151
|
Pohl S, Angermann A, Jeschke A, Hendrickx G, Yorgan TA, Makrypidi-Fraune G, Steigert A, Kuehn SC, Rolvien T, Schweizer M, Koehne T, Neven M, Winter O, Velho RV, Albers J, Streichert T, Pestka JM, Baldauf C, Breyer S, Stuecker R, Muschol N, Cox TM, Saftig P, Paganini C, Rossi A, Amling M, Braulke T, Schinke T. The Lysosomal Protein Arylsulfatase B Is a Key Enzyme Involved in Skeletal Turnover. J Bone Miner Res 2018; 33:2186-2201. [PMID: 30075049 DOI: 10.1002/jbmr.3563] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/10/2018] [Accepted: 06/20/2018] [Indexed: 12/24/2022]
Abstract
Skeletal pathologies are frequently observed in lysosomal storage disorders, yet the relevance of specific lysosomal enzymes in bone remodeling cell types is poorly defined. Two lysosomal enzymes, ie, cathepsin K (Ctsk) and Acp5 (also known as tartrate-resistant acid phosphatase), have long been known as molecular marker proteins of differentiated osteoclasts. However, whereas the cysteine protease Ctsk is directly involved in the degradation of bone matrix proteins, the molecular function of Acp5 in osteoclasts is still unknown. Here we show that Acp5, in concert with Acp2 (lysosomal acid phosphatase), is required for dephosphorylation of the lysosomal mannose 6-phosphate targeting signal to promote the activity of specific lysosomal enzymes. Using an unbiased approach we identified the glycosaminoglycan-degrading enzyme arylsulfatase B (Arsb), mutated in mucopolysaccharidosis type VI (MPS-VI), as an osteoclast marker, whose activity depends on dephosphorylation by Acp2 and Acp5. Similar to Acp2/Acp5-/- mice, Arsb-deficient mice display lysosomal storage accumulation in osteoclasts, impaired osteoclast activity, and high trabecular bone mass. Of note, the most prominent lysosomal storage accumulation was observed in osteocytes from Arsb-deficient mice, yet this pathology did not impair production of sclerostin (Sost) and Fgf23. Because the influence of enzyme replacement therapy (ERT) on bone remodeling in MPS-VI is still unknown, we additionally treated Arsb-deficient mice by weekly injection of recombinant human ARSB from 12 to 24 weeks of age. We found that the high bone mass phenotype of Arsb-deficient mice and the underlying bone cell deficits were fully corrected by ERT in the trabecular compartment. Taken together, our results do not only show that the function of Acp5 in osteoclasts is linked to dephosphorylation and activation of lysosomal enzymes, they also provide an important proof-of-principle for the feasibility of ERT to correct bone cell pathologies in lysosomal storage disorders. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Sandra Pohl
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandra Angermann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Jeschke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gretl Hendrickx
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georgia Makrypidi-Fraune
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Steigert
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja C Kuehn
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Department of Electron Microscopy, Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Koehne
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Orthodontics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mona Neven
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olga Winter
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renata Voltolini Velho
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joachim Albers
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Streichert
- Department of Clinical Chemistry, University Hospital Cologne, Cologne, Germany
| | - Jan M Pestka
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Baldauf
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Breyer
- Department of Orthopedics, Children's Hospital Hamburg-Altona, Hamburg, Germany
| | - Ralf Stuecker
- Department of Orthopedics, Children's Hospital Hamburg-Altona, Hamburg, Germany
| | - Nicole Muschol
- Department of Electron Microscopy, Center of Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timothy M Cox
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Chiara Paganini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Antonio Rossi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Braulke
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
152
|
Rosi S. The final frontier: Transient microglia reduction after cosmic radiation exposure mitigates cognitive impairments and modulates phagocytic activity. Brain Circ 2018; 4:109-113. [PMID: 30450416 PMCID: PMC6187945 DOI: 10.4103/bc.bc_24_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/24/2018] [Accepted: 09/10/2018] [Indexed: 11/21/2022] Open
Abstract
Microglia are the primary immune element within the brain, which are responsible for monitoring synapse function and neuron health. Exposure to cosmic radiation has the potential to cause long-term cognitive deficits in rodent models and therefore indicates a difficult challenge for future astronauts piloting interplanetary travel. Here, we discuss the potential of transient microglia depletion after the injury to ameliorate the harsh microenvironment of the brain and eliminate any potential long-term cognitive effects. Repopulation of microglia enables phagocytic phenotypes to be circumvented, via the reduction of Phagocytic and lysosomal markers, potentially being responsible for increased neuroprotection. Brief depletion of microglia after irradiation mitigated the development of any long-term memory deficits, comparable to healthy animals. Chronically, microglial levels were not affected by cosmic radiation followed by temporary microglia depletion. Following repopulation, improved recognition memory was paralleled by downregulated complement receptor C5aR. Preserved synapse function also demonstrated the therapeutic ability of microglia depletion as it corresponded with fewer phagocytic microglia phenotypes. The understanding of long-term radiation-induced cognitive impairments is vital for the protection of future astronauts and equally as important for current cancer patients. Temporary microglia depletion showed promise in preventing any deleterious cognitive impairments following exposure to elements of cosmic radiation, such as helium and high-charge nuclei.
Collapse
Affiliation(s)
- Susanna Rosi
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA.,Department of Neurological Surgery, University of California, San Francisco, CA, USA.,Weill Institute for Neuroscience, University of California, San Francisco, CA, USA.,Kavli Institute of Fundamental Neuroscience, University of California, San Francisco, CA, USA
| |
Collapse
|
153
|
Joseph C, Green AC, Kwang D, Purton LE. Extrinsic Regulation of Hematopoietic Stem Cells and Lymphocytes by Vitamin A. CURRENT STEM CELL REPORTS 2018. [DOI: 10.1007/s40778-018-0142-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
154
|
Plein A, Fantin A, Denti L, Pollard JW, Ruhrberg C. Erythro-myeloid progenitors contribute endothelial cells to blood vessels. Nature 2018; 562:223-228. [PMID: 30258231 PMCID: PMC6289247 DOI: 10.1038/s41586-018-0552-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 08/17/2018] [Indexed: 12/16/2022]
Abstract
The earliest blood vessels in mammalian embryos are formed when endothelial cells differentiate from angioblasts and coalesce into tubular networks. Thereafter, the endothelium is thought to expand solely by proliferation of pre-existing endothelial cells. Here we show that a complementary source of endothelial cells is recruited into pre-existing vasculature after differentiation from the earliest precursors of erythrocytes, megakaryocytes and macrophages, the erythro-myeloid progenitors (EMPs) that are born in the yolk sac. A first wave of EMPs contributes endothelial cells to the yolk sac endothelium, and a second wave of EMPs colonizes the embryo and contributes endothelial cells to intraembryonic endothelium in multiple organs, where they persist into adulthood. By demonstrating that EMPs constitute a hitherto unrecognized source of endothelial cells, we reveal that embryonic blood vascular endothelium expands in a dual mechanism that involves both the proliferation of pre-existing endothelial cells and the incorporation of endothelial cells derived from haematopoietic precursors.
Collapse
Affiliation(s)
- Alice Plein
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
155
|
Huang H, Wang J, Zhang Y, Zhu G, Li YP, Ping J, Chen W. Bone resorption deficiency affects tooth root development in RANKL mutant mice due to attenuated IGF-1 signaling in radicular odontoblasts. Bone 2018; 114:161-171. [PMID: 29292230 DOI: 10.1016/j.bone.2017.12.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/27/2017] [Accepted: 12/28/2017] [Indexed: 01/05/2023]
Abstract
The tooth root is essential for normal tooth physiological function. Studies on mice with mutations or targeted gene deletions revealed that osteoclasts (OCs) play an important role in tooth root development. However, knowledge on the cellular and molecular mechanism underlying how OCs mediate root formation is limited. During bone formation, growth factors (e.g. Insulin-like growth factor-1, IGF-1) liberated from bone matrix by osteoclastic bone resorption stimulate osteoblast differentiation. Thus, we hypothesize that OC-osteoblast coupling may also apply to OC-odontoblast coupling; therefore OCs may have a direct impact on odontoblast differentiation through the release of growth factor(s) from bone matrix, and consequently regulate tooth root formation. To test this hypothesis, we used a receptor activator of NF-κB ligand (RANKL) knockout mouse model in which OC differentiation and function was entirely blocked. We found that molar root formation and tooth eruption were defective in RANKL-/- mice. Disrupted elongation and disorganization of Hertwig's epithelial root sheath (HERS) was observed in RANKL-/- mice. Reduced expression of nuclear factor I C (NFIC), osterix, and dentin sialoprotein, markers essential for radicular (root) odontogenic cell differentiation indicated that odontoblast differentiation was disrupted in RANKL deficient mice likely contributing to the defect in root formation. Moreover, down-regulation of IGF/AKT/mTOR activity in odontoblast indicated that IGF signaling transduction in odontoblasts of the mutant mice was impaired. Treating odontoblast cells in vitro with conditioned medium from RANKL-/- OCs cultured on bone slices resulted in inhibition of odontoblast differentiation. Moreover, depletion of IGF-1 in bone resorption-conditioned medium (BRCM) from wild-type (WT) OC significantly compromised the ability of WT osteoclastic BRCM to induce odontoblast differentiation while addition of IGF-1 into RANKL-/- osteoclastic BRCM rescued impaired odontoblast differentiation, confirming that root and eruption defect in RANKL deficiency mice may result from failure of releasing of IGF-1 from bone matrix through OC bone resorption. These results suggest that OCs are important for odontoblast differentiation and tooth root formation, possibly through IGF/AKT/mTOR signaling mediated by cell-bone matrix interaction. These findings provide significant insights into regulatory mechanism of tooth root development, and also lay the foundation for root regeneration studies.
