201
|
Abstract
The Cre-LoxP technology permits gene ablation in specific cell lineages, at chosen differentiation stages of this lineage and in an inducible manner. It has allowed tremendous advances in our understanding of skeleton biology and related pathophysiological mechanisms, through the generation of loss/gain of function or cell tracing experiments based on the creation of an expanding toolbox of transgenic mice expressing the Cre recombinase in skeletal stem cells, chondrocytes, osteoblasts, or osteoclasts. In this chapter, we provide an overview of the different Cre-LoxP systems and Cre mouse lines used in the bone field, we discuss their advantages, limitations, and we outline best practices to interpret results obtained from the use of Cre mice.
Collapse
Affiliation(s)
- Florent Elefteriou
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Greig Couasnay
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
202
|
Kegelman CD, Nijsure MP, Moharrer Y, Pearson HB, Dawahare JH, Jordan KM, Qin L, Boerckel JD. YAP and TAZ Promote Periosteal Osteoblast Precursor Expansion and Differentiation for Fracture Repair. J Bone Miner Res 2021; 36:143-157. [PMID: 32835424 PMCID: PMC7988482 DOI: 10.1002/jbmr.4166] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022]
Abstract
In response to bone fracture, periosteal progenitor cells proliferate, expand, and differentiate to form cartilage and bone in the fracture callus. These cellular functions require the coordinated activation of multiple transcriptional programs, and the transcriptional regulators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) regulate osteochondroprogenitor activation during endochondral bone development. However, recent observations raise important distinctions between the signaling mechanisms used to control bone morphogenesis and repair. Here, we tested the hypothesis that YAP and TAZ regulate osteochondroprogenitor activation during endochondral bone fracture healing in mice. Constitutive YAP and/or TAZ deletion from Osterix-expressing cells impaired both cartilage callus formation and subsequent mineralization. However, this could be explained either by direct defects in osteochondroprogenitor differentiation after fracture or by developmental deficiencies in the progenitor cell pool before fracture. Consistent with the second possibility, we found that developmental YAP/TAZ deletion produced long bones with impaired periosteal thickness and cellularity. Therefore, to remove the contributions of developmental history, we next generated adult onset-inducible knockout mice (using Osx-CretetOff ) in which YAP and TAZ were deleted before fracture but after normal development. Adult onset-induced YAP/TAZ deletion had no effect on cartilaginous callus formation but impaired bone formation at 14 days post-fracture (dpf). Earlier, at 4 dpf, adult onset-induced YAP/TAZ deletion impaired the proliferation and expansion of osteoblast precursor cells located in the shoulder of the callus. Further, activated periosteal cells isolated from this region at 4 dpf exhibited impaired osteogenic differentiation in vitro upon YAP/TAZ deletion. Finally, confirming the effects on osteoblast function in vivo, adult onset-induced YAP/TAZ deletion impaired bone formation in the callus shoulder at 7 dpf before the initiation of endochondral ossification. Together, these data show that YAP and TAZ promote the expansion and differentiation of periosteal osteoblast precursors to accelerate bone fracture healing. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Christopher D Kegelman
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yasaman Moharrer
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Hope B Pearson
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - James H Dawahare
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Kelsey M Jordan
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
203
|
Rossetti R, Rós FA, Souza LEBD, Maçonetto JDM, Costa PNMD, Ferreira FU, Borges JS, Carvalho JVD, Morotti NP, Kashima S, Covas DT. Hypoxia-cultured mouse mesenchymal stromal cells from bone marrow and compact bone display different phenotypic traits. Exp Cell Res 2020; 399:112434. [PMID: 33340494 DOI: 10.1016/j.yexcr.2020.112434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/10/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022]
Abstract
It has been suggested that the bone marrow microenvironment harbors two distinct populations of mesenchymal stromal cells (MSC), one with a perivascular location and other present in the endosteum. A better understanding of the biology of these MSC subsets has been pursued in order to refine its clinical application. However, most comparative characterizations of mouse MSC have been performed in normoxia. This can result in misleading interpretations since mouse MSC subsets with low/defective p53 activity are known to be selected during culture in normoxia. Here, we report a comprehensive in vitro characterization of mouse MSC isolated from bone marrow (BM-MSC) and compact bone (CB-MSC) expanded and assayed under hypoxia for their morphology, clonogenic efficiency and differentiation capacity. We found that, under hypoxia, compact bone is richer in absolute numbers of MSC and isolation of MSC from compact bone is associated with a reduced risk of hematopoietic cell carryover. In addition, CB-MSC have higher in vitro osteogenic capacity than BM-MSC, while adipogenic differentiation potential is similar. These findings reinforce the hypothesis of the existence of MSC in bone marrow and compact bone representing functionally distinct cell populations and highlight the compact bone as an efficient source of murine MSC under physiological oxygen concentrations.
Collapse
Affiliation(s)
- Rafaela Rossetti
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil.
| | - Felipe Augusto Rós
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Lucas Eduardo Botelho de Souza
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Juliana de Matos Maçonetto
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Péricles Natan Mendes da Costa
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Fernanda Ursoli Ferreira
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Josiane Serrano Borges
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Julianne Vargas de Carvalho
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Nayara Patrícia Morotti
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Simone Kashima
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
204
|
Dysregulated Immune Responses by ASK1 Deficiency Alter Epithelial Progenitor Cell Fate and Accelerate Metaplasia Development during H. pylori Infection. Microorganisms 2020; 8:microorganisms8121995. [PMID: 33542169 PMCID: PMC7765114 DOI: 10.3390/microorganisms8121995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
The mechanism of H. pylori-induced atrophy and metaplasia has not been fully understood. Here, we demonstrate the novel role of Apoptosis signal-regulating kinase 1 (ASK1) and downstream MAPKs as a regulator of host immune responses and epithelial maintenance against H. pylori infection. ASK1 gene deficiency resulted in enhanced inflammation with numerous inflammatory cells including Gr-1+CD11b+ myeloid-derived suppressor cells (MDSCs) recruited into the infected stomach. Increase of IL-1β release from apoptotic macrophages and enhancement of TH1-polarized immune responses caused STAT1 and NF-κB activation in epithelial cells in ASK1 knockout mice. Dysregulated immune and epithelial activation in ASK1 knockout mice led to dramatic expansion of gastric progenitor cells and massive metaplasia development. Bone marrow transplantation experiments revealed that ASK1 in inflammatory cells is critical for inducing immune disorder and metaplastic changes in epithelium, while ASK1 in epithelial cells regulates cell proliferation in stem/progenitor zone without changes in inflammation and differentiation. These results suggest that H. pylori-induced immune cells may regulate epithelial homeostasis and cell fate as an inflammatory niche via ASK1 signaling.
Collapse
|
205
|
Gazit VA, Swietlicki EA, Liang MU, Surti A, McDaniel R, Geisman M, Alvarado DM, Ciorba MA, Bochicchio G, Ilahi O, Kirby J, Symons WJ, Davidson NO, Levin MS, Rubin DC. Stem cell and niche regulation in human short bowel syndrome. JCI Insight 2020; 5:137905. [PMID: 33141758 PMCID: PMC7714413 DOI: 10.1172/jci.insight.137905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 10/28/2020] [Indexed: 12/20/2022] Open
Abstract
Loss of functional small bowel surface area following surgical resection for disorders such as Crohn’s disease, intestinal ischemic injury, radiation enteritis, and in children, necrotizing enterocolitis, atresia, and gastroschisis, may result in short bowel syndrome, with attendant high morbidity, mortality, and health care costs in the United States. Following resection, the remaining small bowel epithelium mounts an adaptive response, resulting in increased crypt cell proliferation, increased villus height, increased crypt depth, and enhanced nutrient and electrolyte absorption. Although these morphologic and functional changes are well described in animal models, the adaptive response in humans is less well understood. Clinically the response is unpredictable and often inadequate. Here we address the hypotheses that human intestinal stem cell populations are expanded and that the stem cell niche is regulated following massive gut resection in short bowel syndrome (SBS). We use intestinal enteroid cultures from patients with SBS to show that the magnitude and phenotype of the adaptive stem cell response are both regulated by stromal niche cells, including intestinal subepithelial myofibroblasts, which are activated by intestinal resection to enhance epithelial stem and proliferative cell responses. Our data suggest that myofibroblast regulation of bone morphogenetic protein signaling pathways plays a role in the gut adaptive response after resection. LGR5+ stem cells are expanded and BMP signaling regulates the stem cell niche in human short bowel syndrome.
Collapse
Affiliation(s)
- Vered A Gazit
- Division of Gastroenterology, John T. Milliken Department of Medicine
| | | | - Miranda U Liang
- Division of Gastroenterology, John T. Milliken Department of Medicine
| | - Adam Surti
- Division of Gastroenterology, John T. Milliken Department of Medicine
| | - Raechel McDaniel
- Division of Gastroenterology, John T. Milliken Department of Medicine
| | - Mackenzie Geisman
- Division of Gastroenterology, John T. Milliken Department of Medicine
| | - David M Alvarado
- Division of Gastroenterology, John T. Milliken Department of Medicine
| | - Matthew A Ciorba
- Division of Gastroenterology, John T. Milliken Department of Medicine
| | | | | | | | | | - Nicholas O Davidson
- Division of Gastroenterology, John T. Milliken Department of Medicine.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marc S Levin
- Division of Gastroenterology, John T. Milliken Department of Medicine.,Veterans Affairs Medical Center, St. Louis, Missouri, USA
| | - Deborah C Rubin
- Division of Gastroenterology, John T. Milliken Department of Medicine.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
206
|
Tan Z, Kong M, Wen S, Tsang KY, Niu B, Hartmann C, Chan D, Hui CC, Cheah KSE. IRX3 and IRX5 Inhibit Adipogenic Differentiation of Hypertrophic Chondrocytes and Promote Osteogenesis. J Bone Miner Res 2020; 35:2444-2457. [PMID: 32662900 DOI: 10.1002/jbmr.4132] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/19/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Maintaining the correct proportions of different cell types in the bone marrow is critical for bone function. Hypertrophic chondrocytes (HCs) and osteoblasts are a lineage continuum with a minor contribution to adipocytes, but the regulatory network is unclear. Mutations in transcription factors, IRX3 and IRX5, result in skeletal patterning defects in humans and mice. We found coexpression of Irx3 and Irx5 in late-stage HCs and osteoblasts in cortical and trabecular bone. Irx3 and Irx5 null mutants display severe bone deficiency in newborn and adult stages. Quantitative analyses of bone with different combinations of functional alleles of Irx3 and Irx5 suggest these two factors function in a dosage-dependent manner. In Irx3 and Irx5 nulls, the amount of bone marrow adipocytes was increased. In Irx5 nulls, lineage tracing revealed that removal of Irx3 specifically in HCs exacerbated reduction of HC-derived osteoblasts and increased the frequency of HC-derived marrow adipocytes. β-catenin loss of function and gain of function specifically in HCs affects the expression of Irx3 and Irx5, suggesting IRX3 and IRX5 function downstream of WNT signaling. Our study shows that IRX3 and IRX5 regulate fate decisions in the transition of HCs to osteoblasts and to marrow adipocytes, implicating their potential roles in human skeletal homeostasis and disorders.
