with angiogenesis by secreting TGF-β1, which may offer a potential therapeutic strategy for the treatment of irradiation-induced osteoporosis.
Collapse
|
29
|
Maupin KA, Himes ER, Plett AP, Chua HL, Singh P, Ghosh J, Mohamad SF, Abeysekera I, Fisher A, Sampson C, Hong JM, Childress P, Alvarez M, Srour EF, Bruzzaniti A, Pelus LM, Orschell CM, Kacena MA. Aging negatively impacts the ability of megakaryocytes to stimulate osteoblast proliferation and bone mass. Bone 2019; 127:452-459. [PMID: 31299382 PMCID: PMC6708771 DOI: 10.1016/j.bone.2019.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/15/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022]
Abstract
Osteoblast number and activity decreases with aging, contributing to the age-associated decline of bone mass, but the mechanisms underlying changes in osteoblast activity are not well understood. Here, we show that the age-associated bone loss critically depends on impairment of the ability of megakaryocytes (MKs) to support osteoblast proliferation. Co-culture of osteoblast precursors with young MKs is known to increase osteoblast proliferation and bone formation. However, co-culture of osteoblast precursors with aged MKs resulted in significantly fewer osteoblasts compared to co-culture with young MKs, and this was associated with the downregulation of transforming growth factor beta. In addition, the ability of MKs to increase bone mass was attenuated during aging as transplantation of GATA1low/low hematopoietic donor cells (which have elevated MKs/MK precursors) from young mice resulted in an increase in bone mass of recipient mice compared to transplantation of young wild-type donor cells, whereas transplantation of GATA1low/low donor cells from old mice failed to enhance bone mass in recipient mice compared to transplantation of old wild-type donor cells. These findings suggest that the preservation or restoration of the MK-mediated induction of osteoblast proliferation during aging may hold the potential to prevent age-associated bone loss and resulting fractures.
Collapse
Affiliation(s)
| | - Evan R Himes
- Indiana University School of Medicine, Indiana, USA
| | | | - Hui Lin Chua
- Indiana University School of Medicine, Indiana, USA
| | | | | | | | | | - Alexa Fisher
- Indiana University School of Medicine, Indiana, USA
| | | | - Jung-Min Hong
- Indiana University School of Dentistry, Indiana, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Alvarez MB, Xu L, Childress PJ, Maupin KA, Mohamad SF, Chitteti BR, Himes E, Olivos DJ, Cheng YH, Conway SJ, Srour EF, Kacena MA. Megakaryocyte and Osteoblast Interactions Modulate Bone Mass and Hematopoiesis. Stem Cells Dev 2019; 27:671-682. [PMID: 29631496 DOI: 10.1089/scd.2017.0178] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Emerging evidence demonstrates that megakaryocytes (MK) play key roles in regulating skeletal homeostasis and hematopoiesis. To test if the loss of MK negatively impacts osteoblastogenesis and hematopoiesis, we generated conditional knockout mice where Mpl, the receptor for the main MK growth factor, thrombopoietin, was deleted specifically in MK (Mplf/f;PF4cre). Unexpectedly, at 12 weeks of age, these mice exhibited a 10-fold increase in platelets, a significant expansion of hematopoietic/mesenchymal precursors, and a remarkable 20-fold increase in femoral midshaft bone volume. We then investigated whether MK support hematopoietic stem cell (HSC) function through the interaction of MK with osteoblasts (OB). LSK cells (Lin-Sca1+CD117+, enriched HSC population) were co-cultured with OB+MK for 1 week (1wk OB+MK+LSK) or OB alone (1wk OB+LSK). A significant increase in colony-forming units was observed with cells from 1wk OB+MK cultures. Competitive repopulation studies demonstrated significantly higher engraftment in mice transplanted with cells from 1wk OB+MK+LSK cultures compared to 1wk OB+LSK or LSK cultured alone for 1 week. Furthermore, single-cell expression analysis of OB cultured±MK revealed adiponectin as the most significantly upregulated MK-induced gene, which is required for optimal long-term hematopoietic reconstitution. Understanding the interactions between MK, OB, and HSC can inform the development of novel treatments to enhance both HSC recovery following myelosuppressive injuries, as well as bone loss diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Marta B Alvarez
- 1 Department of Orthopaedic Surgery, Indiana University School of Medicine , Indianapolis, Indiana
| | - LinLin Xu
- 2 Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Paul J Childress
- 1 Department of Orthopaedic Surgery, Indiana University School of Medicine , Indianapolis, Indiana
| | - Kevin A Maupin
- 1 Department of Orthopaedic Surgery, Indiana University School of Medicine , Indianapolis, Indiana
| | - Safa F Mohamad
- 2 Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | | | - Evan Himes
- 1 Department of Orthopaedic Surgery, Indiana University School of Medicine , Indianapolis, Indiana
| | - David J Olivos
- 1 Department of Orthopaedic Surgery, Indiana University School of Medicine , Indianapolis, Indiana
| | - Ying-Hua Cheng
- 1 Department of Orthopaedic Surgery, Indiana University School of Medicine , Indianapolis, Indiana
| | - Simon J Conway
- 3 Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine , Indianapolis, Indiana
| | - Edward F Srour