Collapse
Affiliation(s)
- Hong Huang
- The Affiliated Hospital of Stomatology, Chongqing Medical University, 5 Shangqingsi Rd, Yuzhong Qu, Chongqing Shi 400065, China; Department of Pathology, School of Medicine, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Jue Wang
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Yan Zhang
- The Affiliated Hospital of Stomatology, Chongqing Medical University, 5 Shangqingsi Rd, Yuzhong Qu, Chongqing Shi 400065, China; Department of Pathology, School of Medicine, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Yi-Ping Li
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Ji Ping
- The Affiliated Hospital of Stomatology, Chongqing Medical University, 5 Shangqingsi Rd, Yuzhong Qu, Chongqing Shi 400065, China.
| | - Wei Chen
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA.
| |
Collapse
|
156
|
Omar I, Guterman-Ram G, Rahat D, Tabach Y, Berger M, Levaot N. Schlafen2 mutation in mice causes an osteopetrotic phenotype due to a decrease in the number of osteoclast progenitors. Sci Rep 2018; 8:13005. [PMID: 30158544 PMCID: PMC6115409 DOI: 10.1038/s41598-018-31428-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/30/2018] [Indexed: 01/14/2023] Open
Abstract
Osteoclasts are the bone resorbing cells that derive from myeloid progenitor cells. Although there have been recent advancements in the ability to identify osteoclast progenitors, very little is known about the molecular mechanisms governing their homeostasis. Here, by analyzing the normalized phylogenetic profiles of the Schlafen (Slfn) gene family, we found that it co-evolved with osteoclast-related genes. Following these findings, we used a Slfn2 loss-of-function mutant mouse, elektra, to study the direct role of Slfn2 in osteoclast development and function. Slfn2eka/eka mice exhibited a profound increase in their cancellous bone mass and a significant reduction in osteoclast numbers. In addition, monocyte cultures from the bone marrow of Slfn2eka/eka mice showed a reduction in osteoclast number and total resorption area. Finally, we show that the bone marrow of Slfn2eka/eka mice have significantly less CD11b-Ly6Chi osteoclast precursors. Overall, our data suggest that Slfn2 is required for normal osteoclast differentiation and that loss of its function in mice results in an osteopetrotic phenotype.
Collapse
Affiliation(s)
- Ibrahim Omar
- The Lautenberg Center for Immunology and Cancer Research, The Biomedical Research Institute Israel Canada of the Faculty of Medicine, The Hebrew University Hadassah Medical School Jerusalem, Jerusalem, Israel
| | - Gali Guterman-Ram
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Dolev Rahat
- Department of Developmental Biology and Cancer Research, The Biomedical Research Institute Israel Canada of the Faculty of Medicine, The Hebrew University Hadassah Medical School Jerusalem, Jerusalem, Israel
| | - Yuval Tabach
- Department of Developmental Biology and Cancer Research, The Biomedical Research Institute Israel Canada of the Faculty of Medicine, The Hebrew University Hadassah Medical School Jerusalem, Jerusalem, Israel
| | - Michael Berger
- The Lautenberg Center for Immunology and Cancer Research, The Biomedical Research Institute Israel Canada of the Faculty of Medicine, The Hebrew University Hadassah Medical School Jerusalem, Jerusalem, Israel.
| | - Noam Levaot
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
157
|
Neutrophil and Macrophage Cell Surface Colony-Stimulating Factor 1 Shed by ADAM17 Drives Mouse Macrophage Proliferation in Acute and Chronic Inflammation. Mol Cell Biol 2018; 38:MCB.00103-18. [PMID: 29891514 DOI: 10.1128/mcb.00103-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/06/2018] [Indexed: 02/04/2023] Open
Abstract
Macrophages are prominent cells in acute and chronic inflammatory diseases. Recent studies highlight a role for macrophage proliferation post-monocyte recruitment under inflammatory conditions. Using an acute peritonitis model, we identify a significant defect in macrophage proliferation in mice lacking the leukocyte transmembrane protease ADAM17. The defect is associated with decreased levels of macrophage colony-stimulating factor 1 (CSF-1) in the peritoneum and is rescued by intraperitoneal injection of CSF-1. Cell surface CSF-1 (csCSF-1) is one of the substrates of ADAM17. We demonstrate that both infiltrated neutrophils and macrophages are major sources of csCSF-1. Furthermore, acute shedding of csCSF-1 following neutrophil extravasation is associated with elevated expression of iRhom2, a member of the rhomboid-like superfamily, which promotes ADAM17 maturation and trafficking to the neutrophil surface. Accordingly, deletion of hematopoietic iRhom2 is sufficient to prevent csCSF-1 release from neutrophils and macrophages and to prevent macrophage proliferation. In acute inflammation, csCSF-1 release and macrophage proliferation are self-limiting due to transient leukocyte recruitment and temporally restricted csCSF-1 expression. In chronic inflammation, such as atherosclerosis, the ADAM17-mediated lesional macrophage proliferative response is prolonged. Our results demonstrate a novel mechanism whereby ADAM17 promotes macrophage proliferation in states of acute and chronic inflammation.
Collapse
|
158
|
Rab GTPases in Osteoclastic Endomembrane Systems. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4541538. [PMID: 30186859 PMCID: PMC6114073 DOI: 10.1155/2018/4541538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
Osteoclasts (OCs) are bone-resorbing cells that maintain bone homeostasis. OC differentiation, survival, and activity are regulated by numerous small GTPases, including those of the Rab family, which are involved in plasma membrane delivery and lysosomal and autophagic degradation pathways. In resorbing OCs, polarized vesicular trafficking pathways also result in formation of the ruffled membrane, the resorbing organelle, and in transcytosis.
Collapse
|
159
|
Dawodu D, Patecki M, Hegermann J, Dumler I, Haller H, Kiyan Y. oxLDL inhibits differentiation and functional activity of osteoclasts via scavenger receptor-A mediated autophagy and cathepsin K secretion. Sci Rep 2018; 8:11604. [PMID: 30072716 PMCID: PMC6072764 DOI: 10.1038/s41598-018-29963-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/19/2018] [Indexed: 12/24/2022] Open
Abstract
Resorptive activity of osteoclasts is important for maintaining bone homeostasis. Endogenous compounds such as oxidized low density lipoprotein (oxLDL) have been shown to disturb this activity. While some studies have investigated the effects of oxLDL on the process of osteoclastogenesis, the underlying mechanism are not fully understood. We show here that oxLDL concentrations of ~10-25 µg protein (0.43-1.0 µM MDA/mg protein) completely blocked the formation of functional osteoclasts. The underlying mechanism implies an inhibition of autophagy that in turn leads to a decreased fusion of cathepsin K (CatK)-loaded lysosomal vesicles with the ruffled border membrane. As result, a lower secretion of CatK and impaired protonation of the resorption lacunae by vacuolar-ATPase (v-ATPase) is observed in the presence of oxLDL. We demonstrate that scavenger receptor A (SR-A) mediates oxLDL effects on osteoclastogenesis and repressing this receptor partially rescued oxLDL effects. Collectively, our data provides an insight into the possible mechanism of oxLDL on osteoclastogenesis suggesting that it does not perturb the packaging of CatK and v-ATPase (V-a3) in the secretory lysosome, but inhibits the fusion of these lysosomes to the ruffled border. The relevance of our findings suggests a distinct link between oxLDL, autophagy and osteoclastogenesis.
Collapse
Affiliation(s)
- Damilola Dawodu
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany
| | - Margret Patecki
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany
| | - Jan Hegermann
- Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Inna Dumler
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany
| | - Yulia Kiyan
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
160
|
Lopez‐Atalaya JP, Askew KE, Sierra A, Gomez‐Nicola D. Development and maintenance of the brain's immune toolkit: Microglia and non-parenchymal brain macrophages. Dev Neurobiol 2018; 78:561-579. [PMID: 29030904 PMCID: PMC6001428 DOI: 10.1002/dneu.22545] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/29/2017] [Accepted: 10/06/2017] [Indexed: 01/10/2023]
Abstract
Microglia and non-parenchymal macrophages located in the perivascular space, the meninges and the choroid plexus are independent immune populations that play vital roles in brain development, homeostasis, and tissue healing. Resident macrophages account for a significant proportion of cells in the brain and their density remains stable throughout the lifespan thanks to constant turnover. Microglia develop from yolk sac progenitors, later evolving through intermediate progenitors in a fine-tuned process in which intrinsic factors and external stimuli combine to progressively sculpt their cell type-specific transcriptional profiles. Recent evidence demonstrates that non-parenchymal macrophages are also generated during early embryonic development. In recent years, the development of powerful fate mapping approaches combined with novel genomic and transcriptomic methodologies have greatly expanded our understanding of how brain macrophages develop and acquire specialized functions, and how cell population dynamics are regulated. Here, we review the transcription factors, epigenetic remodeling, and signaling pathways orchestrating the embryonic development of microglia and non-parenchymal macrophages. Next, we describe the dynamics of the macrophage populations of the brain and discuss the role of progenitor cells, to gain a better understanding of their functions in the healthy and diseased brain. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 561-579, 2018.