Collapse
Affiliation(s)
- Zhijia Tan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, HKSAR, China
| | - Mingpeng Kong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, HKSAR, China
| | - Songjia Wen
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, HKSAR, China
| | - Kwok Yeung Tsang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, HKSAR, China
| | - Ben Niu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, HKSAR, China
| | - Christine Hartmann
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Faculty of Medicine, University of Münster, Münster, Germany
| | - Danny Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, HKSAR, China
| | - Chi-Chung Hui
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Kathryn S E Cheah
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, HKSAR, China
| |
Collapse
|
207
|
Cao Y, Buckels EJ, Matthews BG. Markers for Identification of Postnatal Skeletal Stem Cells In Vivo. Curr Osteoporos Rep 2020; 18:655-665. [PMID: 33034805 DOI: 10.1007/s11914-020-00622-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The adult skeleton contains stem cells involved in growth, homeostasis, and healing. Mesenchymal or skeletal stem cells are proposed to provide precursors to osteoblasts, chondrocytes, marrow adipocytes, and stromal cells. We review the evidence for existence and functionality of different skeletal stem cell pools, and the tools available for identifying or targeting these populations in mouse and human tissues. RECENT FINDINGS Lineage tracing and single cell-based techniques in mouse models indicate that multiple pools of stem cells exist in postnatal bone. These include growth plate stem cells, stem and progenitor cells in the diaphysis, reticular cells that only form bone in response to injury, and injury-responsive periosteal stem cells. New staining protocols have also been described for prospective isolation of human skeletal stem cells. Several populations of postnatal skeletal stem and progenitor cells have been identified in mice, and we have an increasing array of tools to target these cells. Most Cre models lack a high degree of specificity to define single populations. Human studies are less advanced and require further efforts to refine methods for identifying stem and progenitor cells in adult bone.
Collapse
Affiliation(s)
- Ye Cao
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand
| | - Emma J Buckels
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand
| | - Brya G Matthews
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand.
| |
Collapse
|
208
|
Roelofs AJ, Kania K, Rafipay AJ, Sambale M, Kuwahara ST, Collins FL, Smeeton J, Serowoky MA, Rowley L, Wang H, Gronewold R, Kapeni C, Méndez-Ferrer S, Little CB, Bateman JF, Pap T, Mariani FV, Sherwood J, Crump JG, De Bari C. Identification of the skeletal progenitor cells forming osteophytes in osteoarthritis. Ann Rheum Dis 2020; 79:1625-1634. [PMID: 32963046 PMCID: PMC8136618 DOI: 10.1136/annrheumdis-2020-218350] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Osteophytes are highly prevalent in osteoarthritis (OA) and are associated with pain and functional disability. These pathological outgrowths of cartilage and bone typically form at the junction of articular cartilage, periosteum and synovium. The aim of this study was to identify the cells forming osteophytes in OA. METHODS Fluorescent genetic cell-labelling and tracing mouse models were induced with tamoxifen to switch on reporter expression, as appropriate, followed by surgery to induce destabilisation of the medial meniscus. Contributions of fluorescently labelled cells to osteophytes after 2 or 8 weeks, and their molecular identity, were analysed by histology, immunofluorescence staining and RNA in situ hybridisation. Pdgfrα-H2BGFP mice and Pdgfrα-CreER mice crossed with multicolour Confetti reporter mice were used for identification and clonal tracing of mesenchymal progenitors. Mice carrying Col2-CreER, Nes-CreER, LepR-Cre, Grem1-CreER, Gdf5-Cre, Sox9-CreER or Prg4-CreER were crossed with tdTomato reporter mice to lineage-trace chondrocytes and stem/progenitor cell subpopulations. RESULTS Articular chondrocytes, or skeletal stem cells identified by Nes, LepR or Grem1 expression, did not give rise to osteophytes. Instead, osteophytes derived from Pdgfrα-expressing stem/progenitor cells in periosteum and synovium that are descendants from the Gdf5-expressing embryonic joint interzone. Further, we show that Sox9-expressing progenitors in periosteum supplied hybrid skeletal cells to the early osteophyte, while Prg4-expressing progenitors from synovial lining contributed to cartilage capping the osteophyte, but not to bone. CONCLUSION Our findings reveal distinct periosteal and synovial skeletal progenitors that cooperate to form osteophytes in OA. These cell populations could be targeted in disease modification for treatment of OA.
Collapse
Affiliation(s)
- Anke J Roelofs
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Karolina Kania
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Alexandra J Rafipay
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Meike Sambale
- Institute of Musculoskeletal Medicine, University Hospital Munster, Munster, Germany
| | - Stephanie T Kuwahara
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Fraser L Collins
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Joanna Smeeton
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Maxwell A Serowoky
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Lynn Rowley
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Hui Wang
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - René Gronewold
- Institute of Musculoskeletal Medicine, University Hospital Munster, Munster, Germany
| | - Chrysa Kapeni
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge, UK
| | - Simón Méndez-Ferrer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge, UK
| | - Christopher B Little
- Raymond Purves Bone and Joint Laboratories, Kolling Institute of Medical Research, The University of Sydney, St Leonards, New South Wales, Australia
| | - John F Bateman
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Thomas Pap
- Institute of Musculoskeletal Medicine, University Hospital Munster, Munster, Germany
| | - Francesca V Mariani
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Joanna Sherwood
- Institute of Musculoskeletal Medicine, University Hospital Munster, Munster, Germany
| | - J Gage Crump
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Cosimo De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
209
|
LOXL2 promotes aggrecan and gender-specific anabolic differences to TMJ cartilage. Sci Rep 2020; 10:20179. [PMID: 33214607 PMCID: PMC7678826 DOI: 10.1038/s41598-020-77178-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 11/05/2020] [Indexed: 12/24/2022] Open
Abstract
In the United States, 5–12% of adults have at least one symptom of temporomandibular joint (TMJ) disorders, including TMJ osteoarthritis (TMJ-OA). However, there is no chondroprotective agent that is approved for clinical application. We showed that LOXL2 is elevated in the regenerative response during fracture healing in mice and has a critical role in chondrogenic differentiation. Indeed, LOXL2 is an anabolic effector that attenuates pro-inflammatory signaling in OA cartilage of the TMJ and knee joint, induces chondroprotective and regenerative responses, and attenuates NF-kB signaling. The specific goal of the study was to evaluate if adenoviral delivery of LOXL2 is anabolic to human and mouse TMJ condylar cartilage in vivo and evaluate the protective and anabolic effect on cartilage-specific factors. We employed two different models to assess TMJ-OA. In one model, clinical TMJ-OA cartilage from 5 different samples in TMJ-OA cartilage plugs were implanted subcutaneously in nude mice. Adenovirus LOXL2 -treated implants showed higher mRNA levels of LOXL2, ACAN, and other anabolic genes compared to the adenovirus-Empty-treated implants. Further characterization by RNA-seq analysis showed LOXL2 promotes proteoglycan networks and extracellular matrix in human TMJ-OA cartilage implants in vivo. In order to evaluate if LOXL2-induced functional and sex-linked differences, both male and female four-month-old chondrodysplasia (Cho/+) mice, which develop progressive TMJ-OA due to a point mutation in the Col11a1 gene, were subjected to intraperitoneal injection with Adv-RFP-LOXL2 every 2 weeks for 12 weeks. The data showed that adenovirus delivery of LOXL2 upregulated LOXL2 and aggrecan (Acan), whereas MMP13 expression was slightly downregulated. The fold change expression of Acan and Runx2 induced by Adv-RFP-LOXL2 was higher in females compared to males. Interestingly, Adv-RFP-LOXL2 injection significantly increased Rankl expression in male but there was no change in females, whereas VegfB gene expression was increased in females, but not in males, as compared to those injected with Adv-RFP-Empty in respective groups. Our findings indicate that LOXL2 can induce specifically the expression of Acan and other anabolic genes in two preclinical models in vivo. Further, LOXL2 has beneficial functions in human TMJ-OA cartilage implants and promotes gender-specific anabolic responses in Cho/+ mice with progressive TMJ-OA, suggesting its merit for further study as an anabolic therapy for TMJ-OA.
Collapse
|
210
|
Oliveira TC, Gomes MS, Gomes AC. The Crossroads between Infection and Bone Loss. Microorganisms 2020; 8:microorganisms8111765. [PMID: 33182721 PMCID: PMC7698271 DOI: 10.3390/microorganisms8111765] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 01/18/2023] Open
Abstract
Bone homeostasis, based on a tight balance between bone formation and bone degradation, is affected by infection. On one hand, some invading pathogens are capable of directly colonizing the bone, leading to its destruction. On the other hand, immune mediators produced in response to infection may dysregulate the deposition of mineral matrix by osteoblasts and/or the resorption of bone by osteoclasts. Therefore, bone loss pathologies may develop in response to infection, and their detection and treatment are challenging. Possible biomarkers of impaired bone metabolism during chronic infection need to be identified to improve the diagnosis and management of infection-associated osteopenia. Further understanding of the impact of infections on bone metabolism is imperative for the early detection, prevention, and/or reversion of bone loss. Here, we review the mechanisms responsible for bone loss as a direct and/or indirect consequence of infection.
Collapse
Affiliation(s)
- Tiago Carvalho Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Faculdade de Ciências da Universidade do Porto, 4169-007 Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar da Universidade do Porto, 4050-313 Porto, Portugal
| | - Maria Salomé Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Instituto de Ciências Biomédicas de Abel Salazar da Universidade do Porto, 4050-313 Porto, Portugal
| | - Ana Cordeiro Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Correspondence:
| |
Collapse
|
211
|
Matsushita Y, Ono W, Ono N. Bone regeneration via skeletal cell lineage plasticity: All hands mobilized for emergencies: Quiescent mature skeletal cells can be activated in response to injury and robustly participate in bone regeneration through cellular plasticity. Bioessays 2020; 43:e2000202. [PMID: 33155283 DOI: 10.1002/bies.202000202] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/16/2020] [Accepted: 09/24/2020] [Indexed: 12/17/2022]
Abstract
An emerging concept is that quiescent mature skeletal cells provide an important cellular source for bone regeneration. It has long been considered that a small number of resident skeletal stem cells are solely responsible for the remarkable regenerative capacity of adult bones. However, recent in vivo lineage-tracing studies suggest that all stages of skeletal lineage cells, including dormant pre-adipocyte-like stromal cells in the marrow, osteoblast precursor cells on the bone surface and other stem and progenitor cells, are concomitantly recruited to the injury site and collectively participate in regeneration of the damaged skeletal structure. Lineage plasticity appears to play an important role in this process, by which mature skeletal cells can transform their identities into skeletal stem cell-like cells in response to injury. These highly malleable, long-living mature skeletal cells, readily available throughout postnatal life, might represent an ideal cellular resource that can be exploited for regenerative medicine.
Collapse
Affiliation(s)
- Yuki Matsushita
- University of Michigan School of Dentistry, Ann Arbor, Michigan, 48109, USA
| | - Wanida Ono
- University of Michigan School of Dentistry, Ann Arbor, Michigan, 48109, USA
| | - Noriaki Ono
- University of Michigan School of Dentistry, Ann Arbor, Michigan, 48109, USA
| |
Collapse
|
212
|
Tevlin R, Longaker MT, Wan DC. Skeletal Stem Cells-A Paradigm Shift in the Field of Craniofacial Bone Tissue Engineering. FRONTIERS IN DENTAL MEDICINE 2020; 1:596706. [PMID: 35664558 PMCID: PMC9161996 DOI: 10.3389/fdmed.2020.596706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Defects of the craniofacial skeleton arise as a direct result of trauma, diseases, oncological resection, or congenital anomalies. Current treatment options are limited, highlighting the importance for developing new strategies to restore form, function, and aesthetics of missing or damaged bone in the face and the cranium. For optimal reconstruction, the goal is to replace "like with like." With the inherent challenges of existing options, there is a clear need to develop alternative strategies to reconstruct the craniofacial skeleton. The success of mesenchymal stem cell-based approaches has been hampered by high heterogeneity of transplanted cell populations with inconsistent preclinical and clinical trial outcomes. Here, we discuss the novel characterization and isolation of mouse skeletal stem cell (SSC) populations and their response to injury, systemic disease, and how their re-activation in vivo can contribute to tissue regeneration. These studies led to the characterization of human SSCs which are able to self-renew, give rise to increasingly fate restricted progenitors, and differentiate into bone, cartilage, and bone marrow stroma, all on the clonal level in vivo without prior in vitro culture. SSCs hold great potential for implementation in craniofacial bone tissue engineering and regenerative medicine. As we begin to better understand the diversity and the nature of skeletal stem and progenitor cells, there is a tangible future whereby a subset of human adult SSCs can be readily purified from bone or activated in situ with broad potential applications in craniofacial tissue engineering.