- 2 Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana.,3 Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine , Indianapolis, Indiana
| | - Melissa A Kacena
- 1 Department of Orthopaedic Surgery, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
31
|
Zhang J, Ghosh J, Mohamad SF, Zhang C, Huang X, Capitano ML, Gunawan AM, Cooper S, Guo B, Cai Q, Broxmeyer HE, Srour EF. CD166 Engagement Augments Mouse and Human Hematopoietic Progenitor Function via Activation of Stemness and Cell Cycle Pathways. Stem Cells 2019; 37:1319-1330. [PMID: 31260147 DOI: 10.1002/stem.3053] [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: 01/31/2018] [Accepted: 06/21/2019] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem (HSC) and progenitor (HPC) cells are regulated by interacting signals and cellular and noncellular elements of the hematopoietic niche. We previously showed that CD166 is a functional marker of murine and human HSC and of cellular components of the murine niche. Selection of murine CD166+ engrafting HSC enriched for marrow repopulating cells. Here, we demonstrate that CD166-CD166 homophilic interactions enhance generation of murine and human HPC in vitro and augment hematopoietic function of these cells. Interactions between cultured CD166+ Lineage- Sca-1+ c-Kit+ (LSK) cells and CD166+ osteoblasts (OBs) significantly enhanced the expansion of colony-forming units (CFUs). Interactions between CD166+ LSK cells and immobilized CD166 protein generated more CFU in short-term cultures than between these cells and bovine serum albumin (BSA) or in cultures initiated with CD166- LSK cells. Similar results were obtained when LSK cells from wildtype (WT) or CD166 knockout (KO) (CD166-/- ) mice were used with immobilized CD166. Human cord blood CD34+ cells expressing CD166 produced significantly higher numbers of CFUs following interaction with immobilized CD166 than their CD166- counterparts. These data demonstrate the positive effects of CD166 homophilic interactions involving CD166 on the surface of murine and human HPCs. Single-cell RNA-seq analysis of CD150+ CD48- (signaling lymphocyte activation molecule (SLAM)) LSK cells from WT and CD166-/- mice incubated with immobilized CD166 protein revealed that engagement of CD166 on these cells activates cytokine, growth factor and hormone signaling, epigenetic pathways, and other genes implicated in maintenance of stem cell pluripotency-related and mitochondria-related signaling pathways. These studies provide tangible evidence implicating CD166 engagement in the maintenance of stem/progenitor cell function. Stem Cells 2019;37:1319-1330.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, AMMS, Beijing, People's Republic of China.,Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, AMMS, Beijing, People's Republic of China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, People's Republic of China
| | - Joydeep Ghosh
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Safa F Mohamad
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Xinxin Huang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Maegan L Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrea M Gunawan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Scott Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bin Guo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Qingchun Cai
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Edward F Srour
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
32
|
Pearson HB, Mason DE, Kegelman CD, Zhao L, Dawahare JH, Kacena MA, Boerckel JD. Effects of Bone Morphogenetic Protein-2 on Neovascularization During Large Bone Defect Regeneration. Tissue Eng Part A 2019; 25:1623-1634. [PMID: 30973074 DOI: 10.1089/ten.tea.2018.0326] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Insufficient blood vessel supply is a primary limiting factor for regenerative approaches to large bone defect repair. Recombinant bone morphogenetic protein-2 (BMP-2) delivery induces robust bone formation and has been observed to enhance neovascularization, but whether the angiogenic effects of BMP-2 are due to direct endothelial cell stimulation or due to indirect paracrine signaling remain unclear. In this study, we evaluated the effects of BMP-2 delivery on vascularized bone regeneration and tested whether BMP-2 induces neovascularization directly or indirectly. We found that delivery of BMP-2 (5 μg) enhanced both bone formation and neovascularization in critically sized (8 mm) rat femoral bone defects; however, BMP-2 did not directly stimulate angiogenesis in vitro. In contrast, conditioned medium from both mesenchymal progenitor cells and osteoblasts induced endothelial cell migration in vitro, suggesting a paracrine mechanism of BMP-2 action. Consistent with this inference, codelivery of BMP-2 with endothelial colony forming cells to a heterotopic site, distant from the skeletal stem cell-rich bone marrow niche, induced ossification but had no effect on neovascularization. Taken together, these data suggest that paracrine activation of osteoprogenitor cells is an important contributor to neovascularization during BMP-2-mediated bone regeneration. Impact Statement In this study, we show that bone morphogenetic protein-2 (BMP-2) robustly induces neovascularization during tissue-engineered large bone defect regeneration, and we found that BMP-2 induced angiogenesis, in part, through paracrine signaling from osteoprogenitor cells.