Collapse
Affiliation(s)
- Jose P. Lopez‐Atalaya
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández‐Consejo Superior de Investigaciones Científicas (UMH‐CSIC), Avenida Ramón y Cajal, s/n, Sant Joan d'AlacantSpain
| | - Katharine E. Askew
- Southampton General Hospital, Biological Sciences, University of Southampton, South Lab&Path Block, LD80C, MP840SO166YDSouthamptonUnited Kingdom
| | - Amanda Sierra
- Achucarro Basque Center for NeuroscienceLeioa48940Spain
- Ikerbasque FoundationBilbao48013Spain
- University of the Basque Country EHU/UPVLeioa48940Spain
| | - Diego Gomez‐Nicola
- Southampton General Hospital, Biological Sciences, University of Southampton, South Lab&Path Block, LD80C, MP840SO166YDSouthamptonUnited Kingdom
| |
Collapse
|
161
|
Cipriani G, Gibbons SJ, Miller KE, Yang DS, Terhaar ML, Eisenman ST, Ördög T, Linden DR, Gajdos GB, Szurszewski JH, Farrugia G. Change in Populations of Macrophages Promotes Development of Delayed Gastric Emptying in Mice. Gastroenterology 2018; 154:2122-2136.e12. [PMID: 29501441 PMCID: PMC5985210 DOI: 10.1053/j.gastro.2018.02.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Muscularis propria macrophages lie close to cells that regulate gastrointestinal motor function, including interstitial cells of Cajal (ICC) and myenteric neurons. In animal models of diabetic gastroparesis, development of delayed gastric emptying has been associated with loss of macrophages that express cytoprotective markers and reduced networks of ICC. Mice with long-term diabetes and normal gastric emptying have macrophages that express anti-inflammatory markers and have normal gastric ICC. Mice homozygous for the osteopetrosis spontaneous mutation in the colony-stimulating factor 1 gene (Csf1op/op) do not have macrophages; when they are given streptozotocin to induce diabetes, they do not develop delayed gastric emptying. We investigated whether population of the gastric muscularis propria of diabetic Csf1op/op mice with macrophages is necessary to change gastric emptying, ICC, and myenteric neurons and investigated the macrophage-derived factors that determine whether diabetic mice do or do not develop delayed gastric emptying. METHODS Wild-type and Csf1op/op mice were given streptozotocin to induce diabetes. Some Csf1op/op mice were given daily intraperitoneal injections of CSF1 for 7 weeks; gastric tissues were collected and cellular distributions were analyzed by immunohistochemistry. CD45+, CD11b+, F4/80+ macrophages were dissociated from gastric muscularis propria, isolated by flow cytometry and analyzed by quantitative real-time polymerase chain reaction. Cultured gastric muscularis propria from Csf1op/op mice was exposed to medium that was conditioned by culture with bone marrow-derived macrophages from wild-type mice. RESULTS Gastric muscularis propria from Csf1op/op mice given CSF1 contained macrophages; 11 of 15 diabetic mice given CSF1 developed delayed gastric emptying and had damaged ICC. In non-diabetic Csf1op/op mice, administration of CSF1 reduced numbers of gastric myenteric neurons but did not affect the proportion of nitrergic neurons or ICC. In diabetic Csf1op/op mice given CSF1 that developed delayed gastric emptying, the proportion of nitrergic neurons was the same as in non-diabetic wild-type controls. Medium conditioned by macrophages previously exposed to oxidative injury caused damage to ICC in cultured gastric muscularis propria from Csf1op/op mice; neutralizing antibodies against IL6R or TNF prevented this damage to ICC. CD45+, CD11b+, and F4/80+ macrophages isolated from diabetic wild-type mice with delayed gastric emptying expressed higher levels of messenger RNAs encoding inflammatory markers (IL6 and inducible nitric oxide synthase) and lower levels of messenger RNAs encoding markers of anti-inflammatory cells (heme oxygenase 1, arginase 1, and FIZZ1) than macrophages isolated from diabetic mice with normal gastric emptying. CONCLUSIONS In studies of Csf1op/op and wild-type mice with diabetes, we found delayed gastric emptying to be associated with increased production of inflammatory factors, and reduced production of anti-inflammatory factors, by macrophages, leading to loss of ICC.
Collapse
|
162
|
Han Y, You X, Xing W, Zhang Z, Zou W. Paracrine and endocrine actions of bone-the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res 2018; 6:16. [PMID: 29844945 PMCID: PMC5967329 DOI: 10.1038/s41413-018-0019-6] [Citation(s) in RCA: 349] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/21/2018] [Accepted: 04/16/2018] [Indexed: 12/17/2022] Open
Abstract
The skeleton is a dynamic organ that is constantly remodeled. Proteins secreted from bone cells, namely osteoblasts, osteocytes, and osteoclasts exert regulation on osteoblastogenesis, osteclastogenesis, and angiogenesis in a paracrine manner. Osteoblasts secrete a range of different molecules including RANKL/OPG, M-CSF, SEMA3A, WNT5A, and WNT16 that regulate osteoclastogenesis. Osteoblasts also produce VEGFA that stimulates osteoblastogenesis and angiogenesis. Osteocytes produce sclerostin (SOST) that inhibits osteoblast differentiation and promotes osteoclast differentiation. Osteoclasts secrete factors including BMP6, CTHRC1, EFNB2, S1P, WNT10B, SEMA4D, and CT-1 that act on osteoblasts and osteocytes, and thereby influenceaA osteogenesis. Osteoclast precursors produce the angiogenic factor PDGF-BB to promote the formation of Type H vessels, which then stimulate osteoblastogenesis. Besides, the evidences over the past decades show that at least three hormones or "osteokines" from bone cells have endocrine functions. FGF23 is produced by osteoblasts and osteocytes and can regulate phosphate metabolism. Osteocalcin (OCN) secreted by osteoblasts regulates systemic glucose and energy metabolism, reproduction, and cognition. Lipocalin-2 (LCN2) is secreted by osteoblasts and can influence energy metabolism by suppressing appetite in the brain. We review the recent progresses in the paracrine and endocrine functions of the secretory proteins of osteoblasts, osteocytes, and osteoclasts, revealing connections of the skeleton with other tissues and providing added insights into the pathogenesis of degenerative diseases affecting multiple organs and the drug discovery process.
Collapse
Affiliation(s)
- Yujiao Han
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Xiuling You
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Wenhui Xing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Zhong Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| |
Collapse
|
163
|
Krukowski K, Feng X, Paladini MS, Chou A, Sacramento K, Grue K, Riparip LK, Jones T, Campbell-Beachler M, Nelson G, Rosi S. Temporary microglia-depletion after cosmic radiation modifies phagocytic activity and prevents cognitive deficits. Sci Rep 2018; 8:7857. [PMID: 29777152 PMCID: PMC5959907 DOI: 10.1038/s41598-018-26039-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/02/2018] [Indexed: 12/19/2022] Open
Abstract
Microglia are the main immune component in the brain that can regulate neuronal health and synapse function. Exposure to cosmic radiation can cause long-term cognitive impairments in rodent models thereby presenting potential obstacles for astronauts engaged in deep space travel. The mechanism/s for how cosmic radiation induces cognitive deficits are currently unknown. We find that temporary microglia depletion, one week after cosmic radiation, prevents the development of long-term memory deficits. Gene array profiling reveals that acute microglia depletion alters the late neuroinflammatory response to cosmic radiation. The repopulated microglia present a modified functional phenotype with reduced expression of scavenger receptors, lysosome membrane protein and complement receptor, all shown to be involved in microglia-synapses interaction. The lower phagocytic activity observed in the repopulated microglia is paralleled by improved synaptic protein expression. Our data provide mechanistic evidence for the role of microglia in the development of cognitive deficits after cosmic radiation exposure.
Collapse
Affiliation(s)
- Karen Krukowski
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA
| | - Xi Feng
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA
| | - Maria Serena Paladini
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA
| | - Austin Chou
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA
| | - Kristen Sacramento
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA
| | - Katherine Grue
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA
| | - Lara-Kirstie Riparip
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA
| | - Tamako Jones
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University, Loma Linda, CA, USA
| | - Mary Campbell-Beachler
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University, Loma Linda, CA, USA
| | - Gregory Nelson
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University, Loma Linda, CA, USA
| | - Susanna Rosi
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA. .,Brain and Spinal Injury Center, University of California, San Francisco, CA, USA. .,Department of Neurological Surgery, University of California, San Francisco, CA, USA. .,Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA. .,Kavli Institute of Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
164
|
Takeshita S, Fumoto T, Ito M, Ikeda K. Serum CTX levels and histomorphometric analysis in Src versus RANKL knockout mice. J Bone Miner Metab 2018; 36:264-273. [PMID: 28589412 DOI: 10.1007/s00774-017-0838-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/28/2017] [Indexed: 12/16/2022]
Abstract
Src knockout (KO) and RANKL KO mice both exhibit near complete osteopetrosis in terms of 3D-bone volume (BV) fraction by micro-CT, whereas the serum CTX concentration of Src KO is apparently normal and that of RANKL KO is 30% of wild-type (WT) despite the fact that they lack osteoclasts. By histomorphometry we found that, whereas eroded surface (ES) and osteoid surface (OS) are zero values in RANKL KO, they are indistinguishable from WT in Src KO; because of marked increase in bone surface (BS), ES/BS and OS/BS of Src KO are 30-40% of WT. While RANKL KO lack both osteoclasts and osteoblasts, Src KO reveal increased numbers of osteoclasts and indistinguishable numbers of osteoblasts compared with WT; again, on the basis of BS, N.Oc/BS is comparable to WT and N.Ob/BS is markedly decreased in Src KO. The apparently increased number of total osteoclasts may be due to increased expression of RANKL found in Src KO bone in vivo. Src has a gene dosage-dependent effect on osteoclast function in vitro, with Src-/- osteoclasts completely lacking bone-resorbing function as determined by CTX release on dentin. Thus, Src KO osteoclasts retain some bone-resorbing function in vivo. The number of osteocytes is proportionally increased in RANKL KO, while Src KO mice have relative osteocyte deficiency, raising the possibility that RANKL and Src has an unrecognized role in osteocyte survival.