Collapse
Affiliation(s)
- Ruth Tevlin
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Derrick C. Wan
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
213
|
Huang S, Jin M, Su N, Chen L. New insights on the reparative cells in bone regeneration and repair. Biol Rev Camb Philos Soc 2020; 96:357-375. [PMID: 33051970 DOI: 10.1111/brv.12659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
Bone possesses a remarkable repair capacity to regenerate completely without scar tissue formation. This unique characteristic, expressed during bone development, maintenance and injury (fracture) healing, is performed by the reparative cells including skeletal stem cells (SSCs) and their descendants. However, the identity and functional roles of SSCs remain controversial due to technological difficulties and the heterogeneity and plasticity of SSCs. Moreover, for many years, there has been a biased view that bone marrow is the main cell source for bone repair. Together, these limitations have greatly hampered our understanding of these important cell populations and their potential applications in the treatment of fractures and skeletal diseases. Here, we reanalyse and summarize current understanding of the reparative cells in bone regeneration and repair and outline recent progress in this area, with a particular emphasis on the temporal and spatial process of fracture healing, the sources of reparative cells, an updated definition of SSCs, and markers of skeletal stem/progenitor cells contributing to the repair of craniofacial and long bones, as well as the debate between SSCs and pericytes. Finally, we also discuss the existing problems, emerging novel technologies and future research directions in this field.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| |
Collapse
|
214
|
Sanjurjo-Rodriguez C, Altaie A, Mastbergen S, Baboolal T, Welting T, Lafeber F, Pandit H, McGonagle D, Jones E. Gene Expression Signatures of Synovial Fluid Multipotent Stromal Cells in Advanced Knee Osteoarthritis and Following Knee Joint Distraction. Front Bioeng Biotechnol 2020; 8:579751. [PMID: 33178674 PMCID: PMC7591809 DOI: 10.3389/fbioe.2020.579751] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Osteoarthritis (OA) is the most common musculoskeletal disorder. Although joint replacement remains the standard of care for knee OA patients, knee joint distraction (KJD), which works by temporarily off-loading the joint for 6–8 weeks, is becoming a novel joint-sparing alternative for younger OA sufferers. The biological mechanisms behind KJD structural improvements remain poorly understood but likely involve joint-resident regenerative cells including multipotent stromal cells (MSCs). In this study, we hypothesized that KJD leads to beneficial cartilage-anabolic and anti-catabolic changes in joint-resident MSCs and investigated gene expression profiles of synovial fluid (SF) MSCs following KJD as compared with baseline. To obtain further insights into the effects of local biomechanics on MSCs present in late OA joints, SF MSC gene expression was studied in a separate OA arthroplasty cohort and compared with subchondral bone (SB) MSCs from medial (more loaded) and lateral (less loaded) femoral condyles from the same joints. In OA arthroplasty cohort (n = 12 patients), SF MSCs expressed lower levels of ossification- and hypotrophy-related genes [bone sialoprotein (IBSP), parathyroid hormone 1 receptor (PTH1R), and runt-related transcription factor 2 (RUNX2)] than did SB MSCs. Interestingly, SF MSCs expressed 5- to 50-fold higher levels of transcripts for classical extracellular matrix turnover molecules matrix metalloproteinase 1 (MMP1), a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5), and tissue inhibitor of metalloproteinase-3 (TIMP3), all (p < 0.05) potentially indicating greater cartilage remodeling ability of OA SF MSCs, compared with SB MSCs. In KJD cohort (n = 9 patients), joint off-loading resulted in sustained, significant increase in SF MSC colonies’ sizes and densities and a notable transcript upregulation of key cartilage core protein aggrecan (ACAN) (weeks 3 and 6), as well as reduction in pro-inflammatory C–C motif chemokine ligand 2 (CCL2) expression (weeks 3 and 6). Additionally, early KJD changes (week 3) were marked by significant increases in MSC chondrogenic commitment markers gremlin 1 (GREM1) and growth differentiation factor 5 (GDF5). In combination, our results reveal distinct transcriptomes on joint-resident MSCs from different biomechanical environments and show that 6-week joint off-loading leads to transcriptional changes in SF MSCs that may be beneficial for cartilage regeneration. Biomechanical factors should be certainly considered in the development of novel MSC-based therapies for OA.
Collapse
Affiliation(s)
- Clara Sanjurjo-Rodriguez
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Physiotherapy, Medicine and Biomedical Sciences department, CIBER-BBN, Institute of Biomedical Research of A Coruña (INIBIC)-Centre of Advanced Scientific Researches (CICA), University of A Coruña, A Coruña, Spain
| | - Ala Altaie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Simon Mastbergen
- University Medical Center Utrecht, Rheumatology & Clinical Immunology, Regenerative Medicine Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Thomas Baboolal
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Tim Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Floris Lafeber
- University Medical Center Utrecht, Rheumatology & Clinical Immunology, Regenerative Medicine Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Hemant Pandit
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
215
|
Esposito A, Wang L, Li T, Miranda M, Spagnoli A. Role of Prx1-expressing skeletal cells and Prx1-expression in fracture repair. Bone 2020; 139:115521. [PMID: 32629173 PMCID: PMC7484205 DOI: 10.1016/j.bone.2020.115521] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
The healing capacity of bones after fracture implies the existence of adult regenerative cells. However, information on identification and functional role of fracture-induced progenitors is still lacking. Paired-related homeobox 1 (Prx1) is expressed during skeletogenesis. We hypothesize that fracture recapitulates Prx1's expression, and Prx1 expressing cells are critical to induce repair. To address our hypothesis, we used a combination of in vivo and in vitro approaches, short and long-term cell tracking analyses of progenies and actively expressing cells, cell ablation studies, and rodent animal models for normal and defective fracture healing. We found that fracture elicits a periosteal and endosteal response of perivascular Prx1+ cells that participate in fracture healing and showed that Prx1-expressing cells have a functional role in the repair process. While Prx1-derived cells contribute to the callus, Prx1's expression decreases concurrently with differentiation into cartilaginous and bone cells, similarly to when Prx1+ cells are cultured in differentiating conditions. We determined that bone morphogenic protein 2 (BMP2), through C-X-C motif-ligand-12 (CXCL12) signaling, modulates the downregulation of Prx1. We demonstrated that fracture elicits an early increase in BMP2 expression, followed by a decrease in CXCL12 that in turn down-regulates Prx1, allowing cells to commit to osteochondrogenesis. In vivo and in vitro treatment with CXCR4 antagonist AMD3100 restored Prx1 expression by modulating the BMP2-CXCL12 axis. Our studies represent a shift in the current research that has primarily focused on the identification of markers for postnatal skeletal progenitors, and instead we characterized the function of a specific population (Prx1+ cells) and their expression marker (Prx1) as a crossroad in fracture repair. The identification of fracture-induced perivascular Prx1+ cells and regulation of Prx1's expression by BMP2 and in turn by CXCL12 in the orchestration of fracture repair, highlights a pathway in which to investigate defective mechanisms and therapeutic targets for fracture non-union.
Collapse
Affiliation(s)
- Alessandra Esposito
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Lai Wang
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Tieshi Li
- Department of Pediatrics, University of Nebraska Medical Center, Children's Hospital & Medical Center, Omaha, NE, USA
| | - Mariana Miranda
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Anna Spagnoli
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA; Department of Pediatrics, Division of Pediatric Endocrinology, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
216
|
Wolock SL, Krishnan I, Tenen DE, Matkins V, Camacho V, Patel S, Agarwal P, Bhatia R, Tenen DG, Klein AM, Welner RS. Mapping Distinct Bone Marrow Niche Populations and Their Differentiation Paths. Cell Rep 2020; 28:302-311.e5. [PMID: 31291568 DOI: 10.1016/j.celrep.2019.06.031] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/05/2019] [Accepted: 06/07/2019] [Indexed: 12/21/2022] Open
Abstract
The bone marrow microenvironment is composed of heterogeneous cell populations of non-hematopoietic cells with complex phenotypes and undefined trajectories of maturation. Among them, mesenchymal cells maintain the production of stromal, bone, fat, and cartilage cells. Resolving these unique cellular subsets within the bone marrow remains challenging. Here, we used single-cell RNA sequencing of non-hematopoietic bone marrow cells to define specific subpopulations. Furthermore, by combining computational prediction of the cell state hierarchy with the known expression of key transcription factors, we mapped differentiation paths to the osteocyte, chondrocyte, and adipocyte lineages. Finally, we validated our findings using lineage-specific reporter strains and targeted knockdowns. Our analysis reveals differentiation hierarchies for maturing stromal cells, determines key transcription factors along these trajectories, and provides an understanding of the complexity of the bone marrow microenvironment.
Collapse
Affiliation(s)
- Samuel L Wolock
- Department of System Biology, Harvard Medical School, Boston, MA, USA
| | - Indira Krishnan
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Danielle E Tenen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Victoria Matkins
- Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Virginia Camacho
- Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sweta Patel
- Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Puneet Agarwal
- Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ravi Bhatia
- Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel G Tenen
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA; Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Allon M Klein
- Department of System Biology, Harvard Medical School, Boston, MA, USA
| | - Robert S Welner
- Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
217
|
Dysregulated Bone Metabolism Is Related to High Expression of miR-151a-3p in Severe Adolescent Idiopathic Scoliosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4243015. [PMID: 33029507 PMCID: PMC7537684 DOI: 10.1155/2020/4243015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/14/2020] [Indexed: 12/05/2022]
Abstract
Adolescent idiopathic scoliosis (AIS) is a common complex disease, and bone homeostasis plays an important role in its pathogenesis. Recent advances in epigenetic research show that dysregulated miRNAs may participate in the development of orthopedic diseases and AIS. The aim of this study was to detect differentially expressed miRNAs in severe AIS and elucidate the mechanism of miRNA deregulation in the pathogenesis of AIS. In the present study, miRNA expression profiles were detected in severe and mild AIS patients as well as healthy controls by miRNA sequencing. Candidate miRNAs were validated in a larger cohort. Primary osteoblasts from severe AIS patients were extracted and isolated to determine the effect of the candidate miRNAs on bone metabolism. Finally, we determined the methylation level in primary osteoblasts from severe AIS patients. The result showed that miR-151a-3p was overexpressed in severe AIS patients. Reduced GREM1 expression was observed in primary osteoblasts from severe AIS patients. miR-151a-3p directly inhibited GREM1 in primary osteoblasts. Relatively lower methylation levels were detected in primary osteoblasts from severe AIS patients. In conclusion, our study revealed that plasma miR-151a-3p levels may serve as a biomarker for severe AIS. Overexpression of miR-151a-3p may interrupt bone homeostasis via inhibiting GREM1 expression. Our result may provide a new biomarker for the early detection of AIS and increase our understanding of the pathogenesis of AIS.
Collapse
|
218
|
Single-cell and spatial transcriptomics approaches of the bone marrow microenvironment. Curr Opin Oncol 2020; 32:146-153. [PMID: 31833957 DOI: 10.1097/cco.0000000000000602] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE OF REVIEW The bone marrow is home to hematopoietic stem cells responsible for lifelong blood production, alongside mesenchymal stem cells required for skeletal regeneration. In the bone marrow, a unique combination of signals derived from a multitude of cell types results in the establishment of so-called niches that regulate stem-cell maintenance and differentiation. Recently, single-cell and spatially resolved transcriptomics technologies have been utilized to characterize the murine bone marrow microenvironment during homeostasis, stress and upon cancer-induced remodeling. In this review, we summarize the major findings of these studies. RECENT FINDINGS Single-cell technologies applied to bone marrow provided the first systematic and label-free identification of bone marrow cell types, enabled their molecular and spatial characterization, and clarified the cellular sources of key prohematopoietic factors. Large transcriptional heterogeneity and novel subpopulations were observed in compartments previously thought to be homogenous. For example, Lepr Cxcl12-abundant reticular cells were shown to constitute the major source of prohematopoietic factors, but consist of subpopulations differing in their adipogenic versus osteogenic priming, morphology and localization. These subpopulations were suggested to act as professional cytokine secreting cells, thereby establishing distinct bone marrow niches. SUMMARY Single-cell and spatially resolved transcriptomics approaches have clarified the molecular identity and localization of bone marrow-resident cell types, paving the road for a deeper exploration of bone marrow niches in the mouse and humans.