Collapse
Affiliation(s)
- Hope B Pearson
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Devon E Mason
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana.,Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher D Kegelman
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Liming Zhao
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - James H Dawahare
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Joel D Boerckel
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana.,Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
33
|
Megakaryocytes in Bone Metastasis: Protection or Progression? Cells 2019; 8:cells8020134. [PMID: 30744029 PMCID: PMC6406759 DOI: 10.3390/cells8020134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 01/07/2023] Open
Abstract
Bone is the primary site where some cancers develop secondary growth, particularly those derived from breast and prostate tissue. The spread of metastasis to distant sites relies on complex mechanisms by which only cells endowed with certain characteristics are able to reach secondary growth sites. Platelets play a pivotal role in tumour growth, by conferring resistance to shear stress to the circulating tumour cells and protection against natural killer cell attack. Mature polyploid megakaryocytes (MKs) reside in close proximity to the vascular sinusoids of bone marrow, where their primary function is to produce platelets. Emerging evidence has demonstrated that MKs are essential for skeletal homeostasis, due to the expression and production of the bone-related proteins osteocalcin, osteonectin, bone morphogenetic protein, osteopontin, bone sialoprotein, and osteoprotegerin. Debate surrounds the role that MKs play in the development of bone metastasis, which is the topic of this mini-review.
Collapse
|
34
|
Fischer L, Herkner C, Kitte R, Dohnke S, Riewaldt J, Kretschmer K, Garbe AI. Foxp3 + Regulatory T Cells in Bone and Hematopoietic Homeostasis. Front Endocrinol (Lausanne) 2019; 10:578. [PMID: 31551927 PMCID: PMC6746882 DOI: 10.3389/fendo.2019.00578] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/08/2019] [Indexed: 12/29/2022] Open
Abstract
The bone represents surprisingly dynamic structures that are subject to constant remodeling by the concerted action of bone-forming osteoblasts and bone-resorbing osteoclasts - two cell subsets of distinct developmental origin that are key in maintaining skeletal integrity throughout life. In general, abnormal bone remodeling due to dysregulated bone resorption and formation is an early event in the manifestation of various human bone diseases, such as osteopetrosis/osteoporosis and arthritis. But bone remodeling is also closely interrelated with lympho-hematopoietic homeostasis, as the bone marrow niche is formed by solid and trabecular bone structures that provide a framework for the long-term maintenance and differentiation of HSCs (>blood lineage cells and osteoclasts) and MSCs (>osteoblasts). Numerous studies in mice and humans have implicated innate and adaptive immune cells in the dynamic regulation of bone homeostasis, but despite considerable clinical relevance, the exact mechanisms of such immuno-bone interplay have remained incompletely understood. This holds particularly true for CD4+ regulatory T (Treg) cells expressing the lineage specification factor Foxp3: Foxp3+ Treg cells have been shown to play an indispensable role in maintaining immune homeostasis, but may also exert critical non-immune functions, which includes the control of metabolic and regenerative processes, as well as the differentiation of HSCs and function of osteoclasts. Here, we summarize our current knowledge on the T cell/bone interplay, with a particular emphasis on our own efforts to dissect the role of Foxp3+ Treg cells in bone and hematopoietic homeostasis, employing experimental settings of gain- and loss-of-Treg cell function. These data make a strong case that Foxp3+ Treg cells impinge on lympho-hematopoiesis through indirect mechanisms, i.e., by acting on osteoclast development and function, which translates into changes in niche size. Furthermore, we propose that, besides disorders that involve inflammatory bone loss, the modulation of Foxp3+ Treg cell function in vivo may represent a suitable approach to reinstate bone homeostasis in non-autoimmune settings of aberrant bone remodeling.
Collapse
Affiliation(s)
- Luise Fischer
- Osteoimmunology, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Caroline Herkner
- Osteoimmunology, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Reni Kitte
- Osteoimmunology, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Sebastian Dohnke
- Osteoimmunology, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Julia Riewaldt
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Karsten Kretschmer
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Annette I. Garbe
- Osteoimmunology, DFG-Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- *Correspondence: Annette I. Garbe
| |
Collapse
|
35
|
Leblanc R, Houssin A, Peyruchaud O. Platelets, autotaxin and lysophosphatidic acid signalling: win-win factors for cancer metastasis. Br J Pharmacol 2018; 175:3100-3110. [PMID: 29777586 PMCID: PMC6031885 DOI: 10.1111/bph.14362] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 12/19/2022] Open
Abstract
Platelets play a crucial role in the survival of metastatic cells in the blood circulation. The interaction of tumour cells with platelets leads to the production of plethoric factors among which our review will focus on lysophosphatidic acid (LPA), because platelets are the highest producers of this bioactive lysophospholipid in the organism. LPA promotes platelet aggregation, and blocking platelet function decreases LPA signalling and leads to inhibition of breast cancer cell metastasis. Autotaxin (ATX), a lysophospholipase D responsible for the basal concentration of LPA in blood, was detected in platelet α-granules. Functionally, active ATX is eventually released following tumour cell-induced platelet aggregation, thereby promoting metastasis. Megakaryocytes do not express ATX but respond to LPA stimulation. Whether LPA-primed megakaryocytes contribute to the recently reported negative action of megakaryocytes on cancer metastasis is not yet known. However, an understanding of the ATX/LPA signalling pathways in platelets, cancer cells and megakaryocytes opens up new approaches for fighting cancer metastasis.