Collapse
Affiliation(s)
- Sunao Takeshita
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan.
| | - Toshio Fumoto
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan
| | - Masako Ito
- Medical Work-Life-Balance Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kyoji Ikeda
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan
| |
Collapse
|
165
|
Sevenich L. Brain-Resident Microglia and Blood-Borne Macrophages Orchestrate Central Nervous System Inflammation in Neurodegenerative Disorders and Brain Cancer. Front Immunol 2018; 9:697. [PMID: 29681904 PMCID: PMC5897444 DOI: 10.3389/fimmu.2018.00697] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/21/2018] [Indexed: 01/09/2023] Open
Abstract
Inflammation is a hallmark of different central nervous system (CNS) pathologies. It has been linked to neurodegenerative disorders as well as primary and metastatic brain tumors. Microglia, the brain-resident immune cells, are emerging as a central player in regulating key pathways in CNS inflammation. Recent insights into neuroinflammation indicate that blood-borne immune cells represent an additional critical cellular component in mediating CNS inflammation. The lack of experimental systems that allow for discrimination between brain-resident and recruited myeloid cells has previously halted functional analysis of microglia and their blood-borne counterparts in brain malignancies. However, recent conceptual and technological advances, such as the generation of lineage tracing models and the identification of cell type-specific markers provide unprecedented opportunities to study the cellular functions of microglia and macrophages by functional interference. The use of different “omic” strategies as well as imaging techniques has significantly increased our knowledge of disease-associated gene signatures and effector functions under pathological conditions. In this review, recent developments in evaluating functions of brain-resident and recruited myeloid cells in neurodegenerative disorders and brain cancers will be discussed and unique or shared cellular traits of microglia and macrophages in different CNS disorders will be highlighted. Insight from these studies will shape our understanding of disease- and cell-type-specific effector functions of microglia or macrophages and will open new avenues for therapeutic intervention that target aberrant functions of myeloid cells in CNS pathologies.
Collapse
Affiliation(s)
- Lisa Sevenich
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| |
Collapse
|
166
|
Abstract
Osteoporosis, characterized by low bone mass and micro-architectural deterioration of bone tissue with increased risk of fracture, can be categorized into two forms: primary and secondary, depending on whether it occurs as part of the natural aging process (estrogen deficiency) or as part of disease pathology. In both forms bone loss is due to an imbalance in the bone remodeling process, with resorption/formation skewed more toward bone loss. Recent studies and emerging evidence consistently demonstrate the potential of the intestinal microbiota to modulate bone health. This review discusses the process of bone remodeling and the pathology of osteoporosis and introduces the intestinal microbiota and its potential to influence bone health. In particular, we highlight recent murine studies that examine how probiotic supplementation can both increase bone density in healthy individuals and protect against primary (estrogen deficiency) as well as secondary osteoporosis. Potential mechanisms are described to account for how probiotic treatments could be exerting their beneficial effect on bone health.
Collapse
|
167
|
Güney Saruhan B, Sağsöz H, Akbalık E, Ketani MA, Erdoğan S. Distribution of CD68-, CD8-, MHCI- and MHCII-positive cells in the bull and ram testis and epididymis. Anat Histol Embryol 2018. [PMID: 29527795 DOI: 10.1111/ahe.12354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The mammalian testis possesses a special immunological environment because of its properties of remarkable immune privilege and effective local innate immunity. The testicular immune privilege protects immunogenic germ cells from systemic immune attack, and local innate immunity is important in preventing testicular microbial infections. Thus, this study aimed to immunohistochemically demonstrate the distribution and localization of CD68-, CD8-, MHCI- and MHCII-positive immune cells in the testes and epididymes. Negative immunoreactivity was detected in the seminiferous tubule epithelium and peritubular myoid cells of the testes upon staining in CD68, CD8 and MHC Class I. Positive CD68 immunoreaction was determined in the Sertoli cells and some Leydig cells. The detection of positive cells for CD8 clearly indicated the presence of lymphocytes. Furthermore, the staining with MHCI intensity was ascertained to vary from weak to moderate in the Sertoli and Leydig cells and connective tissue cells. MHCII-positive immunoreactivity was determined in myoid cells and Leydig cells in the interstitial area. The epithelium of the epididymis showed positive staining for CD68 and CD8, but the stroma displayed a rather weak staining. In the ram epididymis, neither intraepithelial nor interstitial positive reaction was observed for MHCI. In the epididymis, the basal cells displayed a stronger staining for MHCII. In conclusion, these cells not only contribute to local immunity through their direct effects on the quality of fertility in males, but also contribute either directly or indirectly to immune privilege by minimizing the development of both autoimmune reactions and potentially harmful risks.
Collapse
Affiliation(s)
- B Güney Saruhan
- Department of Histology and Embryology, Dicle University, Diyarbakır, Turkey
| | - H Sağsöz
- Department of Histology and Embryology, Dicle University, Diyarbakır, Turkey
| | - E Akbalık
- Department of Histology and Embryology, Dicle University, Diyarbakır, Turkey
| | - M A Ketani
- Department of Histology and Embryology, Dicle University, Diyarbakır, Turkey
| | - S Erdoğan
- Department of Anatomy, Namık Kemal University, Tekirdağ, Turkey
| |
Collapse
|
168
|
Abstract
The bone remodelling cycle replaces old and damaged bone and is a highly regulated, lifelong process essential for preserving bone integrity and maintaining mineral homeostasis. During the bone remodelling cycle, osteoclastic resorption is tightly coupled to osteoblastic bone formation. The remodelling cycle occurs within the basic multicellular unit and comprises five co-ordinated steps; activation, resorption, reversal, formation and termination. These steps occur simultaneously but asynchronously at multiple different locations within the skeleton. Study of rare human bone disease and animal models have helped to elucidate the cellular and molecular mechanisms that regulate the bone remodelling cycle. The key signalling pathways controlling osteoclastic bone resorption and osteoblastic bone formation are receptor activator of nuclear factor-κB (RANK)/RANK ligand/osteoprotegerin and canonical Wnt signalling. Cytokines, growth factors and prostaglandins act as paracrine regulators of the cycle, whereas endocrine regulators include parathyroid hormone, vitamin D, calcitonin, growth hormone, glucocorticoids, sex hormones, and thyroid hormone. Disruption of the bone remodelling cycle and any resulting imbalance between bone resorption and formation leads to metabolic bone disease, most commonly osteoporosis. The advances in understanding the cellular and molecular mechanisms underlying bone remodelling have also provided targets for pharmacological interventions which include antiresorptive and anabolic therapies. This review will describe the remodelling process and its regulation, discuss osteoporosis and summarize the commonest pharmacological interventions used in its management.
Collapse
Affiliation(s)
- J S Kenkre
- 1 Section of Investigative Medicine, Imperial College London, London, UK
| | - Jhd Bassett
- 2 Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
169
|
Brook N, Brook E, Dharmarajan A, Dass CR, Chan A. Breast cancer bone metastases: pathogenesis and therapeutic targets. Int J Biochem Cell Biol 2018; 96:63-78. [DOI: 10.1016/j.biocel.2018.01.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/31/2017] [Accepted: 01/04/2018] [Indexed: 01/03/2023]
|
170
|
Nakamichi Y, Udagawa N, Suda T, Takahashi N. Mechanisms involved in bone resorption regulated by vitamin D. J Steroid Biochem Mol Biol 2018; 177:70-76. [PMID: 29146302 DOI: 10.1016/j.jsbmb.2017.11.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/26/2017] [Accepted: 11/12/2017] [Indexed: 12/31/2022]
Abstract
Active forms of vitamin D enhance osteoclastogenesis in vitro and in vivo through the vitamin D receptor (VDR) in osteoblast-lineage cells consisting of osteoblasts and osteocytes. This pro-resorptive activity was evident basically with higher concentrations of active vitamin D than those expected in physiological conditions. Nevertheless, vitamin D compounds have been used in Japan for treating osteoporosis to increase bone mineral density (BMD). Of note, the increase in BMD by long-term treatment with pharmacological (=near-physiological) doses of vitamin D compounds was caused by the suppression of bone resorption. Therefore, whether vitamin D expresses pro-resorptive or anti-resorptive properties seems to be dependent on the treatment protocols. We established osteoblast lineage-specific and osteoclast-specific VDR conditional knockout (cKO) mice using Osterix-Cre transgenic mice and Cathepsin K-Cre knock-in mice, respectively. According to our observation using these cKO mouse lines, neither VDR in osteoblast-lineage cells nor that in osteoclasts played important roles for osteoclastogenesis and bone resorption at homeostasis. However, using our cKO lines, we observed that VDR in osteoblast-lineage cells, but not osteoclasts, was involved in the anti-resorptive properties of pharmacological doses of vitamin D compounds in vivo. Two different osteoblast-lineage VDR cKO mouse lines were reported. One is a VDR cKO mouse line using alpha 1, type I collagen (Col1a1)-Cre transgenic mice (here we call Col1a1-VDR-cKO mice) and the other is that using dentin matrix protein 1 (Dmp1)-Cre transgenic mice (Dmp1-VDR-cKO mice). Col1a1-VDR-cKO mice exhibited slightly increased bone mass due to lowered bone resorption. In contrast, Dmp1-VDR-cKO mice exhibited no difference in BMD in agreement with our results regarding Ob-VDR-cKO mice. Here we discuss contradictory results and multiple modes of actions of vitamin D in bone resorption in detail. (279 words).