Collapse
|
219
|
Ortinau LC, Wang H, Lei K, Deveza L, Jeong Y, Hara Y, Grafe I, Rosenfeld SB, Lee D, Lee B, Scadden DT, Park D. Identification of Functionally Distinct Mx1+αSMA+ Periosteal Skeletal Stem Cells. Cell Stem Cell 2020; 25:784-796.e5. [PMID: 31809737 DOI: 10.1016/j.stem.2019.11.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/11/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022]
Abstract
The periosteum is critical for bone maintenance and healing. However, the in vivo identity and specific regulatory mechanisms of adult periosteum-resident skeletal stem cells are unknown. Here, we report animal models that selectively and durably label postnatal Mx1+αSMA+ periosteal stem cells (P-SSCs) and establish that P-SSCs are a long-term repopulating, functionally distinct SSC subset responsible for lifelong generation of periosteal osteoblasts. P-SSCs rapidly migrate toward an injury site, supply osteoblasts and chondrocytes, and recover new periosteum. Notably, P-SSCs specifically express CCL5 receptors, CCR3 and CCR5. Real-time intravital imaging revealed that the treatment with CCL5 induces P-SSC migration in vivo and bone healing, while CCL5/CCR5 deletion, CCR5 inhibition, or local P-SSC ablation reduces osteoblast number and delays bone healing. Human periosteal cells express CCR5 and undergo CCL5-mediated migration. Thus, the adult periosteum maintains genetically distinct SSC subsets with a CCL5-dependent migratory mechanism required for bone maintenance and injury repair.
Collapse
Affiliation(s)
- Laura C Ortinau
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hamilton Wang
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Kevin Lei
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Lorenzo Deveza
- Department of Orthopedic Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Youngjae Jeong
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yannis Hara
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ingo Grafe
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Scott B Rosenfeld
- Texas Children's Hospital, 6701 Fannin Street, Houston, TX 77030, USA
| | - Dongjun Lee
- Department of Convergence of Medical Science, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Brendan Lee
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - David T Scadden
- Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Dongsu Park
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
220
|
Scott RW, Arostegui M, Schweitzer R, Rossi FMV, Underhill TM. Hic1 Defines Quiescent Mesenchymal Progenitor Subpopulations with Distinct Functions and Fates in Skeletal Muscle Regeneration. Cell Stem Cell 2020; 25:797-813.e9. [PMID: 31809738 DOI: 10.1016/j.stem.2019.11.004] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/10/2019] [Accepted: 11/11/2019] [Indexed: 01/01/2023]
Abstract
Many adult tissues contain resident stem cells, such as the Pax7+ satellite cells within skeletal muscle, that regenerate parenchymal elements following damage. Tissue-resident mesenchymal progenitors (MPs) also participate in regeneration, although their function and fate in this process are unclear. Here, we identify Hypermethylated in cancer 1 (Hic1) as a marker of MPs in skeletal muscle and further show that Hic1 deletion leads to MP hyperplasia. Single-cell RNA-seq and ATAC-seq analysis of Hic1+ MPs in skeletal muscle shows multiple subpopulations, which we further show have distinct functions and lineage potential. Hic1+ MPs orchestrate multiple aspects of skeletal muscle regeneration by providing stage-specific immunomodulation and trophic and mechanical support. During muscle regeneration, Hic1+ derivatives directly contribute to several mesenchymal compartments including Col22a1-expressing cells within the myotendinous junction. Collectively, these findings demonstrate that HIC1 regulates MP quiescence and identifies MP subpopulations with transient and enduring roles in muscle regeneration.
Collapse
Affiliation(s)
- R Wilder Scott
- Department of Cellular and Physiological Sciences, 2222 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada; School of Biomedical Engineering and the Biomedical Research Centre, 2222 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Martin Arostegui
- Department of Cellular and Physiological Sciences, 2222 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Ronen Schweitzer
- Research Division, Shriners Hospital for Children, Portland, OR 97239, USA
| | - Fabio M V Rossi
- Department of Medical Genetics, 2222 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada; School of Biomedical Engineering and the Biomedical Research Centre, 2222 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - T Michael Underhill
- Department of Cellular and Physiological Sciences, 2222 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada; School of Biomedical Engineering and the Biomedical Research Centre, 2222 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
221
|
Julien A, Perrin S, Duchamp de Lageneste O, Carvalho C, Bensidhoum M, Legeai-Mallet L, Colnot C. FGFR3 in Periosteal Cells Drives Cartilage-to-Bone Transformation in Bone Repair. Stem Cell Reports 2020; 15:955-967. [PMID: 32916123 PMCID: PMC7561512 DOI: 10.1016/j.stemcr.2020.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/18/2022] Open
Abstract
Most organs and tissues in the body, including bone, can repair after an injury due to the activation of endogenous adult stem/progenitor cells to replace the damaged tissue. Inherent dysfunctions of the endogenous stem/progenitor cells in skeletal repair disorders are still poorly understood. Here, we report that Fgfr3Y637C/+ over-activating mutation in Prx1-derived skeletal stem/progenitor cells leads to failure of fracture consolidation. We show that periosteal cells (PCs) carrying the Fgfr3Y637C/+ mutation can engage in osteogenic and chondrogenic lineages, but following transplantation do not undergo terminal chondrocyte hypertrophy and transformation into bone causing pseudarthrosis. Instead, Prx1Cre;Fgfr3Y637C/+ PCs give rise to fibrocartilage and fibrosis. Conversely, wild-type PCs transplanted at the fracture site of Prx1Cre;Fgfr3Y637C/+ mice allow hypertrophic cartilage transition to bone and permit fracture consolidation. The results thus highlight cartilage-to-bone transformation as a necessary step for bone repair and FGFR3 signaling within PCs as a key regulator of this transformation. Fgfr3Y367C activating mutation in skeletal stem/progenitor cells prevents bone healing Intrinsic deficiencies in transplanted Prx1Cre;Fgfr3Y637C/+ PCs cause pseudarthrosis Prx1Cre;Fgfr3Y637C/+ PCs cannot support cartilage-to-bone transformation Wild-type PCs can rescue the Prx1Cre;Fgfr3Y637C/+ pseudarthrosis phenotype
Collapse
Affiliation(s)
- Anais Julien
- Paris University, Imagine Institute, INSERM UMR 1163, 75015, Paris, France
| | - Simon Perrin
- Paris University, Imagine Institute, INSERM UMR 1163, 75015, Paris, France
| | | | - Caroline Carvalho
- Paris University, Imagine Institute, INSERM UMR 1163, 75015, Paris, France
| | - Morad Bensidhoum
- Paris university, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging (B3OA), UMR CNRS 7052, INSERM 1271
| | - Laurence Legeai-Mallet
- Paris University, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, 75015, Paris, France
| | - Céline Colnot
- Paris University, Imagine Institute, INSERM UMR 1163, 75015, Paris, France.
| |
Collapse
|
222
|
Salhotra A, Shah HN, Levi B, Longaker MT. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol 2020; 21:696-711. [PMID: 32901139 DOI: 10.1038/s41580-020-00279-w] [Citation(s) in RCA: 540] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
Abstract
Bone development occurs through a series of synchronous events that result in the formation of the body scaffold. The repair potential of bone and its surrounding microenvironment - including inflammatory, endothelial and Schwann cells - persists throughout adulthood, enabling restoration of tissue to its homeostatic functional state. The isolation of a single skeletal stem cell population through cell surface markers and the development of single-cell technologies are enabling precise elucidation of cellular activity and fate during bone repair by providing key insights into the mechanisms that maintain and regenerate bone during homeostasis and repair. Increased understanding of bone development, as well as normal and aberrant bone repair, has important therapeutic implications for the treatment of bone disease and ageing-related degeneration.
Collapse
Affiliation(s)
- Ankit Salhotra
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Harsh N Shah
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
223
|
Spatial Distributions, Characteristics, and Applications of Craniofacial Stem Cells. Stem Cells Int 2020; 2020:8868593. [PMID: 32908545 PMCID: PMC7475745 DOI: 10.1155/2020/8868593] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 02/05/2023] Open
Abstract
Stem cells play an irreplaceable role in the development, homeostasis, and regeneration of the craniofacial bone. Multiple populations of tissue-resident craniofacial skeletal stem cells have been identified in different stem cell niches, including the cranial periosteum, jawbone marrow, temporomandibular joint, cranial sutures, and periodontium. These cells exhibit self-renewal and multidirectional differentiation abilities. Here, we summarized the properties of craniofacial skeletal stem cells, based on their spatial distribution. Specifically, we focused on the in vivo genetic fate mapping of stem cells, by exploring specific stem cell markers and observing their lineage commitment in both the homeostatic and regenerative states. Finally, we discussed their application in regenerative medicine.
Collapse
|
224
|
Thorup AS, Dell'Accio F, Eldridge SE. Lessons from joint development for cartilage repair in the clinic. Dev Dyn 2020; 250:360-376. [PMID: 32738003 DOI: 10.1002/dvdy.228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
More than 250 years ago, William Hunter stated that when cartilage is destroyed it never recovers. In the last 20 years, the understanding of the mechanisms that lead to joint formation and the knowledge that some of these mechanisms are reactivated in the homeostatic responses of cartilage to injury has offered an unprecedented therapeutic opportunity to achieve cartilage regeneration. Very large investments in ambitious clinical trials are finally revealing that, although we do not have perfect medicines yet, disease modification is a feasible possibility for human osteoarthritis.
Collapse
Affiliation(s)
- Anne-Sophie Thorup
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Suzanne E Eldridge
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
225
|
Li H, Wang H, Pan J, Li J, Zhang K, Duan W, Liang H, Chen K, Geng D, Shi Q, Yang H, Li B, Chen H. Nanoscaled Bionic Periosteum Orchestrating the Osteogenic Microenvironment for Sequential Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:36823-36836. [PMID: 32706234 DOI: 10.1021/acsami.0c06906] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Periosteum orchestrates bone repair. Previously developed artificial periosteum was mainly focusing on materials modification to simply enhance bone formation, but few were attempting to make the artificial periosteum fit different bone repair stages. Here, we constructed a functionalized periosteum, which was composed of an electrospun scaffold grafted with leptin receptor antibody (LepR-a) and BMP2-loaded hollow MnO2 (h-MnO2) nanoparticles through a polydopamine (PDA)-assisted technique. The bionic periosteum showed suitable mechanical properties and favorable biocompatibility. It effectively recruited skeletal stem cells (SSCs) through antigen-antibody interactions, as in in vitro cell adhesion tests, we observed that more SSCs attached to the LepR-a-grafted periosteum compared to the control group. In vivo, the LepR-a-grafted periosteum covered on the cranial defect in Prx1-Cre/ERT2, -EGFP mice recruited more Prx1-EGFP cells to the fracture site compared to control groups at post-surgery day 3, 7, and 14. Co-staining with Sp7 indicated that most of the recruited Prx1-EGFP cells underwent osteogenic lineage commitment. Sustained BMP2 release from h-MnO2 promoted osteogenesis by accelerating the osteogenic differentiation of recruited SSCs, as demonstrated by alkaline phosphatase (ALP) and alizarin red staining (ARS) in vitro and microcomputed tomography (micro-CT) in vivo. Interestingly, we also observed the growth of osteogenic coupled capillaries (CD31hiEmcnhi) in the bone repair site, which might be induced by increased platelet-derived growth factor-BB (PDGF-BB) in the regenerative microenvironment subsequent to SSCs' differentiation. Taken together, the findings from this study indicate that the multifunctionalized periosteum efficiently recruited and motivated the SSCs in vivo and orchestrated the osteogenic microenvironment for bone repair in a sequence manner. Thus, the construction of the bionic periosteum to couple with natural bone regeneration stages has been demonstrated to be effective in facilitating bone healing.