Collapse
Affiliation(s)
- Raphael Leblanc
- Centre de Recherche en Cancérologie de Marseille, INSERM, CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Audrey Houssin
- INSERM, UMR_S1033, Université Claude Bernard Lyon-1, Lyon, France
| | | |
Collapse
|
36
|
Olivos DJ, Perrien DS, Hooker A, Cheng YH, Fuchs RK, Hong JM, Bruzzaniti A, Chun K, Eischen CM, Kacena MA, Mayo LD. The proto-oncogene function of Mdm2 in bone. J Cell Biochem 2018; 119:8830-8840. [PMID: 30011084 DOI: 10.1002/jcb.27133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 05/07/2018] [Indexed: 12/19/2022]
Abstract
Mouse double minute 2 (Mdm2) is a multifaceted oncoprotein that is highly regulated with distinct domains capable of cellular transformation. Loss of Mdm2 is embryonically lethal, making it difficult to study in a mouse model without additional genetic alterations. Global overexpression through increased Mdm2 gene copy number (Mdm2Tg ) results in the development of hematopoietic neoplasms and sarcomas in adult animals. In these mice, we found an increase in osteoblastogenesis, differentiation, and a high bone mass phenotype. Since it was difficult to discern the cell lineage that generated this phenotype, we generated osteoblast-specific Mdm2 overexpressing (Mdm2TgOb ) mice in 2 different strains, C57BL/6 and DBA. These mice did not develop malignancies; however, these animals and the MG63 human osteosarcoma cell line with high levels of Mdm2 showed an increase in bone mineralization. Importantly, overexpression of Mdm2 corrected age-related bone loss in mice, providing a role for the proto-oncogenic activity of Mdm2 in bone health of adult animals.
Collapse
Affiliation(s)
- David J Olivos
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Daniel S Perrien
- Departments of Medicine and Orthopaedic Surgery and Rehabilitation, Vanderbilt University Medical Center, and Tennessee Valley Healthcare System, Nashville, Tennessee.,Department of Veterans Affairs, Nashville, Tennessee
| | - Adam Hooker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ying-Hua Cheng
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Robyn K Fuchs
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, Indiana
| | - Jung Min Hong
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Angela Bruzzaniti
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Kristin Chun
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christine M Eischen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lindsey D Mayo
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
37
|
Yorgan T, David JP, Amling M, Schinke T. The high bone mass phenotype of Lrp5-mutant mice is not affected by megakaryocyte depletion. Biochem Biophys Res Commun 2018; 497:659-666. [PMID: 29454962 DOI: 10.1016/j.bbrc.2018.02.127] [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: 02/12/2018] [Accepted: 02/14/2018] [Indexed: 11/18/2022]
Abstract
Bone remodeling is a continuously ongoing process mediated by bone-resorbing osteoclasts and bone-forming osteoblasts. One key regulator of bone formation is the putative Wnt co-receptor Lrp5, where activating mutations in the extracellular domain cause increased bone formation in mice and humans. We have previously reported that megakaryocyte numbers are increased the bone marrow of mice carrying a high bone mass mutation (HBM) of Lrp5 (Lrp5G170V). Since megakaryocytes can promote bone formation, we addressed the question, if the bone remodeling phenotype of Lrp5G170V mice is affected by megakaryocyte depletion. For that purpose we took advantage of a mouse model carrying a mutation of the Mpl gene, encoding the thrombopoietin receptor. These mice (Mplhlb219) were crossed with Lrp5G170V mice to generate animals carrying both mutations in a homozygous state. Using μCT, undecalcified histology and bone-specific histomorphometry of 12 weeks old littermates we observed that megakaryocyte number was remarkably decreased in Mplhlb219/Lrp5G170V mice, yet the high bone mass phenotype of Lrp5G170V mice was not significantly affected by the homozygous Mpl mutation. Finally, when we analyzed 24 weeks old wildtype and Mplhlb219 mice we did not observe a statistically significant alteration of bone remodeling in the latter ones. Taken together, our results demonstrate that an increased number of bone marrow megakaryocytes does not contribute to the increased bone formation caused by Lrp5 activation.