Collapse
Affiliation(s)
- Yuko Nakamichi
- Institute for Oral Science, Matsumoto Dental University, Shiojiri, Nagano 399-0781, Japan.
| | - Nobuyuki Udagawa
- Department of Biochemistry, Matsumoto Dental University, Shiojiri, Nagano 399-0781, Japan
| | - Tatsuo Suda
- Research Center for Genomic Medicine, Saitama Medical University, Saitama 350-1241, Japan
| | - Naoyuki Takahashi
- Institute for Oral Science, Matsumoto Dental University, Shiojiri, Nagano 399-0781, Japan
| |
Collapse
|
171
|
Xu X, Yan Y, Liu Z, Qi J, Qian N, Zhou H, Zhou Q, Wang T, Huang P, Guo L, Jiang M, Deng L. The prevention of latanoprost on osteoclastgenesis in vitro and lipopolysaccharide-induced murine calvaria osteolysis in vivo. J Cell Biochem 2018; 119:4680-4691. [PMID: 29274286 DOI: 10.1002/jcb.26646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/20/2017] [Indexed: 02/02/2023]
Abstract
Identification of agents that inhibit osteoclast formation and function is important for the treatment of osteolytic diseases which feature excessive osteoclast formation and bone resorption. Latanoprost (LTP), an analog of prostaglandin F2α, is a medication which works to lower pressure inside the eyes. Prostaglandin F2α was reported to regulate bone metabolism, however, the effect of LTP in osteoclastogenesis is still unknown. Here, we found that LTP suppressed RANKL-induced osteoclastogenesis in a dose-dependent manner as illustrated by TRAP activity and TRAP staining. In addition, the osteoclast function was also reduced by LTP treatment, as indicated in less osteoclastic resorption pit areas. Furthermore, LTP inhibited the mRNA expressions of osteoclast marker genes such as TRAP and cathepsin K. In order to illustrate its molecular mechanism, we examined the changing of mRNA and protein levels of NFATc1 and c-fos by LTP treatment, as well as the phosphorylation of ERK, AKT, JNK, and p38. The results suggested that LTP inhibited RANKL-induced osteoclastgenesis and function by inhibiting ERK, AKT, JNK, and p38 cascade, following by the c-fos/NFATc1 pathway. In agreement with in vitro results, using an in vivo lipopolysaccharide-induced murine calvaria osteolysis mouse model, we found that administration of LTP was able to reverse the lipopolysaccharide-induced bone loss. Together, these data demonstrated that LTP attenuated the bone loss in lipopolysaccharide-induced murine calvaria osteolysis mice through inhibiting osteoclast formation and function. Our study thus provided the evidences that LTP was a potential treatment option against osteolytic bone diseases.
Collapse
Affiliation(s)
- Xing Xu
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yufei Yan
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhuochao Liu
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jin Qi
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Niandong Qian
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hanbing Zhou
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Zhou
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tianqi Wang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ping Huang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lei Guo
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Jiang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lianfu Deng
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
172
|
Amarasekara DS, Yun H, Kim S, Lee N, Kim H, Rho J. Regulation of Osteoclast Differentiation by Cytokine Networks. Immune Netw 2018; 18:e8. [PMID: 29503739 PMCID: PMC5833125 DOI: 10.4110/in.2018.18.e8] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 12/20/2022] Open
Abstract
Cytokines play a pivotal role in maintaining bone homeostasis. Osteoclasts (OCs), the sole bone resorbing cells, are regulated by numerous cytokines. Macrophage colony-stimulating factor and receptor activator of NF-κB ligand play a central role in OC differentiation, which is also termed osteoclastogenesis. Osteoclastogenic cytokines, including tumor necrosis factor-α, IL-1, IL-6, IL-7, IL-8, IL-11, IL-15, IL-17, IL-23, and IL-34, promote OC differentiation, whereas anti-osteoclastogenic cytokines, including interferon (IFN)-α, IFN-β, IFN-γ, IL-3, IL-4, IL-10, IL-12, IL-27, and IL-33, downregulate OC differentiation. Therefore, dynamic regulation of osteoclastogenic and anti-osteoclastogenic cytokines is important in maintaining the balance between bone-resorbing OCs and bone-forming osteoblasts (OBs), which eventually affects bone integrity. This review outlines the osteoclastogenic and anti-osteoclastogenic properties of cytokines with regard to osteoimmunology, and summarizes our current understanding of the roles these cytokines play in osteoclastogenesis.
Collapse
Affiliation(s)
| | - Hyeongseok Yun
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Sumi Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Nari Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Hyunjong Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
173
|
Wang L, Iorio C, Yan K, Yang H, Takeshita S, Kang S, Neel BG, Yang W. A ERK/RSK-mediated negative feedback loop regulates M-CSF-evoked PI3K/AKT activation in macrophages. FASEB J 2018; 32:875-887. [PMID: 29046360 PMCID: PMC5888401 DOI: 10.1096/fj.201700672rr] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/02/2017] [Indexed: 12/20/2022]
Abstract
Activation of the RAS/ERK and its downstream signaling components is essential for growth factor-induced cell survival, proliferation, and differentiation. The Src homology-2 domain containing protein tyrosine phosphatase 2 (SHP2), encoded by protein tyrosine phosphatase, non-receptor type 11 ( Ptpn11), is a positive mediator required for most, if not all, receptor tyrosine kinase-evoked RAS/ERK activation, but differentially regulates the PI3K/AKT signaling cascade in various cellular contexts. The precise mechanisms underlying the differential effects of SHP2 deficiency on the PI3K pathway remain unclear. We found that mice with myelomonocytic cell-specific [ Tg(LysM-Cre); Ptpn11fl/fl mice] Ptpn11 deficiency exhibit mild osteopetrosis. SHP2-deficient bone marrow macrophages (BMMs) showed decreased proliferation in response to M-CSF and decreased osteoclast generation. M-CSF-evoked ERK1/2 activation was decreased, whereas AKT activation was enhanced in SHP2-deficient BMMs. ERK1/2, via its downstream target RSK2, mediates this negative feedback by negatively regulating phosphorylation of M-CSF receptor at Tyr721 and, consequently, its binding to p85 subunit of PI3K and PI3K activation. Pharmacologic inhibition of RSK or ERK phenotypically mimics the signaling defects observed in SHP2-deficient BMMs. Furthermore, this increase in PI3K/AKT activation enables BMM survival in the setting of SHP2 deficiency.-Wang, L., Iorio, C., Yan, K., Yang, H., Takeshita, S., Kang, S., Neel, B.G., Yang, W. An ERK/RSK-mediated negative feedback loop regulates M-CSF-evoked PI3K/AKT activation in macrophages.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Orthopaedics, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Caterina Iorio
- Department of Medical Biophysics, Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Kevin Yan
- Department of Orthopaedics, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Howard Yang
- College of Engineering, University of Rhode Island, Kingston, Rhode Island, USA
| | - Sunao Takeshita
- Department of Bone and Joint Disease, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Sumin Kang
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Benjamin G. Neel
- Department of Medical Biophysics, Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Wentian Yang
- Department of Orthopaedics, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| |
Collapse
|
174
|
Singh K, Piprode V, Mhaske ST, Barhanpurkar-Naik A, Wani MR. IL-3 Differentially Regulates Membrane and Soluble RANKL in Osteoblasts through Metalloproteases and the JAK2/STAT5 Pathway and Improves the RANKL/OPG Ratio in Adult Mice. THE JOURNAL OF IMMUNOLOGY 2018; 200:595-606. [DOI: 10.4049/jimmunol.1601528] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Bone remodeling comprises balanced activities between osteoclasts and osteoblasts, which is regulated by various factors, including hormones and cytokines. We previously reported that IL-3 inhibits osteoclast differentiation and pathological bone loss. IL-3 also enhances osteoblast differentiation and bone formation from mesenchymal stem cells. However, the role of IL-3 in regulation of osteoblast–osteoclast interactions and underlying mechanisms is not yet delineated. In this study, we investigated the role of IL-3 on the regulation of osteoblast-specific molecules, receptor activator of NF-κB ligand (RANKL), and osteoprotegerin (OPG) that modulate bone homeostasis. We found that IL-3 increases RANKL expression at both the transcriptional and translational levels, and it showed no effect on OPG expression in calvarial osteoblasts. The increased RANKL expression by IL-3 induces mononuclear osteoclasts; however, it does not induce multinuclear osteoclasts. Interestingly, IL-3 decreases soluble RANKL by reducing ectodomain shedding of membrane RANKL through downregulation of metalloproteases mainly a disintegrin and metalloproteinase (ADAM)10, ADAM17, ADAM19, and MMP3. Moreover, IL-3 increases membrane RANKL by activating the JAK2/STAT5 pathway. Furthermore, IL-3 enhances RANKL expression in mesenchymal stem cells of wild-type mice but not in STAT5a knockout mice. Interestingly, IL-3 restores RANKL expression in adult mice by enhancing bone-specific RANKL and decreasing serum RANKL. Furthermore, IL-3 increases the serum OPG level in adult mice. Thus, our results reveal, to our knowledge for the first time, that IL-3 differentially regulates two functional forms of RANKL through metalloproteases and the JAK2/STAT5 pathway, and it helps in restoring the decreased RANKL/OPG ratio in adult mice. Notably, our studies indicate the novel role of IL-3 in regulating bone homeostasis in important skeletal disorders.