Collapse
Affiliation(s)
- Hanwen Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Huan Wang
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Jun Pan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Jiaying Li
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Kai Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Weifeng Duan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Huan Liang
- Medical College, Yangzhou University, 136 Jiangyang Road, Yangzhou, Jiangsu 225009, P. R. China
| | - Kangwu Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Dechun Geng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Qin Shi
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Bin Li
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Hao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
- Medical College, Yangzhou University, 136 Jiangyang Road, Yangzhou, Jiangsu 225009, P. R. China
| |
Collapse
|
226
|
Articular cartilage regeneration by activated skeletal stem cells. Nat Med 2020; 26:1583-1592. [PMID: 32807933 DOI: 10.1038/s41591-020-1013-2] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease resulting in irreversible, progressive destruction of articular cartilage1. The etiology of OA is complex and involves a variety of factors, including genetic predisposition, acute injury and chronic inflammation2-4. Here we investigate the ability of resident skeletal stem-cell (SSC) populations to regenerate cartilage in relation to age, a possible contributor to the development of osteoarthritis5-7. We demonstrate that aging is associated with progressive loss of SSCs and diminished chondrogenesis in the joints of both mice and humans. However, a local expansion of SSCs could still be triggered in the chondral surface of adult limb joints in mice by stimulating a regenerative response using microfracture (MF) surgery. Although MF-activated SSCs tended to form fibrous tissues, localized co-delivery of BMP2 and soluble VEGFR1 (sVEGFR1), a VEGF receptor antagonist, in a hydrogel skewed differentiation of MF-activated SSCs toward articular cartilage. These data indicate that following MF, a resident stem-cell population can be induced to generate cartilage for treatment of localized chondral disease in OA.
Collapse
|
227
|
Koliaraki V, Prados A, Armaka M, Kollias G. The mesenchymal context in inflammation, immunity and cancer. Nat Immunol 2020; 21:974-982. [PMID: 32747813 DOI: 10.1038/s41590-020-0741-2] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022]
Abstract
Mesenchymal cells are mesoderm-derived stromal cells that are best known for providing structural support to organs, synthesizing and remodeling the extracellular matrix (ECM) and regulating development, homeostasis and repair of tissues. Recent detailed mechanistic insights into the biology of fibroblastic mesenchymal cells have revealed they are also significantly involved in immune regulation, stem cell maintenance and blood vessel function. It is now becoming evident that these functions, when defective, drive the development of complex diseases, such as various immunopathologies, chronic inflammatory disease, tissue fibrosis and cancer. Here, we provide a concise overview of the contextual contribution of fibroblastic mesenchymal cells in physiology and disease and bring into focus emerging evidence for both their heterogeneity at the single-cell level and their tissue-specific, spatiotemporal functional diversity.
Collapse
Affiliation(s)
- Vasiliki Koliaraki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| | - Alejandro Prados
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Marietta Armaka
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - George Kollias
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece. .,Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece. .,Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
228
|
Targeted Disruption of Bone Marrow Stromal Cell-Derived Gremlin1 Limits Multiple Myeloma Disease Progression In Vivo. Cancers (Basel) 2020; 12:cancers12082149. [PMID: 32756430 PMCID: PMC7464474 DOI: 10.3390/cancers12082149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 12/14/2022] Open
Abstract
In most instances, multiple myeloma (MM) plasma cells (PCs) are reliant on factors made by cells of the bone marrow (BM) stroma for their survival and growth. To date, the nature and cellular composition of the BM tumor microenvironment and the critical factors which drive tumor progression remain imprecisely defined. Our studies show that Gremlin1 (Grem1), a highly conserved protein, which is abundantly secreted by a subset of BM mesenchymal stromal cells, plays a critical role in MM disease development. Analysis of human and mouse BM stromal samples by quantitative PCR showed that GREM1/Grem1 expression was significantly higher in the MM tumor-bearing cohorts compared to healthy controls (p < 0.05, Mann–Whitney test). Additionally, BM-stromal cells cultured with 5TGM1 MM PC line expressed significantly higher levels of Grem1, compared to stromal cells alone (p < 0.01, t-test), suggesting that MM PCs promote increased Grem1 expression in stromal cells. Furthermore, the proliferation of 5TGM1 MM PCs was found to be significantly increased when co-cultured with Grem1-overexpressing stromal cells (p < 0.01, t-test). To examine the role of Grem1 in MM disease in vivo, we utilized the 5TGM1/KaLwRij mouse model of MM. Our studies showed that, compared to immunoglobulin G (IgG) control antibody-treated mice, mice treated with an anti-Grem1 neutralizing antibody had a decrease in MM tumor burden of up to 81.2% (p < 0.05, two-way ANOVA). The studies presented here demonstrate, for the first time, a novel positive feedback loop between MM PCs and BM stroma, and that inhibiting this vicious cycle with a neutralizing antibody can dramatically reduce tumor burden in a preclinical mouse model of MM.
Collapse
|
229
|
Gilliam EA, Schlieve CR, Fowler KL, Rea JN, Schall KA, Huang S, Spence JR, Grikscheit TC. Grading TESI: Crypt and villus formation in tissue-engineered small intestine alters with stem/progenitor cell source. Am J Physiol Gastrointest Liver Physiol 2020; 319:G261-G279. [PMID: 32597710 DOI: 10.1152/ajpgi.00387.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The small intestine has a remarkable ability to enhance its absorptive and digestive surface area through the formation of villi, a process known as villification. We sought to learn whether developing mouse and human tissue-engineered small intestine (TESI) followed known developmental biology routes to villification, such as Sonic hedgehog (SHH)/Indian hedgehog (IHH) and bone morphogenetic protein 4 (BMP4)/forkhead box F1 (FOXF1) signaling to identify targets to enhance the development of TESI. After generating TESI from prenatal and postnatal stem cell sources, we evaluated the effect of cell source derivation on villification with a grading scheme to approximate developmental stage. χ2 analysis compared the prevalence of TESI grade from each stem cell source. RNAscope probes detected genes known to direct villification and the development of the crypt-villus axis in mouse and human development. These were compared in TESI derived from various pluripotent and progenitor cell donor cell types as well as native human fetal and postnatal tissues. Prenatal and pluripotent cell sources form mature villus and crypt-like structures more frequently than postnatal donor sources, and there are alternate routes to villus formation. Human TESI recapitulates epithelial to mesenchymal crosstalk of several genes identified in development, with fetal and pluripotent donor-derived TESI arriving at villus formation following described developmental patterns. However, postnatal TESI is much less likely to form complete villus-crypt patterns and demonstrates alternate SHH/IHH and BMP4/FOXF1 signaling patterns. Grading TESI and other cellular constructs may assist discoveries to support future human therapies.NEW & NOTEWORTHY The small intestine can enhance its absorptive and digestive surface area through a process known as villification. Tissue-engineered small intestine achieves mature villification at varying levels of success between differing sources. We have developed a consistent grading schema of morphology and characterized it across multiple developmental pathways, allowing objective comparison between differing constructs and sources.
Collapse
Affiliation(s)
- Elizabeth A Gilliam
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Christopher R Schlieve
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Kathryn L Fowler
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California
| | - Jessica N Rea
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Kathy A Schall
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Sha Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jason R Spence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan.,Program of Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Tracy C Grikscheit
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California.,Keck Medical School, University of Southern California, Los Angeles, California
| |
Collapse
|
230
|
Lukač N, Katavić V, Novak S, Šućur A, Filipović M, Kalajzić I, Grčević D, Kovačić N. What do we know about bone morphogenetic proteins and osteochondroprogenitors in inflammatory conditions? Bone 2020; 137:115403. [PMID: 32371019 DOI: 10.1016/j.bone.2020.115403] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/10/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Osteochondroprogenitors are crucial for embryonic bone development and postnatal processes such as bone repair in response to fracture injury, and their dysfunction may contribute to insufficient repair of structural damage in inflammatory arthritides. In the fracture healing, the early inflammatory phase is crucial for normal callus development and new bone formation. This process involves a complex interplay of many molecules and cell types, responsible for recruitment, expansion and differentiation of osteochondroprogenitor populations. In inflammatory arthritides, inflammation induces bone resorption and causes insufficient bone formation, which leads to local and systemic bone loss. While bone loss is a predominant feature in rheumatoid arthritis, inflammation also induces pathologic bone formation at enthesial sites in seronegative spondyloarthropathies. Bone morphogenetic proteins (BMP) are involved in cell proliferation, differentiation and apoptosis, and have fundamental roles in maintenance of postnatal bone homeostasis. They are crucial regulators of the osteochondroprogenitor pool and drive their proliferation, differentiation, and lifespan during bone regeneration. In this review, we summarize the effects of inflammation on osteochondroprogenitor populations during fracture repair and in inflammatory arthritides, with special focus on inflammation-mediated modulation of BMP signaling. We also present data in which we describe a population of murine synovial osteochondroprogenitor cells, which are reduced in arthritis, and characterize their expression of genes involved in regulation of bone homeostasis, emphasizing the up-regulation of BMP pathways in early progenitor subset. Based on the presented data, it may be concluded that during an inflammatory response, innate immune cells induce osteochondroprogenitors by providing signals for their recruitment, by producing BMPs and other osteogenic factors for paracrine effects, and by secreting inflammatory cytokines that may positively regulate osteogenic pathways. On the other hand, inflammatory cells may secrete cytokines that interfere with osteogenic pathways, proapoptotic factors that reduce the pool of osteochondroprogenitor cells, as well as BMP and Wnt antagonists. The net effect is strongly context-dependent and influenced by the local milieu of cells, cytokines, and growth factors. Further elucidation of the interplay between inflammatory signals and BMP-mediated bone formation may provide valuable tools for therapeutic targeting.
Collapse
Affiliation(s)
- Nina Lukač
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vedran Katavić
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sanja Novak
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Alan Šućur
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Maša Filipović
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivo Kalajzić
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Danka Grčević
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia.
| |
Collapse
|
231
|
Oya Y, Hayakawa Y, Koike K. Tumor microenvironment in gastric cancers. Cancer Sci 2020; 111:2696-2707. [PMID: 32519436 PMCID: PMC7419059 DOI: 10.1111/cas.14521] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment favors the growth and expansion of cancer cells. Many cell types are involved in the tumor microenvironment such as inflammatory cells, fibroblasts, nerves, and vascular endothelial cells. These stromal cells contribute to tumor growth by releasing various molecules to either directly activate the growth signaling in cancer cells or remodel surrounding areas. This review introduces recent advances in findings on the interactions within the tumor microenvironment such as in cancer-associated fibroblasts (CAFs), immune cells, and endothelial cells, in particular those established in mouse gastric cancer models. In mice, myofibroblasts in the gastric stroma secrete R-spondin and support normal gastric stem cells. Most CAFs promote tumor growth in a paracrine manner, but CAF population appears to be heterogeneous in terms of their function and origin, and include both tumor-promoting and tumor-restraining populations. Among immune cell populations, tumor-associated macrophages, including M1 and M2 macrophages, and myeloid-derived suppressor cells (MDSCs), are reported to directly or indirectly promote gastric tumorigenesis by secreting soluble factors or modulating immune responses. Endothelial cells or blood vessels not only fuel tumors with nutrients, but also interact with cancer stem cells and immune cells by secreting chemokines or cytokines, and act as a cancer niche. Understanding these interactions within the tumor microenvironment would contribute to unraveling new therapeutic targets.