Collapse
Affiliation(s)
- Timur Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany
| | - Jean-Pierre David
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany.
| |
Collapse
|
38
|
Ling T, Crispino JD, Zingariello M, Martelli F, Migliaccio AR. GATA1 insufficiencies in primary myelofibrosis and other hematopoietic disorders: consequences for therapy. Expert Rev Hematol 2018; 11:169-184. [PMID: 29400094 PMCID: PMC6108178 DOI: 10.1080/17474086.2018.1436965] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION GATA1, the founding member of a family of transcription factors, plays important roles in the development of hematopoietic cells of several lineages. Although loss of GATA1 has been known to impair hematopoiesis in animal models for nearly 25 years, the link between GATA1 defects and human blood diseases has only recently been realized. Areas covered: Here the current understanding of the functions of GATA1 in normal hematopoiesis and how it is altered in disease is reviewed. GATA1 is indispensable mainly for erythroid and megakaryocyte differentiation. In erythroid cells, GATA1 regulates early stages of differentiation, and its deficiency results in apoptosis. In megakaryocytes, GATA1 controls terminal maturation and its deficiency induces proliferation. GATA1 alterations are often found in diseases involving these two lineages, such as congenital erythroid and/or megakaryocyte deficiencies, including Diamond Blackfan Anemia (DBA), and acquired neoplasms, such as acute megakaryocytic leukemia (AMKL) and the myeloproliferative neoplasms (MPNs). Expert commentary: Since the first discovery of GATA1 mutations in AMKL, the number of diseases that are associated with impaired GATA1 function has increased to include DBA and MPNs. With respect to the latter, we are only just now appreciating the link between enhanced JAK/STAT signaling, GATA1 deficiency and disease pathogenesis.
Collapse
Affiliation(s)
- Te Ling
- Division of Hematology/Oncology, Northwestern University, Chicago, IL, USA
| | - John D. Crispino
- Division of Hematology/Oncology, Northwestern University, Chicago, IL, USA
| | | | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Roma, Italy
| | - Anna Rita Migliaccio
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
| |
Collapse
|
39
|
Xu X, Zheng L, Yuan Q, Zhen G, Crane JL, Zhou X, Cao X. Transforming growth factor-β in stem cells and tissue homeostasis. Bone Res 2018; 6:2. [PMID: 29423331 PMCID: PMC5802812 DOI: 10.1038/s41413-017-0005-4] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/12/2017] [Accepted: 11/15/2017] [Indexed: 02/05/2023] Open
Abstract
TGF-β 1-3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Gehua Zhen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Janet L. Crane
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
40
|
Osteomacs interact with megakaryocytes and osteoblasts to regulate murine hematopoietic stem cell function. Blood Adv 2017; 1:2520-2528. [PMID: 29296903 DOI: 10.1182/bloodadvances.2017011304] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/01/2017] [Indexed: 12/15/2022] Open
Abstract
Networking between hematopoietic stem cells (HSCs) and cells of the hematopoietic niche is critical for stem cell function and maintenance of the stem cell pool. We characterized calvariae-resident osteomacs (OMs) and their interaction with megakaryocytes to sustain HSC function and identified distinguishing properties between OMs and bone marrow (BM)-derived macrophages. OMs, identified as CD45+F4/80+ cells, were easily detectable (3%-5%) in neonatal calvarial cells. Coculture of neonatal calvarial cells with megakaryocytes for 7 days increased OM three- to sixfold, demonstrating that megakaryocytes regulate OM proliferation. OMs were required for the hematopoiesis-enhancing activity of osteoblasts, and this activity was augmented by megakaryocytes. Serial transplantation demonstrated that HSC repopulating potential was best maintained by in vitro cultures containing osteoblasts, OMs, and megakaryocytes. With or without megakaryocytes, BM-derived macrophages were unable to functionally substitute for neonatal calvarial cell-associated OMs. In addition, OMs differentiated into multinucleated, tartrate resistant acid phosphatase-positive osteoclasts capable of bone resorption. Nine-color flow cytometric analysis revealed that although BM-derived macrophages and OMs share many cell surface phenotypic similarities (CD45, F4/80, CD68, CD11b, Mac2, and Gr-1), only a subgroup of OMs coexpressed M-CSFR and CD166, thus providing a unique profile for OMs. CD169 was expressed by both OMs and BM-derived macrophages and therefore was not a distinguishing marker between these 2 cell types. These results demonstrate that OMs support HSC function and illustrate that megakaryocytes significantly augment the synergistic activity of osteoblasts and OMs. Furthermore, this report establishes for the first time that the crosstalk between OMs, osteoblasts, and megakaryocytes is a novel network supporting HSC function.
Collapse
|
41
|
Hoggatt J, Kfoury Y, Scadden DT. Hematopoietic Stem Cell Niche in Health and Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 11:555-81. [PMID: 27193455 DOI: 10.1146/annurev-pathol-012615-044414] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Regulation of stem cells in adult tissues is a key determinant of how well an organism can respond to the stresses of physiological challenge and disease. This is particularly true of the hematopoietic system, where demands on host defenses can call for an acute increase in cell production. Hematopoietic stem cells receive the regulatory signals for cell production in adult mammals in the bone marrow, a tissue with higher-order architectural and functional organization than previously appreciated. Here, we review the data defining particular structural components and heterologous cells in the bone marrow that participate in hematopoietic stem cell function. Further, we explore the case for stromal-hematopoietic cell interactions contributing to neoplastic myeloid disease. As the hematopoietic regulatory networks in the bone marrow are revealed, it is anticipated that strategies will emerge for how to enhance or inhibit production of specific blood cells. In that way, the control of hematopoiesis will enter the domain of therapies to modulate broad aspects of hematopoiesis, both normal and malignant.