Collapse
Affiliation(s)
- Kanupriya Singh
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Vikrant Piprode
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Suhas T. Mhaske
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Amruta Barhanpurkar-Naik
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Mohan R. Wani
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| |
Collapse
|
175
|
Guterman-Ram G, Pesic M, Orenbuch A, Czeiger T, Aflalo A, Levaot N. Dual-specificity tyrosine phosphorylation-regulated kinase 2 regulates osteoclast fusion in a cell heterotypic manner. J Cell Physiol 2018; 233:617-629. [PMID: 28332708 DOI: 10.1002/jcp.25922] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 03/21/2017] [Indexed: 12/14/2022]
Abstract
Monocyte fusion into osteoclasts, bone resorbing cells, plays a key role in bone remodeling and homeostasis; therefore, aberrant cell fusion may be involved in a variety of debilitating bone diseases. Research in the last decade has led to the discovery of genes that regulate osteoclast fusion, but the basic molecular and cellular regulatory mechanisms underlying the fusion process are not completely understood. Here, we reveal a role for Dyrk2 in osteoclast fusion. We demonstrate that Dyrk2 down regulation promotes osteoclast fusion, whereas its overexpression inhibits fusion. Moreover, Dyrk2 also promotes the fusion of foreign-body giant cells, indicating that Dyrk2 plays a more general role in cell fusion. In an earlier study, we showed that fusion is a cell heterotypic process initiated by fusion-founder cells that fuse to fusion-follower cells, the latter of which are unable to initiate fusion. Here, we show that Dyrk2 limits the expansion of multinucleated founder cells through the suppression of the fusion competency of follower cells.
Collapse
Affiliation(s)
- Gali Guterman-Ram
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Milena Pesic
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ayelet Orenbuch
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tal Czeiger
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anastasia Aflalo
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Noam Levaot
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Regenerative Medicine and Stem Cell (RMSC) Research Center, Beer-Sheva, Israel
| |
Collapse
|
176
|
Harwani SC. Macrophages under pressure: the role of macrophage polarization in hypertension. Transl Res 2018; 191:45-63. [PMID: 29172035 PMCID: PMC5733698 DOI: 10.1016/j.trsl.2017.10.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/05/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023]
Abstract
Hypertension is a multifactorial disease involving the nervous, renal, and cardiovascular systems. Macrophages are the most abundant and ubiquitous immune cells, placing them in a unique position to serve as key mediators between these components. The polarization of macrophages confers vast phenotypic and functional plasticity, allowing them to act as proinflammatory, homeostatic, and anti-inflammatory agents. Key differences between the M1 and M2 phenotypes, the 2 subsets at the extremes of this polarization spectrum, place macrophages at a juncture to mediate many mechanisms involved in the pathogenesis of hypertension. Neuronal and non-neuronal regulation of the immune system, that is, the "neuroimmuno" axis, plays an integral role in the polarization of macrophages. In hypertension, the neuroimmuno axis results in synchronization of macrophage mobilization from immune cell reservoirs and their chemotaxis, via increased expression of chemoattractants, to end organs critical in the development of hypertension. This complicated system is largely coordinated by the dichotomous actions of the autonomic neuronal and non-neuronal activation of cholinergic, adrenergic, and neurohormonal receptors on macrophages, leading to their ability to "switch" between phenotypes at sites of active inflammation. Data from experimental models and human studies are in concordance with each other and support a central role for macrophage polarization in the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Sailesh C Harwani
- Department of Internal Medicine, Iowa City, IA; Center for Immunology and Immune Based Diseases, Iowa City, IA; Abboud Cardiovascular Research Center, Iowa City, Io.
| |
Collapse
|
177
|
Abstract
Macrophages are a heterogeneous population of innate immune cells and are distributed in most adult tissues. Certain tissue-resident macrophages with a prenatal origin, together with postnatal monocyte-derived macrophages, serve as the host scavenger system to eliminate invading pathogens, malignant cells, senescent cells, dead cells, cellular debris, and other foreign substances. As a key member of the mononuclear phagocyte system, macrophages play essential roles in regulation of prenatal development, tissue homeostasis, and disease progression. Over the past two decades, considerable efforts have been made to generate genetic models of macrophage ablation in mice. These models support investigations of the precise functions of tissue-specific macrophages under physiological and pathological conditions. Herein, we overview the currently available mouse strains for in vivo genetic ablation of macrophages and discuss their respective advantages and limitations.
Collapse
Affiliation(s)
- Li Hua
- The Jackson Laboratory, Bar Harbor, ME, USA
| | | | | | | |
Collapse
|
178
|
Kittaka M, Mayahara K, Mukai T, Yoshimoto T, Yoshitaka T, Gorski JP, Ueki Y. Cherubism Mice Also Deficient in c-Fos Exhibit Inflammatory Bone Destruction Executed by Macrophages That Express MMP14 Despite the Absence of TRAP+ Osteoclasts. J Bone Miner Res 2018; 33:167-181. [PMID: 28914985 PMCID: PMC5771992 DOI: 10.1002/jbmr.3295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 11/10/2022]
Abstract
Currently, it is believed that osteoclasts positive for tartrate-resistant acid phosphatase (TRAP+) are the exclusive bone-resorbing cells responsible for focal bone destruction in inflammatory arthritis. Recently, a mouse model of cherubism (Sh3bp2KI/KI ) with a homozygous gain-of-function mutation in the SH3-domain binding protein 2 (SH3BP2) was shown to develop auto-inflammatory joint destruction. Here, we demonstrate that Sh3bp2KI/KI mice also deficient in the FBJ osteosarcoma oncogene (c-Fos) still exhibit noticeable bone erosion at the distal tibia even in the absence of osteoclasts at 12 weeks old. Levels of serum collagen I C-terminal telopeptide (ICTP), a marker of bone resorption generated by matrix metalloproteinases (MMPs), were elevated, whereas levels of serum cross-linked C-telopeptide (CTX), another resorption marker produced by cathepsin K, were not increased. Collagenolytic MMP levels were increased in the inflamed joints of the Sh3bp2KI/KI mice deficient in c-Fos. Resorption pits contained a large number of F4/80+ macrophages and genetic depletion of macrophages rescued these erosive changes. Importantly, administration of NSC405020, an MMP14 inhibitor targeted to the hemopexin (PEX) domain, suppressed bone erosion in c-Fos-deficient Sh3bp2KI/KI mice. After activation of the NF-κB pathway, macrophage colony-stimulating factor (M-CSF)-dependent macrophages from c-Fos-deficient Sh3bp2KI/KI mice expressed increased amounts of MMP14 compared with wild-type macrophages. Interestingly, receptor activator of NF-κB ligand (RANKL)-deficient Sh3bp2KI/KI mice failed to show notable bone erosion, whereas c-Fos deletion did restore bone erosion to the RANKL-deficient Sh3bp2KI/KI mice, suggesting that osteolytic transformation of macrophages requires both loss-of-function of c-Fos and gain-of-function of SH3BP2 in this model. These data provide the first genetic evidence that cells other than osteoclasts can cause focal bone destruction in inflammatory bone disease and suggest that MMP14 is a key mediator conferring pathological bone-resorbing capacity on c-Fos-deficient Sh3bp2KI/KI macrophages. In summary, the paradigm that osteoclasts are the exclusive cells executing inflammatory bone destruction may need to be reevaluated based on our findings with c-Fos-deficient cherubism mice lacking osteoclasts. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mizuho Kittaka
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Kotoe Mayahara
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,Department of Orthodontics, Nihon University, School of Dentistry, Tokyo, Japan
| | - Tomoyuki Mukai
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Tetsuya Yoshimoto
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Teruhito Yoshitaka
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Jeffrey P Gorski
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,University of Missouri-Kansas City (UMKC) Center of Excellence in the Study of Dental and Musculoskeletal Tissues (CEMT), Kansas City, MO, USA
| | - Yasuyoshi Ueki
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,University of Missouri-Kansas City (UMKC) Center of Excellence in the Study of Dental and Musculoskeletal Tissues (CEMT), Kansas City, MO, USA
| |
Collapse
|
179
|
Grössinger EM, Kang M, Bouchareychas L, Sarin R, Haudenschild DR, Borodinsky LN, Adamopoulos IE. Ca 2+-Dependent Regulation of NFATc1 via KCa3.1 in Inflammatory Osteoclastogenesis. THE JOURNAL OF IMMUNOLOGY 2017; 200:749-757. [PMID: 29246953 DOI: 10.4049/jimmunol.1701170] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/02/2017] [Indexed: 12/21/2022]
Abstract
In inflammatory arthritis, the dysregulation of osteoclast activity by proinflammatory cytokines, including TNF, interferes with bone remodeling during inflammation through Ca2+-dependent mechanisms causing pathological bone loss. Ca2+-dependent CREB/c-fos activation via Ca2+-calmodulin kinase IV (CaMKIV) induces transcriptional regulation of osteoclast-specific genes via NFATc1, which facilitate bone resorption. In leukocytes, Ca2+ regulation of NFAT-dependent gene expression oftentimes involves the activity of the Ca2+-activated K+ channel KCa3.1. In this study, we evaluate KCa3.1 as a modulator of Ca2+-induced NFAT-dependent osteoclast differentiation in inflammatory bone loss. Microarray analysis of receptor activator of NF-κB ligand (RANKL)-activated murine bone marrow macrophage (BMM) cultures revealed unique upregulation of KCa3.1 during osteoclastogenesis. The expression of KCa3.1 in vivo was confirmed by immunofluorescence staining on multinucleated cells at the bone surface of inflamed mouse joints. Experiments on in vitro BMM cultures revealed that KCa3.1-/- and TRAM-34 treatment significantly reduced the expression of osteoclast-specific genes (p < 0.05) alongside decreased osteoclast formation (p < 0.0001) in inflammatory (RANKL+TNF) and noninflammatory (RANKL) conditions. In particular, live cell Ca2+ imaging and Western blot analysis showed that TRAM-34 pretreatment decreased transient RANKL-induced Ca2+ amplitudes in BMMs by ∼50% (p < 0.0001) and prevented phosphorylation of CaMKIV. KCa3.1-/- reduced RANKL+/-TNF-stimulated phosphorylation of CREB and expression of c-fos in BMMs (p < 0.01), culminating in decreased NFATc1 protein expression and transcriptional activity (p < 0.01). These data indicate that KCa3.1 regulates Ca2+-dependent NFATc1 expression via CaMKIV/CREB during inflammatory osteoclastogenesis in the presence of TNF, corroborating its role as a target candidate for the treatment of bone erosion in inflammatory arthritis.