Collapse
Affiliation(s)
- Yukiko Oya
- Department of GastroenterologyGraduate school of Medicinethe University of TokyoTokyoJapan
| | - Yoku Hayakawa
- Department of GastroenterologyGraduate school of Medicinethe University of TokyoTokyoJapan
| | - Kazuhiko Koike
- Department of GastroenterologyGraduate school of Medicinethe University of TokyoTokyoJapan
| |
Collapse
|
232
|
Todd GM, Gao Z, Hyvönen M, Brazil DP, Ten Dijke P. Secreted BMP antagonists and their role in cancer and bone metastases. Bone 2020; 137:115455. [PMID: 32473315 DOI: 10.1016/j.bone.2020.115455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/23/2020] [Accepted: 05/23/2020] [Indexed: 02/08/2023]
Abstract
Bone morphogenetic proteins (BMPs) are multifunctional secreted cytokines that act in a highly context-dependent manner. BMP action extends beyond the induction of cartilage and bone formation, to encompass pivotal roles in controlling tissue and organ homeostasis during development and adulthood. BMPs signal via plasma membrane type I and type II serine/threonine kinase receptors and intracellular SMAD transcriptional effectors. Exquisite temporospatial control of BMP/SMAD signalling and crosstalk with other cellular cues is achieved by a series of positive and negative regulators at each step in the BMP/SMAD pathway. The interaction of BMP ligand with its receptors is carefully controlled by a diverse set of secreted antagonists that bind BMPs and block their interaction with their cognate BMP receptors. Perturbations in this BMP/BMP antagonist balance are implicated in a range of developmental disorders and diseases, including cancer. Here, we provide an overview of the structure and function of secreted BMP antagonists, and summarize recent novel insights into their role in cancer progression and bone metastasis. Gremlin1 (GREM1) is a highly studied BMP antagonist, and we will focus on this molecule in particular and its role in cancer. The therapeutic potential of pharmacological inhibitors for secreted BMP antagonists for cancer and other human diseases will also be discussed.
Collapse
Affiliation(s)
- Grace M Todd
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland, UK
| | - Zhichun Gao
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland, UK
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK.
| | - Derek P Brazil
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland, UK.
| | - Peter Ten Dijke
- Oncode Institute, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
233
|
Oichi T, Otsuru S, Usami Y, Enomoto-Iwamoto M, Iwamoto M. Wnt signaling in chondroprogenitors during long bone development and growth. Bone 2020; 137:115368. [PMID: 32380258 PMCID: PMC7354209 DOI: 10.1016/j.bone.2020.115368] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 02/08/2023]
Abstract
Wnt signaling together with other signaling pathways governs cartilage development and the growth plate function during long bone formation and growth. β-catenin-dependent Wnt signaling is a specific lineage determinant of skeletal mesenchymal cells toward chondrogenic or osteogenic direction. Once cartilage forms and the growth plate organize, Wnt signaling continues to regulate proliferation and differentiation of the growth plate chondrocytes. Although chondrocytes in the growth plate have a high capacity to proliferate, new cells must be supplied to the growth plate from chondroprogenitor population. Advances in in vivo cell tracking techniques have demonstrated the importance of Wnt signaling in driving tissue renewal. The Wnt-responsive cells, genetically marked by the Wnt-reporter system, are found as stem cells in various tissues. Similarly, Wnt-responsive cells are found in the periphery of the growth plate and expanded to constitute entire column structure, indicating that Wnt signaling participates in the regulation of chondroprogenitors in the growth plate. This review will discuss advancements in research of progenitors in the growth plate, specifically focusing on Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Takeshi Oichi
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Satoru Otsuru
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Yu Usami
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Masahiro Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
234
|
Bone marrow fat: friend or foe in people with diabetes mellitus? Clin Sci (Lond) 2020; 134:1031-1048. [PMID: 32337536 DOI: 10.1042/cs20200220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 12/22/2022]
Abstract
Global trends in the prevalence of overweight and obesity put the adipocyte in the focus of huge medical interest. This review highlights a new topic in adipose tissue biology, namely the emerging pathogenic role of fat accumulation in bone marrow (BM). Specifically, we summarize current knowledge about the origin and function of BM adipose tissue (BMAT), provide evidence for the association of excess BMAT with diabetes and related cardiovascular complications, and discuss potential therapeutic approaches to correct BMAT dysfunction. There is still a significant uncertainty about the origins and function of BMAT, although several subpopulations of stromal cells have been suggested to have an adipogenic propensity. BM adipocytes are higly plastic and have a distinctive capacity to secrete adipokines that exert local and endocrine functions. BM adiposity is abundant in elderly people and has therefore been interpreted as a component of the whole-body ageing process. BM senescence and BMAT accumulation has been also reported in patients and animal models with Type 2 diabetes, being more pronounced in those with ischaemic complications. Understanding the mechanisms responsible for excess and altered function of BMAT could lead to new treatments able to preserve whole-body homeostasis.
Collapse
|
235
|
Kurenkova AD, Medvedeva EV, Newton PT, Chagin AS. Niches for Skeletal Stem Cells of Mesenchymal Origin. Front Cell Dev Biol 2020; 8:592. [PMID: 32754592 PMCID: PMC7366157 DOI: 10.3389/fcell.2020.00592] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
With very few exceptions, all adult tissues in mammals are maintained and can be renewed by stem cells that self-renew and generate the committed progeny required. These functions are regulated by a specific and in many ways unique microenvironment in stem cell niches. In most cases disruption of an adult stem cell niche leads to depletion of stem cells, followed by impairment of the ability of the tissue in question to maintain its functions. The presence of stem cells, often referred to as mesenchymal stem cells (MSCs) or multipotent bone marrow stromal cells (BMSCs), in the adult skeleton has long been realized. In recent years there has been exceptional progress in identifying and characterizing BMSCs in terms of their capacity to generate specific types of skeletal cells in vivo. Such BMSCs are often referred to as skeletal stem cells (SSCs) or skeletal stem and progenitor cells (SSPCs), with the latter term being used throughout this review. SSPCs have been detected in the bone marrow, periosteum, and growth plate and characterized in vivo on the basis of various genetic markers (i.e., Nestin, Leptin receptor, Gremlin1, Cathepsin-K, etc.). However, the niches in which these cells reside have received less attention. Here, we summarize the current scientific literature on stem cell niches for the SSPCs identified so far and discuss potential factors and environmental cues of importance in these niches in vivo. In this context we focus on (i) articular cartilage, (ii) growth plate cartilage, (iii) periosteum, (iv) the adult endosteal compartment, and (v) the developing endosteal compartment, in that order.
Collapse
Affiliation(s)
- Anastasiia D Kurenkova
- Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Ekaterina V Medvedeva
- Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Phillip T Newton
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Andrei S Chagin
- Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
236
|
The osteogenic commitment of CD271+CD56+ bone marrow stromal cells (BMSCs) in osteoarthritic femoral head bone. Sci Rep 2020; 10:11145. [PMID: 32636407 PMCID: PMC7341749 DOI: 10.1038/s41598-020-67998-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA), the most common joint disorder, is characterised by progressive structural changes in both the cartilage and the underlying subchondral bone. In late disease stages, subchondral bone sclerosis has been linked to heightened osteogenic commitment of bone marrow stromal cells (BMSCs). This study utilised cell sorting and immunohistochemistry to identify a phenotypically-distinct, osteogenically-committed BMSC subset in human OA trabecular bone. Femoral head trabecular bone tissue digests were sorted into CD45-CD271+CD56+CD146-, CD45-CD271+CD56-CD146+ and CD45-CD271+CD56-CD146-(termed double-negative, DN) subsets, and CD45+CD271-hematopoietic-lineage cells served as control. Compared to the CD146+ subset, the CD56+ subset possessed a lower-level expression of adipocyte-associated genes and significantly over 100-fold higher-level expression of many osteoblast-related genes including osteopontin and osteocalcin, whilst the DN subset presented a transcriptionally ‘intermediate’ BMSC population. All subsets were tri-potential following culture-expansion and were present in control non-OA trabecular bone. However, while in non-OA bone CD56+ cells only localised on the bone surface, in OA bone they were additionally present in the areas of new bone formation rich in osteoblasts and newly-embedded osteocytes. In summary, this study reveals a distinct osteogenically-committed CD271+CD56+ BMSC subset and implicates it in subchondral bone sclerosis in hip OA. CD271+CD56+ subset may represent a future therapeutic target for OA and other bone-associated pathologies.
Collapse
|
237
|
Doherty L, Sanjay A. LGRs in Skeletal Tissues: An Emerging Role for Wnt-Associated Adult Stem Cell Markers in Bone. JBMR Plus 2020; 4:e10380. [PMID: 32666024 PMCID: PMC7340442 DOI: 10.1002/jbm4.10380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/18/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023] Open
Abstract
Leucine-rich repeat-containing G protein-coupled receptors (LGRs) are adult stem cell markers that have been described across various stem cell niches, and expression of LGRs and their corresponding ligands (R-spondins) has now been reported in multiple bone-specific cell types. The skeleton harbors elusive somatic stem cell populations that are exceedingly compartment-specific and under tight regulation from various signaling pathways. Skeletal progenitors give rise to multiple tissues during development and during regenerative processes of bone, requiring postnatal endochondral and intramembranous ossification. The relevance of LGRs and the LGR/R-spondin ligand interaction in bone and tooth biology is becoming increasingly appreciated. LGRs may define specific stem cell and progenitor populations and their behavior during both development and regeneration, and their role as Wnt-associated receptors with specific ligands poses these proteins as unique therapeutic targets via potential R-spondin agonism. This review seeks to outline the current literature on LGRs in the context of bone and its associated tissues, and points to key future directions for studying the functional role of LGRs and ligands in skeletal biology. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic SurgeryUConn HealthFarmingtonCTUSA
| | - Archana Sanjay
- Department of Orthopaedic SurgeryUConn HealthFarmingtonCTUSA
| |
Collapse
|
238
|
Hu B, Lv X, Chen H, Xue P, Gao B, Wang X, Zhen G, Crane JL, Pan D, Liu S, Ni S, Wu P, Su W, Liu X, Ling Z, Yang M, Deng R, Li Y, Wang L, Zhang Y, Wan M, Shao Z, Chen H, Yuan W, Cao X. Sensory nerves regulate mesenchymal stromal cell lineage commitment by tuning sympathetic tones. J Clin Invest 2020; 130:3483-3498. [PMID: 32191640 PMCID: PMC7324175 DOI: 10.1172/jci131554] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 03/11/2020] [Indexed: 12/30/2022] Open
Abstract
The sensory nerve was recently identified as being involved in regulation of bone mass accrual. We previously discovered that prostaglandin E2 (PGE2) secreted by osteoblasts could activate sensory nerve EP4 receptor to promote bone formation by inhibiting sympathetic activity. However, the fundamental units of bone formation are active osteoblasts, which originate from mesenchymal stromal/stem cells (MSCs). Here, we found that after sensory denervation, knockout of the EP4 receptor in sensory nerves, or knockout of COX-2 in osteoblasts, could significantly promote adipogenesis and inhibit osteogenesis in adult mice. Furthermore, injection of SW033291 (a small molecule that locally increases the PGE2 level) or propranolol (a beta blocker) significantly promoted osteogenesis and inhibited adipogenesis. This effect of SW033291, but not propranolol, was abolished in conditional EP4-KO mice under normal conditions or in the bone repair process. We conclude that the PGE2/EP4 sensory nerve axis could regulate MSC differentiation in bone marrow of adult mice.