Collapse
Affiliation(s)
- Jonathan Hoggatt
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| | - Youmna Kfoury
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| | - David T Scadden
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| |
Collapse
|
42
|
Fibroblast growth factor 2 supports osteoblastic niche cells during hematopoietic homeostasis recovery after bone marrow suppression. Cell Commun Signal 2017; 15:25. [PMID: 28662672 PMCID: PMC5492158 DOI: 10.1186/s12964-017-0181-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 06/16/2017] [Indexed: 11/30/2022] Open
Abstract
Background Hematopoietic stem cell (HSC) maintenance requires a specific microenvironment. HSC niches can be activated by tissue damaging chemotherapeutic drugs and various cell signaling molecules such as SDF-1 and FGF, which might also result in bone marrow stress. Recent research has insufficiently shown that endosteal osteolineage cells and other niche constituents recover after marrow injury. Methods We investigated the role of FGF2 in the osteoblastic niche cells during hematopoietic homeostasis recovery after bone marrow injury. Mice were treated with 5-fluorouracil (5FU) to eliminate actively cycling cells in the bone marrow. Primary osteoblasts were isolated and subjected to cell culture. Real-time PCR, western blot and immunohistochemical staining were performed to study niche-related genes, osteoblast markers, and FGF2 signaling. Proliferation rate were analyzed by marker gene Ki67 and colony formation assay. Also, osterix-positive osteoprogenitor cells were isolated by FACS from Osx-GFP-Cre mice after 5FU treatment, and subjected to RNA-sequencing and analyzed for Fgf receptors and niche markers. Results The endosteal osteolineage cells isolated from 5FU-treated mice showed increased expression of the niche-related genes Sdf-1, Jagged-1, Scf, N-cad, Angpt1 and Vcam-1 and the osteoblast marker genes Osx, Opn, Runx2, and Alp, indicating that BM stress upon 5FU treatment activated the osteoblastic niche. Endosteal osteoblast expanded from a single layer to several layers 3 and 6 days after 5FU treatment. During the early recovery phase in 5FU-activated osteoblastic niches increased FGF2 expression and activated its downstream pERK. FGF2 treatment resulted in increased proliferation rate and the expression of niche marker genes in 5FU-activated osteoblastic niche cells. RNA-seq analysis in Osterix-positive osteoprogenitor cells isolated from 5FU-treated Osx-GFP mice showed significantly increased expression of Fgf receptors Fgfr1, 2 and 3. Although osteoblastic niche cells were damaged by 5FU treatment in the beginning, the increased number of OB layers in the recovery phase may be derived from resident osteoprogenitor cells by FGF2 activation under stress. Conclusions Taken together, FGF2 signaling can regulate osteoblastic niche cells to support HSC homeostasis in response to bone marrow damage.
Collapse
|
43
|
Osteoblasts support megakaryopoiesis through production of interleukin-9. Blood 2017; 129:3196-3209. [DOI: 10.1182/blood-2016-11-749838] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/19/2017] [Indexed: 12/16/2022] Open
Abstract
Key Points
Osteoblast-produced IL-9 supports megakaryopoiesis and platelet formation. IL-9 is a promising therapeutic agent for treatment of thrombocytopenia.
Collapse
|
44
|
Olivos DJ, Alvarez M, Cheng YH, Hooker RA, Ciovacco WA, Bethel M, McGough H, Yim C, Chitteti BR, Eleniste PP, Horowitz MC, Srour EF, Bruzzaniti A, Fuchs RK, Kacena MA. Lnk Deficiency Leads to TPO-Mediated Osteoclastogenesis and Increased Bone Mass Phenotype. J Cell Biochem 2017; 118:2231-2240. [PMID: 28067429 DOI: 10.1002/jcb.25874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/06/2017] [Indexed: 11/11/2022]
Abstract
The Lnk adapter protein negatively regulates the signaling of thrombopoietin (TPO), the main megakaryocyte (MK) growth factor. Lnk-deficient (-/-) mice have increased TPO signaling and increased MK number. Interestingly, several mouse models exist in which increased MK number leads to a high bone mass phenotype. Here we report the bone phenotype of these mice. MicroCT and static histomorphometric analyses at 20 weeks showed the distal femur of Lnk-/- mice to have significantly higher bone volume fraction and trabecular number compared to wild-type (WT) mice. Notably, despite a significant increase in the number of osteoclasts (OC), and decreased bone formation rate in Lnk-/- mice compared to WT mice, Lnk-/- mice demonstrated a 2.5-fold greater BV/TV suggesting impaired OC function in vivo. Additionally, Lnk-/- mouse femurs exhibited non-significant increases in mid-shaft cross-sectional area, yet increased periosteal BFR compared to WT femurs was observed. Lnk-/- femurs also had non-significant increases in polar moment of inertia and decreased cortical bone area and thickness, resulting in reduced bone stiffness, modulus, and strength compared to WT femurs. Of note, Lnk is expressed by OC lineage cells and when Lnk-/- OC progenitors are cultured in the presence of TPO, significantly more OC are observed than in WT cultures. Lnk is also expressed in osteoblast (OB) cells and in vitro reduced alkaline phosphatase activity was observed in Lnk-/- cultures. These data suggest that both direct effects on OB and OC as well as indirect effects of MK in regulating OB contributes to the observed high bone mass. J. Cell. Biochem. 118: 2231-2240, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David J Olivos