Collapse
Affiliation(s)
- Eva M Grössinger
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616
| | - Mincheol Kang
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616
| | - Laura Bouchareychas
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616
| | - Ritu Sarin
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616
| | | | - Laura N Borodinsky
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616; and.,Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children - Northern California, Sacramento, CA 95817
| | - Iannis E Adamopoulos
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616; .,Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children - Northern California, Sacramento, CA 95817
| |
Collapse
|
180
|
Yamazaki T, Nalbandian A, Uchida Y, Li W, Arnold TD, Kubota Y, Yamamoto S, Ema M, Mukouyama YS. Tissue Myeloid Progenitors Differentiate into Pericytes through TGF-β Signaling in Developing Skin Vasculature. Cell Rep 2017; 18:2991-3004. [PMID: 28329690 DOI: 10.1016/j.celrep.2017.02.069] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/24/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Mural cells (pericytes and vascular smooth muscle cells) are essential for the regulation of vascular networks and maintenance of vascular integrity, but their origins are diverse in different tissues and not known in the organs that arise from the ectoderm, such as skin. Here, we show that tissue-localized myeloid progenitors contribute to pericyte development in embryonic skin vasculature. A series of in vivo fate-mapping experiments indicates that tissue myeloid progenitors differentiate into pericytes. Furthermore, depletion of tissue myeloid cells and their progenitors in PU.1 (also known as Spi1) mutants results in defective pericyte development. Fluorescence-activated cell sorting (FACS)-isolated myeloid cells and their progenitors from embryonic skin differentiate into pericytes in culture. At the molecular level, transforming growth factor-β (TGF-β) induces pericyte differentiation in culture. Furthermore, type 2 TGF-β receptor (Tgfbr2) mutants exhibit deficient pericyte development in skin vasculature. Combined, these data suggest that pericytes differentiate from tissue myeloid progenitors in the skin vasculature through TGF-β signaling.
Collapse
Affiliation(s)
- Tomoko Yamazaki
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Ani Nalbandian
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Yutaka Uchida
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Wenling Li
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, 1550 4(th) Street, San Francisco, CA 94158, USA
| | - Yoshiaki Kubota
- Department of Vascular Biology, The Sakaguchi Laboratory, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Seiji Yamamoto
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
181
|
Abstract
The link between inappropriate salt retention in the kidney and hypertension is well recognized. However, growing evidence suggests that the immune system can play surprising roles in sodium homeostasis, such that the study of inflammatory cells and their secreted effectors has provided important insights into salt sensitivity. As part of the innate immune system, myeloid cells have diverse roles in blood pressure regulation, ranging from prohypertensive actions in the kidney, vasculature, and brain, to effects in the skin that attenuate blood pressure elevation. In parallel, T lymphocyte subsets, as key constituents of the adaptive immune compartment, have variable effects on renal sodium handling and the hypertensive response, accruing from the functions of the cytokines that they produce. Conversely, salt can directly modulate the phenotypes of myeloid and T cells, illustrating bidirectional regulatory mechanisms through which sodium and the immune system coordinately impact blood pressure. This review details the complex interplay between myeloid cells, T cells, and salt in the pathogenesis of essential hypertension.
Collapse
Affiliation(s)
- A Justin Rucker
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Durham Veterans Affairs Medical Center, Durham, North Carolina 27705, USA
| | - Nathan P Rudemiller
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Durham Veterans Affairs Medical Center, Durham, North Carolina 27705, USA
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Durham Veterans Affairs Medical Center, Durham, North Carolina 27705, USA
| |
Collapse
|
182
|
Uehara N, Kukita A, Kyumoto-Nakamura Y, Yamaza T, Yasuda H, Kukita T. Osteoblast-derived Laminin-332 is a novel negative regulator of osteoclastogenesis in bone microenvironments. J Transl Med 2017; 97:1235-1244. [PMID: 28581488 DOI: 10.1038/labinvest.2017.55] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/06/2017] [Accepted: 04/06/2017] [Indexed: 01/02/2023] Open
Abstract
Laminin-332 (Lm-332), a major basement membrane protein, has been shown to provide a niche for some stem cells. Here, we found that Lm-332 was expressed in osteoblasts, and is implicated in the regulation of osteoclast differentiation. Immunofluorescence analysis of laminin-β3, a unique component of Lm-332, indicated specific expression of laminin-β3 in osteoblast-like cells localized on bone surface. RT-PCR analysis confirmed that α3, β3, and γ2 chains of Lm-332 were all expressed in primary osteoblasts prepared from mouse calvaria. Lm-332 markedly inhibited osteoclastogenesis induced by receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL) when bone marrow-derived macrophages (BMMs) were cultured on Lm-332-coated plates. Lm-332 also blocked RANKL-induced activation of mitogen-activated protein kinases (MAPKs) (ERK, JNK, and p38) and expression of NFATc1, c-Fos, and c-Jun. Lm-332 suppressed osteoclast differentiation while retaining macrophage phenotypes, including nonspecific esterase activity and gene expression of lysozyme and EGF-like module-containing mucin-like hormone receptor-like 1 (Emr1). Furthermore, the treatment of primary osteoblasts with osteoclastogenic factors dramatically suppressed expression of Lm-332. These findings suggest that Lm-332 produced by osteoblasts in bone tissues has a pivotal role in controlling normal bone remodeling through suppressing osteoclastogenesis.
Collapse
Affiliation(s)
- Norihisa Uehara
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Akiko Kukita
- Department of Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Yukari Kyumoto-Nakamura
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Hisataka Yasuda
- Bioindustry Division, Oriental Yeast Company, Limited, Tokyo, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
183
|
Kramer AC, Blake AL, Taisto ME, Lehrke MJ, Webber BR, Lund TC. Dermatopontin in Bone Marrow Extracellular Matrix Regulates Adherence but Is Dispensable for Murine Hematopoietic Cell Maintenance. Stem Cell Reports 2017; 9:770-778. [PMID: 28844660 PMCID: PMC5599243 DOI: 10.1016/j.stemcr.2017.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 01/03/2023] Open
Abstract
The hematopoietic marrow microenvironment is composed of multiple cell types embedded in an extracellular matrix (ECM). We have explored marrow ECM using mass spectrometry and found dermatopontin (DPT), a small non-collagenous ECM protein, to be present. We found that DPT cooperates with other ECM proteins to promote hematopoietic cell adherence in vitro on plastic as well as OP9 stromal cells. We generated constitutional DPT−/− mice that were viable and had no peripheral lympho-hematopoietic abnormalities. The composition of the marrow of wild-type and DPT−/− mice was equivalent in terms of cellularity, CFU-C, LSK (Lineage−, SCA-1+, KIT+), and LSK-SLAM (LSK, CD48−, CD150+) frequencies. These data suggest that DPT fosters adherence but is not required for steady-state hematopoiesis in vivo. There are likely overlapping cellular adhesion mechanisms that can compensate to maintain the hematopoietic niche in the absence of DPT. Murine marrow can be decellularized to reveal the ECM Dermatopontin is a component of the marrow ECM DPT can promote HSPC adherence in vitro DPT is dispensable for native hematopoiesis
Collapse
Affiliation(s)
- Ashley C Kramer
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota Medical School, MMC 366, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Amanda L Blake
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota Medical School, MMC 366, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Mandy E Taisto
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota Medical School, MMC 366, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Michael J Lehrke
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota Medical School, MMC 366, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Beau R Webber
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota Medical School, MMC 366, 420 Delaware Street SE, Minneapolis, MN 55455, USA; B-MoGen Biotechnologies, Inc., 614 McKinley Place NE, Minneapolis, MN 55413, USA
| | - Troy C Lund
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota Medical School, MMC 366, 420 Delaware Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
184
|
C/EBPβ is required for survival of Ly6C - monocytes. Blood 2017; 130:1809-1818. [PMID: 28807982 DOI: 10.1182/blood-2017-03-772962] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/07/2017] [Indexed: 02/06/2023] Open
Abstract
The transcription factor CCAAT/enhancer-binding protein β (C/EBPβ) is highly expressed in monocytes/macrophages. However, its roles in monopoiesis are largely unknown. Here, we investigated the roles of C/EBPβ in monopoiesis. Further subdivision of monocytes revealed that Cebpb messenger RNA was highly upregulated in Ly6C- monocytes in bone marrow. Accordingly, the number of Ly6C- monocytes was significantly reduced in Cebpb-/- mice. Bone marrow chimera experiments and Mx1-Cre-mediated deletion of Cebpb revealed a cell-intrinsic and monocyte-specific requirement for C/EBPβ in monopoiesis. In Cebpb-/- mice, turnover of Ly6C- monocytes was highly accelerated and apoptosis of Ly6C- monocytes was increased. Expression of Csf1r, which encodes a receptor for macrophage colony-stimulating factor, was significantly reduced in Ly6C- monocytes of Cebpb-/- mice. C/EBPβ bound to positive regulatory elements of Csf1r and promoted its transcription. Collectively, these results indicate that C/EBPβ is a critical factor for Ly6C- monocyte survival, at least in part through upregulation of Csf1r.