Collapse
Affiliation(s)
- Bo Hu
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
- Section of Spine Surgery, Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiao Lv
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Chen
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Peng Xue
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bo Gao
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiao Wang
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gehua Zhen
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Janet L. Crane
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Dayu Pan
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shen Liu
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shuangfei Ni
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Panfeng Wu
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Weiping Su
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiaonan Liu
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zemin Ling
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mi Yang
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ruoxian Deng
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yusheng Li
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ying Zhang
- Section of Spine Surgery, Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huajiang Chen
- Section of Spine Surgery, Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wen Yuan
- Section of Spine Surgery, Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
239
|
Selective deletion of MyD88 signaling in α-SMA positive cells ameliorates experimental intestinal fibrosis via post-transcriptional regulation. Mucosal Immunol 2020; 13:665-678. [PMID: 32020030 PMCID: PMC7316631 DOI: 10.1038/s41385-020-0259-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 02/04/2023]
Abstract
Intestinal fibrosis leading to strictures remains a significant clinical problem in inflammatory bowel diseases (IBD). The role of bacterial components in activating intestinal mesenchymal cells and driving fibrogenesis is largely unexplored. Tamoxifen-inducible α-SMA promoter Cre mice crossed with floxed MyD88 mice were subjected to chronic dextran sodium sulfate colitis. MyD88 was deleted prior to or after induction of colitis. Human intestinal myofibroblasts (HIMF) were exposed to various bacterial components and assessed for fibronectin (FN) and collagen I (Col1) production. RNA sequencing was performed. Post-transcriptional regulation was assessed by polysome profiling assay. Selective deletion of MyD88 in α-SMA-positive cells prior to, but not after induction of, experimental colitis decreased the degree of intestinal fibrosis. HIMF selectively responded to flagellin with enhanced FN or Col1 protein production in a MyD88-dependent manner. RNA sequencing suggested minimal transcriptional changes induced by flagellin in HIMF. Polysome profiling revealed higher proportions of FN and Col1 mRNA in the actively translated fractions of flagellin exposed HIMF, which was mediated by eIF2 alpha and 4EBP1. In conclusion, selectivity of flagellin-induced ECM secretion in HIMF is post-transcriptionally regulated. The results may represent a novel and targetable link between the gut microbiota and intestinal fibrogenesis.
Collapse
|
240
|
Zheng G, Xie ZY, Wang P, Wu YF, Shen HY. Recent advances of single-cell RNA sequencing technology in mesenchymal stem cell research. World J Stem Cells 2020; 12:438-447. [PMID: 32742561 PMCID: PMC7360991 DOI: 10.4252/wjsc.v12.i6.438] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/13/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells with great potential for clinical applications. However, little is known about their cell heterogeneity at a single-cell resolution, which severely impedes the development of MSC therapy. In this review, we focus on advances in the identification of novel surface markers and functional subpopulations of MSCs made by single-cell RNA sequencing and discuss their participation in the pathophysiology of stem cells and related diseases. The challenges and future directions of single-cell RNA sequencing in MSCs are also addressed in this review.
Collapse
Affiliation(s)
- Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Zhong-Yu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Yan-Feng Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| | - Hui-Yong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| |
Collapse
|
241
|
Zhang D, Zhang S, Wang J, Li Q, Xue H, Sheng R, Xiong Q, Qi X, Wen J, Fan Y, Zhou B, Yuan Q. LepR-Expressing Stem Cells Are Essential for Alveolar Bone Regeneration. J Dent Res 2020; 99:1279-1286. [PMID: 32585118 DOI: 10.1177/0022034520932834] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cells play a critical role in bone regeneration. Multiple populations of skeletal stem cells have been identified in long bone, while their identity and functions in alveolar bone remain unclear. Here, we identified a quiescent leptin receptor–expressing (LepR+) cell population that contributed to intramembranous bone formation. Interestingly, these LepR+ cells became activated in response to tooth extraction and generated the majority of the newly formed bone in extraction sockets. In addition, genetic ablation of LepR+ cells attenuated extraction socket healing. The parabiosis experiments revealed that the LepR+ cells in the healing sockets were derived from resident tissue rather than peripheral blood circulation. Further studies on the mechanism suggested that these LepR+ cells were responsive to parathyroid hormone/parathyroid hormone 1 receptor (PTH/PTH1R) signaling. Collectively, we demonstrate that LepR+ cells, a postnatal skeletal stem cell population, are essential for alveolar bone regeneration of extraction sockets.
Collapse
Affiliation(s)
- D. Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - S. Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J. Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q. Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H. Xue
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - R. Sheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q. Xiong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X. Qi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J. Wen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y. Fan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - B.O. Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Q. Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
242
|
Gooding S, Leedham SJ. Gremlin 1 - small protein, big impact: the multiorgan consequences of disrupted BMP antagonism †. J Pathol 2020; 251:349-352. [PMID: 32472605 PMCID: PMC8576570 DOI: 10.1002/path.5479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 12/31/2022]
Abstract
Highly conserved, complex and interacting morphogen signalling pathways regulate adult stem cells and control cell fate determination across numerous different organs. In homeostasis, the bone morphogenetic protein (BMP) pathway predominantly promotes cell differentiation. Localised expression of ligand sequestering BMP antagonists, such as Gremlin 1 (Grem1), necessarily restricts BMP activity within the stem cell niche and facilitate stemness and self‐renewal. In a new paper, Rowan, Jahns et al show that acute deletion of Grem1 in adult mice, using a ubiquitous ROSA26‐Cre recombinase, induced not only severe intestinal enteropathy but also hypocellular bone marrow failure suggestive of stem cell niche collapse in both tissues. Grem1 has an increasingly recognised pleiotrophic role in a number of organ systems and is implicated across a wide range of disease states. Although the importance of Grem1 in intestinal stem cell regulation has been well described, a putative function in haematopoietic niche maintenance is novel and requires further exploration. Moreover, the complex and context‐specific regulation of Grem1, among a host of functionally convergent but structurally disparate BMP antagonists, warrants further research as we learn more about the pathogenic consequences of deranged expression of this small, but important, protein. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sarah Gooding
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Simon J Leedham
- Intestinal Stem Cell Biology Laboratory, Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
243
|
Sharma T, Cotney J, Singh V, Sanjay A, Reichenberger EJ, Ueki Y, Maye P. Investigating global gene expression changes in a murine model of cherubism. Bone 2020; 135:115315. [PMID: 32165349 PMCID: PMC7305689 DOI: 10.1016/j.bone.2020.115315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/25/2020] [Accepted: 03/08/2020] [Indexed: 11/22/2022]
Abstract
Cherubism is a rare genetic disorder caused primarily by mutations in SH3BP2 resulting in excessive bone resorption and fibrous tissue overgrowth in the lower portions of the face. Bone marrow derived cell cultures derived from a murine model of cherubism display poor osteogenesis and spontaneous osteoclast formation. To develop a deeper understanding for the potential underlying mechanisms contributing to these phenotypes in mice, we compared global gene expression changes in hematopoietic and mesenchymal cell populations between cherubism and wild type mice. In the hematopoietic population, not surprisingly, upregulated genes were significantly enriched for functions related to osteoclastogenesis. However, these upregulated genes were also significantly enriched for functions associated with inflammation including arachidonic acid/prostaglandin signaling, regulators of coagulation and autoinflammation, extracellular matrix remodeling, and chemokine expression. In the mesenchymal population, we observed down regulation of osteoblast and adventitial reticular cell marker genes. Regulators of BMP and Wnt pathway associated genes showed numerous changes in gene expression, likely implicating the down regulation of BMP signaling and possibly the activation of certain Wnt pathways. Analyses of the cherubism derived mesenchymal population also revealed interesting changes in gene expression related to inflammation including the expression of distinct granzymes, chemokines, and sulfotransferases. These studies reveal complex changes in gene expression elicited from a cherubic mutation in Sh3bp2 that are informative to the mechanisms responding to inflammatory stimuli and repressing osteogenesis. The outcomes of this work are likely to have relevance not only to cherubism, but other inflammatory conditions impacting the skeleton.
Collapse
Affiliation(s)
- Tulika Sharma
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health, United States of America
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut Health, United States of America
| | - Vijender Singh
- Computational Biology Core, Institute for Systems Genomics, University of Connecticut, United States of America
| | - Archana Sanjay
- Department of Orthopedic Surgery, University of Connecticut Health, United States of America
| | - Ernst J Reichenberger
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health, United States of America
| | - Yasuyoshi Ueki
- Department of Biomedical Sciences and Comprehensive Care, Indiana University, United States of America
| | - Peter Maye
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health, United States of America.
| |
Collapse
|
244
|
Rowan SC, Jahns H, Mthunzi L, Piouceau L, Cornwell J, Doody R, Frohlich S, Callanan JJ, McLoughlin P. Gremlin 1 depletion in vivo causes severe enteropathy and bone marrow failure. J Pathol 2020; 251:117-122. [PMID: 32297672 PMCID: PMC7384058 DOI: 10.1002/path.5450] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 03/11/2020] [Accepted: 04/02/2020] [Indexed: 12/19/2022]
Abstract
The intestinal epithelium is perpetually renewed from a stem cell niche in the base of crypts to maintain a healthy bowel mucosa. Exit from this niche and maturation of epithelial cells requires tightly controlled gradients in BMP signalling, progressing from low BMP signalling at the crypt base to high signalling at the luminal surface. The BMP antagonist gremlin 1 (Grem1) is highly expressed by subepithelial myofibroblasts adjacent to the intestinal crypts but its role in regulating the stem cell niche and epithelial renewal in vivo has not been explored. To explore the effects of Grem1 loss in adulthood following normal growth and development, we bred mice (ROSA26CreER‐Grem1flx/flx) in which Grem1 could be deleted by tamoxifen administration. While Grem1 remained intact, these mice were healthy, grew normally, and reproduced successfully. Following Grem1 depletion, the mice became unwell and were euthanised (at 7–13 days). Post‐mortem examination revealed extensive mucosal abnormalities throughout the small and large intestines with failure of epithelial cell replication and maturation, villous atrophy, and features of malabsorption. Bone marrow hypoplasia was also observed with associated early haematopoietic failure. These results demonstrate an essential homeostatic role for gremlin 1 in maintaining normal bowel epithelial function in adulthood, suggesting that abnormalities in gremlin 1 expression can contribute to enteropathies. We also identified a previously unsuspected requirement for gremlin 1 in normal haematopoiesis. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Simon C Rowan
- University College Dublin, School of Medicine and Conway Institute, Dublin, Ireland
| | - Hanne Jahns
- University College Dublin, School of Veterinary Medicine, Dublin, Ireland
| | - Liberty Mthunzi
- University College Dublin, School of Medicine and Conway Institute, Dublin, Ireland
| | - Lucie Piouceau
- University College Dublin, School of Medicine and Conway Institute, Dublin, Ireland
| | - Joanna Cornwell
- University College Dublin, School of Medicine and Conway Institute, Dublin, Ireland
| | - Róisín Doody
- University College Dublin, School of Medicine and Conway Institute, Dublin, Ireland
| | | | - John J Callanan
- University College Dublin, School of Veterinary Medicine, Dublin, Ireland
| | - Paul McLoughlin
- University College Dublin, School of Medicine and Conway Institute, Dublin, Ireland
| |
Collapse
|
245
|
Xu GP, Zhang XF, Sun L, Chen EM. Current and future uses of skeletal stem cells for bone regeneration. World J Stem Cells 2020; 12:339-350. [PMID: 32547682 PMCID: PMC7280866 DOI: 10.4252/wjsc.v12.i5.339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/07/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
The postnatal skeleton undergoes growth, modeling, and remodeling. The human skeleton is a composite of diverse tissue types, including bone, cartilage, fat, fibroblasts, nerves, blood vessels, and hematopoietic cells. Fracture nonunion and bone defects are among the most challenging clinical problems in orthopedic trauma. The incidence of nonunion or bone defects following fractures is increasing. Stem and progenitor cells mediate homeostasis and regeneration in postnatal tissue, including bone tissue. As multipotent stem cells, skeletal stem cells (SSCs) have a strong effect on the growth, differentiation, and repair of bone regeneration. In recent years, a number of important studies have characterized the hierarchy, differential potential, and bone formation of SSCs. Here, we describe studies on and applications of SSCs and/or mesenchymal stem cells for bone regeneration.