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Marta Alvarez
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ying-Hua Cheng
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Richard Adam Hooker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Wendy A Ciovacco
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| | - Monique Bethel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Haley McGough
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christopher Yim
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Pierre P Eleniste
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| | - Edward F Srour
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Angela Bruzzaniti
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Robyn K Fuchs
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, Indiana
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
45
|
|
46
|
Holen I. WITHDRAWN: The bone microenvironment – Multiple players involved in cancer progression. J Bone Oncol 2016. [DOI: 10.1016/j.jbo.2016.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
47
|
Hamed SA. Markers of bone turnover in patients with epilepsy and their relationship to management of bone diseases induced by antiepileptic drugs. Expert Rev Clin Pharmacol 2015; 9:267-286. [PMID: 26589104 DOI: 10.1586/17512433.2016.1123617] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Data from cross-sectional and prospective studies revealed that patients with epilepsy and on long-term treatment with antiepileptic drugs (AEDs) are at increased risk for metabolic bone diseases. Bone diseases were reported in about 50% of patients on AEDs. Low bone mineral density, osteopenia/osteoporosis, osteomalacia, rickets, altered concentration of bone turnover markers and fractures were reported with phenobarbital, phenytoin, carbamazepine, valproate, oxcarbazepine and lamotrigine. The mechanisms for AEDs-induced bone diseases are heterogeneous and include hypovitaminosis D, hypocalcemia and direct acceleration of bone loss and/or reduction of bone formation. This article reviews the evidence, predictors and mechanisms of AEDs-induced bone abnormalities and its clinical implications. For patients on AEDs, regular monitoring of bone health is recommended. Prophylactic administration of calcium and vitamin D is recommended for all patients. Treatment doses of calcium and vitamin D and even anti-resorptive drug therapy are reserved for patients at high risk of pathological fracture.
Collapse
Affiliation(s)
- Sherifa A Hamed
- a Department of Neurology and Psychiatry , Assiut University Hospital , Assiut , Egypt
| |
Collapse
|
48
|
Hamed SA. Markers of bone turnover in patients with epilepsy and their relationship to management of bone diseases induced by antiepileptic drugs. Expert Rev Clin Pharmacol 2015; 9:267-286. [PMID: 26589104 DOI: org/10.1586/17512433.2016.1123617] [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] [Indexed: 11/08/2022]
Abstract
Data from cross-sectional and prospective studies revealed that patients with epilepsy and on long-term treatment with antiepileptic drugs (AEDs) are at increased risk for metabolic bone diseases. Bone diseases were reported in about 50% of patients on AEDs. Low bone mineral density, osteopenia/osteoporosis, osteomalacia, rickets, altered concentration of bone turnover markers and fractures were reported with phenobarbital, phenytoin, carbamazepine, valproate, oxcarbazepine and lamotrigine. The mechanisms for AEDs-induced bone diseases are heterogeneous and include hypovitaminosis D, hypocalcemia and direct acceleration of bone loss and/or reduction of bone formation. This article reviews the evidence, predictors and mechanisms of AEDs-induced bone abnormalities and its clinical implications. For patients on AEDs, regular monitoring of bone health is recommended. Prophylactic administration of calcium and vitamin D is recommended for all patients. Treatment doses of calcium and vitamin D and even anti-resorptive drug therapy are reserved for patients at high risk of pathological fracture.
Collapse
Affiliation(s)
- Sherifa A Hamed
- a Department of Neurology and Psychiatry , Assiut University Hospital , Assiut , Egypt
| |
Collapse
|
49
|
Eleniste PP, Patel V, Posritong S, Zero O, Largura H, Cheng YH, Himes ER, Hamilton M, Ekwealor JTB, Kacena MA, Bruzzaniti A. Pyk2 and Megakaryocytes Regulate Osteoblast Differentiation and Migration Via Distinct and Overlapping Mechanisms. J Cell Biochem 2015; 117:1396-406. [PMID: 26552846 DOI: 10.1002/jcb.25430] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/09/2015] [Indexed: 01/08/2023]
Abstract
Osteoblast differentiation and migration are necessary for bone formation during bone remodeling. Mice lacking the proline-rich tyrosine kinase Pyk2 (Pyk2-KO) have increased bone mass, in part due to increased osteoblast proliferation. Megakaryocytes (MKs), the platelet-producing cells, also promote osteoblast proliferation in vitro and bone-formation in vivo via a pathway that involves Pyk2. In the current study, we examined the mechanism of action of Pyk2, and the role of MKs, on osteoblast differentiation and migration. We found that Pyk2-KO osteoblasts express elevated alkaline phosphatase (ALP), type I collagen and osteocalcin mRNA levels as well as increased ALP activity, and mineralization, confirming that Pyk2 negatively regulates osteoblast function. Since Pyk2 Y402 phosphorylation is important for its catalytic activity and for its protein-scaffolding functions, we expressed the phosphorylation-mutant (Pyk2(Y402F) ) and kinase-mutant (Pyk2(K457A) ) in Pyk2-KO osteoblasts. Both Pyk2(Y402F) and Pyk2(K457A) reduced ALP activity, whereas only kinase-inactive Pyk2(K457A) inhibited Pyk2-KO osteoblast migration. Consistent with a role for Pyk2 on ALP activity, co-culture of MKs with osteoblasts led to a decrease in the level of phosphorylated Pyk2 (pY402) as well as a decrease in ALP activity. Although, Pyk2-KO osteoblasts exhibited increased migration compared to wild-type osteoblasts, Pyk2 expression was not required necessary for the ability of MKs to stimulate osteoblast migration. Together, these data suggest that osteoblast differentiation and migration are inversely regulated by MKs via distinct Pyk2-dependent and independent signaling pathways. Novel drugs that distinguish between the kinase-dependent or protein-scaffolding functions of Pyk2 may provide therapeutic specificity for the control of bone-related diseases.
Collapse
Affiliation(s)
- Pierre P Eleniste
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Vruti Patel
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Sumana Posritong
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Odette Zero
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Heather Largura
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Ying-Hua Cheng
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Evan R Himes
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Matthew Hamilton
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jenna T B Ekwealor
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Angela Bruzzaniti
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| |
Collapse
|
50
|
Haider MT, Hunter KD, Robinson SP, Graham TJ, Corey E, Dear TN, Hughes R, Brown NJ, Holen I. Rapid modification of the bone microenvironment following short-term treatment with Cabozantinib in vivo. Bone 2015; 81:581-592. [PMID: 26279137 PMCID: PMC4768060 DOI: 10.1016/j.bone.2015.08.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/18/2015] [Accepted: 08/04/2015] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Bone metastasis remains incurable with treatment restricted to palliative care. Cabozantinib (CBZ) is targeted against multiple receptor tyrosine kinases involved in tumour pathobiology, including hepatocyte growth factor receptor (MET) and vascular endothelial growth factor receptor 2 (VEGFR-2). CBZ has demonstrated clinical activity in advanced prostate cancer with resolution of lesions visible on bone scans, implicating a potential role of the bone microenvironment as a mediator of CBZ effects. We characterised the effects of short-term administration of CBZ on bone in a range of in vivo models to determine how CBZ affects bone in the absence of tumour. METHODS Studies were performed in a variety of in vivo models including male and female BALB/c nude mice (age 6-17-weeks). Animals received CBZ (30 mg/kg, 5× weekly) or sterile H2O control for 5 or 10 days. Effects on bone integrity (μCT), bone cell activity (PINP, TRAP ELISA), osteoblast and osteoclast number/mm trabecular bone surface, area of epiphyseal growth plate cartilage, megakaryocyte numbers and bone marrow composition were assessed. Effects of longer-term treatment (15-day & 6-week administration) were assessed in male NOD/SCID and beige SCID mice. RESULTS CBZ treatment had significant effects on the bone microenvironment, including reduced osteoclast and increased osteoblast numbers compared to control. Trabecular bone structure was altered after 8 administrations. A significant elongation of the epiphyseal growth plate, in particular the hypertrophic chondrocyte zone, was observed in all CBZ treated animals irrespective of administration schedule. Both male and female BALB/c nude mice had increased megakaryocyte numbers/mm(2) tissue after 10-day CBZ treatment, in addition to vascular ectasia, reduced bone marrow cellularity and extravasation of red blood cells into the extra-vascular bone marrow. All CBZ-induced effects were transient and rapidly lost following cessation of treatment. CONCLUSION Short-term administration of CBZ induces rapid, reversible effects on the bone microenvironment in vivo highlighting a potential role in mediating treatment responses.
Collapse
Affiliation(s)
| | - Keith D Hunter
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK.
| | - Simon P Robinson
- CR-UK Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, Surrey, UK.
| | - Timothy J Graham
- CR-UK Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, Surrey, UK.
| | - Eva Corey
- Department of Urology, University of Washington Medical Center, Seattle, WA, USA.
| | - T Neil Dear
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| | - Russell Hughes
- Department of Oncology, University of Sheffield, Sheffield, UK.
| | - Nicola J Brown
- Department of Oncology, University of Sheffield, Sheffield, UK.
| | - Ingunn Holen
- Department of Oncology, University of Sheffield, Sheffield, UK.
| |
Collapse
|