Collapse
Affiliation(s)
- Guo-Ping Xu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xiang-Feng Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Lu Sun
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, MA 02115, United States
| | - Er-Man Chen
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
246
|
Kagan BJ, Rosello‐Diez A. Integrating levels of bone growth control: From stem cells to body proportions. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e384. [DOI: 10.1002/wdev.384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/09/2020] [Accepted: 04/16/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Brett J. Kagan
- Australian Regenerative Medicine Institute Monash University Clayton Australia
| | | |
Collapse
|
247
|
Zhong L, Yao L, Tower RJ, Wei Y, Miao Z, Park J, Shrestha R, Wang L, Yu W, Holdreith N, Huang X, Zhang Y, Tong W, Gong Y, Ahn J, Susztak K, Dyment N, Li M, Long F, Chen C, Seale P, Qin L. Single cell transcriptomics identifies a unique adipose lineage cell population that regulates bone marrow environment. eLife 2020; 9:e54695. [PMID: 32286228 PMCID: PMC7220380 DOI: 10.7554/elife.54695] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/11/2020] [Indexed: 12/14/2022] Open
Abstract
Bone marrow mesenchymal lineage cells are a heterogeneous cell population involved in bone homeostasis and diseases such as osteoporosis. While it is long postulated that they originate from mesenchymal stem cells, the true identity of progenitors and their in vivo bifurcated differentiation routes into osteoblasts and adipocytes remain poorly understood. Here, by employing large scale single cell transcriptome analysis, we computationally defined mesenchymal progenitors at different stages and delineated their bi-lineage differentiation paths in young, adult and aging mice. One identified subpopulation is a unique cell type that expresses adipocyte markers but contains no lipid droplets. As non-proliferative precursors for adipocytes, they exist abundantly as pericytes and stromal cells that form a ubiquitous 3D network inside the marrow cavity. Functionally they play critical roles in maintaining marrow vasculature and suppressing bone formation. Therefore, we name them marrow adipogenic lineage precursors (MALPs) and conclude that they are a newly identified component of marrow adipose tissue.
Collapse
Affiliation(s)
- Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, The First Hospital of China Medical UniversityShenyangChina
| | - Robert J Tower
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhen Miao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Jihwan Park
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of PennsylvaniaPhiladelphiaUnited States
| | - Rojesh Shrestha
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of PennsylvaniaPhiladelphiaUnited States
| | - Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, Shandong University Qilu Hospital, Shandong UniversityJinanChina
| | - Wei Yu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Nicholas Holdreith
- Division of Hematology, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pediatrics, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Xiaobin Huang
- Department of Pediatrics, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Yejia Zhang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical CenterPhiladelphiaUnited States
| | - Wei Tong
- Division of Hematology, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pediatrics, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Yanqing Gong
- Division of Transnational Medicine and Human Genetics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Jaimo Ahn
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of PennsylvaniaPhiladelphiaUnited States
| | - Nathanial Dyment
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopaedics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery/Pharmacology, University of Pennsylvania, School of Dental MedicinePhiladelphiaUnited States
| | - Patrick Seale
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
248
|
May RD, Frauchiger DA, Albers CE, Tekari A, Benneker LM, Klenke FM, Hofstetter W, Gantenbein B. Application of Cytokines of the Bone Morphogenetic Protein (BMP) Family in Spinal Fusion - Effects on the Bone, Intervertebral Disc and Mesenchymal Stromal Cells. Curr Stem Cell Res Ther 2020; 14:618-643. [PMID: 31455201 PMCID: PMC7040507 DOI: 10.2174/1574888x14666190628103528] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Low back pain is a prevalent socio-economic burden and is often associated with damaged or degenerated intervertebral discs (IVDs). When conservative therapy fails, removal of the IVD (discectomy), followed by intersomatic spinal fusion, is currently the standard practice in clinics. The remaining space is filled with an intersomatic device (cage) and with bone substitutes to achieve disc height compensation and bone fusion. As a complication, in up to 30% of cases, spinal non-fusions result in a painful pseudoarthrosis. Bone morphogenetic proteins (BMPs) have been clinically applied with varied outcomes. Several members of the BMP family, such as BMP2, BMP4, BMP6, BMP7, and BMP9, are known to induce osteogenesis. Questions remain on why hyper-physiological doses of BMPs do not show beneficial effects in certain patients. In this respect, BMP antagonists secreted by mesenchymal cells, which might interfere with or block the action of BMPs, have drawn research attention as possible targets for the enhancement of spinal fusion or the prevention of non-unions. Examples of these antagonists are noggin, gremlin1 and 2, chordin, follistatin, BMP3, and twisted gastrulation. In this review, we discuss current evidence of the osteogenic effects of several members of the BMP family on osteoblasts, IVD cells, and mesenchymal stromal cells. We consider in vitro and in vivo studies performed in human, mouse, rat, and rabbit related to BMP and BMP antagonists in the last two decades. We give insights into the effects that BMP have on the ossification of the spine. Furthermore, the benefits, pitfalls, and possible safety concerns using these cytokines for the improvement of spinal fusion are discussed.
Collapse
Affiliation(s)
- Rahel Deborah May
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Christoph Emmanuel Albers
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Adel Tekari
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Lorin Michael Benneker
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Frank Michael Klenke
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Willy Hofstetter
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Benjamin Gantenbein
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
249
|
Čamernik K, Mihelič A, Mihalič R, Haring G, Herman S, Marolt Presen D, Janež A, Trebše R, Marc J, Zupan J. Comprehensive analysis of skeletal muscle- and bone-derived mesenchymal stem/stromal cells in patients with osteoarthritis and femoral neck fracture. Stem Cell Res Ther 2020; 11:146. [PMID: 32245507 PMCID: PMC7118858 DOI: 10.1186/s13287-020-01657-z] [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: 11/26/2019] [Revised: 02/24/2020] [Accepted: 03/18/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Mesenchymal stem/stromal cells (MSCs) can replenish the aged cells of the musculoskeletal system in adult life. Stem cell exhaustion and decrease in their regenerative potential have been suggested to be hallmarks of aging. Here, we investigated whether muscle- and bone-derived MSCs of patients with osteoarthritis and osteoporosis are affected by this exhaustion, compared to healthy donors. METHODS Patients with primary osteoarthritis, femoral neck fractures due to osteoporosis, and healthy donors (controls) were included. MSCs were isolated from the skeletal muscle and subchondral bone from each patient and compared using ex vivo and in vitro analyses, including immunophenotyping, colony-forming unit fibroblast assays, growth kinetics, cell senescence, multilineage potential, and MSC marker gene expression profiling. RESULTS Freshly isolated cells from muscle from patients with osteoarthritis showed a lower proportion of CD45/CD19/CD14/CD34-negative cells compared to patients with osteoporosis and healthy donors. Freshly isolated muscle cells from patients with osteoarthritis and osteoporosis also showed higher clonogenicity compared to healthy donors. MSCs from both tissues of osteoarthritis patients showed significantly reduced osteogenesis and MSCs from the bone also reduced adipogenesis. Chondrogenic pellet diameter was reduced in bone-derived MSCs from both patient groups compared to healthy donors. A significant positive correlation was observed between adipogenesis and CD271 expression in muscle-derived MSCs. CD73 was significantly lower in bone-derived MSCs from osteoarthritis patients, compared to osteoporosis patients. Gene expression profiling showed significantly lower expression of MSC marker gene leptin receptor, LEPR, previously identified as the major source of the bone and adipocytes in the adult bone marrow, in bone-derived MSCs from patients with osteoarthritis in comparison with osteoporotic patients and healthy donors. CONCLUSIONS Our results show deficient ex vivo and in vitro properties of both skeletal muscle- and bone-derived MSCs in osteoarthritis and osteoporosis patients, compared to healthy donors. In bone-derived MSCs from patients with osteoarthritis, we also identified a lower expression of the leptin receptor, a marker of MSCs that present a major source of MSCs in the adult bone marrow. This suggests that exhaustion of skeletal muscle- and bone-derived MSCs is a hallmark of osteoarthritis and osteoporosis, which defines the need for further clinical trials of stem cell transplantation in these patients.
Collapse
Affiliation(s)
- Klemen Čamernik
- University of Ljubljana, Faculty of Pharmacy, Chair of Clinical Biochemistry, Askerceva 7, 1000, Ljubljana, Slovenia
| | - Anže Mihelič
- Valdoltra Orthopaedic Hospital, Jadranska 31, SI-6280, Ankaran, Slovenia
| | - Rene Mihalič
- Valdoltra Orthopaedic Hospital, Jadranska 31, SI-6280, Ankaran, Slovenia
| | - Gregor Haring
- University of Ljubljana, Faculty of Medicine, Institute of Forensic Medicine, Korytkova 2, 1000, Ljubljana, Slovenia
| | - Simon Herman
- Clinical Department of Traumatology, University Medical Centre, Zaloska cesta 7, 1000, Ljubljana, Slovenia
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, A-1200, Vienna, Austria
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre, Zaloska cesta 2, 1000, Ljubljana, Slovenia
| | - Rihard Trebše
- Valdoltra Orthopaedic Hospital, Jadranska 31, SI-6280, Ankaran, Slovenia
| | - Janja Marc
- University of Ljubljana, Faculty of Pharmacy, Chair of Clinical Biochemistry, Askerceva 7, 1000, Ljubljana, Slovenia
| | - Janja Zupan
- University of Ljubljana, Faculty of Pharmacy, Chair of Clinical Biochemistry, Askerceva 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
250
|
Lienemann PS, Vallmajo‐Martin Q, Papageorgiou P, Blache U, Metzger S, Kiveliö A, Milleret V, Sala A, Hoehnel S, Roch A, Reuten R, Koch M, Naveiras O, Weber FE, Weber W, Lutolf MP, Ehrbar M. Smart Hydrogels for the Augmentation of Bone Regeneration by Endogenous Mesenchymal Progenitor Cell Recruitment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903395. [PMID: 32274319 PMCID: PMC7141038 DOI: 10.1002/advs.201903395] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/27/2019] [Indexed: 04/14/2023]
Abstract
The treatment of bone defects with recombinant bone morphogenetic protein-2 (BMP-2) requires high doses precluding broad clinical application. Here, a bioengineering approach is presented that strongly improves low-dose BMP-2-based bone regeneration by mobilizing healing-associated mesenchymal progenitor cells (MPCs). Smart synthetic hydrogels are used to trap and study endogenous MPCs trafficking to bone defects. Hydrogel-trapped and prospectively isolated MPCs differentiate into multiple lineages in vitro and form bone in vivo. In vitro screenings reveal that platelet-derived growth factor BB (PDGF-BB) strongly recruits prospective MPCs making it a promising candidate for the engineering of hydrogels that enrich endogenous MPCs in vivo. However, PDGF-BB inhibits BMP-2-mediated osteogenesis both in vitro and in vivo. In contrast, smart two-way dynamic release hydrogels with fast-release of PDGF-BB and sustained delivery of BMP-2 beneficially promote the healing of bone defects. Collectively, it is shown that modulating the dynamics of endogenous progenitor cells in vivo by smart synthetic hydrogels significantly improves bone healing and holds great potential for other advanced applications in regenerative medicine.
Collapse
Affiliation(s)
- Philipp S. Lienemann
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Queralt Vallmajo‐Martin
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Panagiota Papageorgiou
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| | - Ulrich Blache
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| | - Stéphanie Metzger
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Anna‐Sofia Kiveliö
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Vincent Milleret
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| | - Ana Sala
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| | - Sylke Hoehnel
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Aline Roch
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Raphael Reuten
- Institute for Dental Research and Oral Musculoskeletal BiologyCenter for BiochemistryUniversity of CologneCologne50931Germany
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal BiologyCenter for BiochemistryUniversity of CologneCologne50931Germany
| | - Olaia Naveiras
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Franz E. Weber
- Department of Cranio‐Maxillofacial SurgeryOral Biotechnology and BioengineeringUniversity Hospital ZurichFrauenklinikstrasse 24Zurich8091Switzerland
| | - Wilfried Weber
- Faculty of Biology and BIOSS Centre for Biological Signalling StudiesUniversity of FreiburgSchänzlestr. 18Freiburg79104Germany
| | - Matthias P. Lutolf
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Martin Ehrbar
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| |
Collapse
|