Collapse
|
185
|
Mansour A, Wakkach A, Blin-Wakkach C. Emerging Roles of Osteoclasts in the Modulation of Bone Microenvironment and Immune Suppression in Multiple Myeloma. Front Immunol 2017; 8:954. [PMID: 28848556 PMCID: PMC5554508 DOI: 10.3389/fimmu.2017.00954] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 07/26/2017] [Indexed: 12/26/2022] Open
Abstract
Multiple myeloma (MM) is one of the most common forms of hematologic malignancy resulting from cancerous proliferation of mature malignant plasma cells (MPCs). But despite the real improvement in therapeutics in the past years, it remains largely incurable. MM is the most frequent cancer to involve bone due to the stimulation of osteoclast (OCL) differentiation and activity. OCLs have a unique capacity to resorb bone. However, recent studies reveal that they are not restrained to this sole function. They participate in the control of angiogenesis, medullary niches, and immune responses, including in MM. Therefore, therapeutic approaches targeting OCLs probably affect not only bone resorption but also many other functions, and OCLs should not be considered anymore only as targets to improve the bone phenotype but also to modulate bone microenvironment. In this review, we explore these novel contributions of OCLs to MM which reveal their strong implication in the MM physiopathology. We also underline the therapeutic interest of targeting OCLs not only to overcome bone lesions, but also to improve bone microenvironment and anti-tumoral immune responses.
Collapse
Affiliation(s)
- Anna Mansour
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France.,Faculté de Médecine, Université Aix-Marseille, Marseille, France
| | - Abdelilah Wakkach
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| |
Collapse
|
186
|
Katsimbri P. The biology of normal bone remodelling. Eur J Cancer Care (Engl) 2017; 26. [PMID: 28786518 DOI: 10.1111/ecc.12740] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2017] [Indexed: 12/01/2022]
Abstract
During life, bone undergoes modelling and remodelling in order to grow or change shape. Bone modelling is the process by which bones change shape or size in response to physiologic influences or mechanical forces that are encountered by the skeleton, while bone remodelling takes place so that bone may maintain its strength and mineral homeostasis. During early childhood, both bone modelling (the formation and shaping of bone) and bone remodelling (the replacement or renewal of old bone) occur. The predominant process in childhood is bone modelling, while in adulthood bone remodelling predominates. The exception to this is after a fracture when we see massive increases in bone formation. During childhood and adolescence growth occurs in the bones longitudinally and radially, while in the growth plates it occurs longitudinally, thus promoting growth in size. Cartilage first proliferates in the epiphyseal and metaphyseal areas of long bones before undergoing mineralisation to form new bone.
Collapse
Affiliation(s)
- P Katsimbri
- 4th Department of Internal Medicine, "Attikon" University Hospital, Athens, Greece
| |
Collapse
|
187
|
Karmaus PWF, Herrada AA, Guy C, Neale G, Dhungana Y, Long L, Vogel P, Avila J, Clish CB, Chi H. Critical roles of mTORC1 signaling and metabolic reprogramming for M-CSF-mediated myelopoiesis. J Exp Med 2017; 214:2629-2647. [PMID: 28784627 PMCID: PMC5584119 DOI: 10.1084/jem.20161855] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 05/17/2017] [Accepted: 06/22/2017] [Indexed: 12/21/2022] Open
Abstract
Karmaus et al. show that loss of mechanistic target of rapamycin complex 1 (mTORC1) signaling impairs myelopoiesis. M-CSF–dependent myelopoiesis requires mTORC1 signaling and anabolic metabolism, which in turn promote expression of M-CSFR and transcription factors PU.1 and IRF8, thereby constituting a feed-forward loop for myelopoiesis. Myelopoiesis is necessary for the generation of mature myeloid cells during homeostatic turnover and immunological insults; however, the metabolic requirements for this process remain poorly defined. Here, we demonstrate that myelopoiesis, including monocyte and macrophage differentiation, requires mechanistic target of rapamycin complex 1 (mTORC1) signaling and anabolic metabolism. Loss of mTORC1 impaired myelopoiesis under steady state and dampened innate immune responses against Listeria monocytogenes infection. Stimulation of hematopoietic progenitors with macrophage colony-stimulating factor (M-CSF) resulted in mTORC1-dependent anabolic metabolism, which in turn promoted expression of M-CSF receptor and transcription factors PU.1 and IRF8, thereby constituting a feed-forward loop for myelopoiesis. Mechanistically, mTORC1 engaged glucose metabolism and initiated a transcriptional program involving Myc activation and sterol biosynthesis after M-CSF stimulation. Perturbation of glucose metabolism or disruption of Myc function or sterol biosynthesis impaired myeloid differentiation. Integrative metabolomic and genomic profiling further identified one-carbon metabolism as a central node in mTORC1-dependent myelopoiesis. Therefore, the interplay between mTORC1 signaling and metabolic reprogramming underlies M-CSF–induced myelopoiesis.
Collapse
Affiliation(s)
- Peer W F Karmaus
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Andrés A Herrada
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Cliff Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, TN
| | - Yogesh Dhungana
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Lingyun Long
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Julian Avila
- Metabolomics Platform, Broad Institute of MIT and Harvard University, Cambridge, MA
| | - Clary B Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard University, Cambridge, MA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
188
|
Yeo CE, Kang WY, Seong SJ, Cho S, Lee HW, Yoon YR, Kim HJ. Neuromedin B and its receptor silencing suppresses osteoclast generation by modulating precursor proliferation via M-CSF/c-Fms/D-type cyclins. Exp Cell Res 2017; 359:112-119. [PMID: 28780306 DOI: 10.1016/j.yexcr.2017.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 01/16/2023]
Abstract
Neuromedin B (NMB), a mammalian bombesin-like peptide, regulates diverse physiological processes, such as energy metabolism, memory and fear behavior, and cellular growth, through its cognate receptor, NMBR. In this study, we report that NMB expression was upregulated during osteoclast development and that silencing NMB or NMBR attenuated osteoclast generation mediated by macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL). We found that knockdown of NMB or NMBR using a small hairpin RNA suppressed M-CSF-induced proliferation of osteoclast precursor cells without altering osteoclast differentiation. Interestingly, NMB or NMBR knockdown reduced the expression of the M-CSF receptor, c-Fms, which is an important modulator of osteoclast development. Consequently, NMB or NMBR silencing inhibited M-CSF/c-Fms-mediated downstream signaling pathways like activation of ERK and Akt and induction of D-type cyclins, cyclin D1 and D2. Moreover, knockdown of NMB or NMBR accelerated apoptosis in osteoclast lineage cells by inducing caspase-3, caspase-9, and Bim expression. In summary, our study demonstrates that the NMB/NMBR axis plays a pivotal role in osteoclast generation by modulating the proliferation and survival of osteoclast lineage cells.
Collapse
Affiliation(s)
- Chae-Eun Yeo
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu 41944, Republic of Korea
| | - Woo Youl Kang
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu 41944, Republic of Korea
| | - Sook Jin Seong
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu 41944, Republic of Korea
| | - Seungil Cho
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu 41944, Republic of Korea
| | - Hae Won Lee
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu 41944, Republic of Korea
| | - Young-Ran Yoon
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu 41944, Republic of Korea.
| | - Hyun-Ju Kim
- Department of Biomedical Science, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Clinical Trial Center, School of Medicine, Kyungpook National University and Hospital, Daegu 41944, Republic of Korea.
| |
Collapse
|
189
|
|
190
|
Abstract
We review here the macrophages found in endocrine tissues, placing emphasis on those residing in the islets of Langerhans of the pancreas. The islets represent the endocrine organ where macrophages have been examined in great detail and where our own studies and experience have been directed.
Collapse
|
191
|
Yamamoto S, Muramatsu M, Azuma E, Ikutani M, Nagai Y, Sagara H, Koo BN, Kita S, O'Donnell E, Osawa T, Takahashi H, Takano KI, Dohmoto M, Sugimori M, Usui I, Watanabe Y, Hatakeyama N, Iwamoto T, Komuro I, Takatsu K, Tobe K, Niida S, Matsuda N, Shibuya M, Sasahara M. A subset of cerebrovascular pericytes originates from mature macrophages in the very early phase of vascular development in CNS. Sci Rep 2017. [PMID: 28634350 PMCID: PMC5478595 DOI: 10.1038/s41598-017-03994-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pericytes are believed to originate from either mesenchymal or neural crest cells. It has recently been reported that pericytes play important roles in the central nervous system (CNS) by regulating blood-brain barrier homeostasis and blood flow at the capillary level. However, the origin of CNS microvascular pericytes and the mechanism of their recruitment remain unknown. Here, we show a new source of cerebrovascular pericytes during neurogenesis. In the CNS of embryonic day 10.5 mouse embryos, CD31+F4/80+ hematopoietic lineage cells were observed in the avascular region around the dorsal midline of the developing midbrain. These cells expressed additional macrophage markers such as CD206 and CD11b. Moreover, the CD31+F4/80+ cells phagocytosed apoptotic cells as functionally matured macrophages, adhered to the newly formed subventricular vascular plexus, and then divided into daughter cells. Eventually, these CD31+F4/80+ cells transdifferentiated into NG2/PDGFRβ/desmin-expressing cerebrovascular pericytes, enwrapping and associating with vascular endothelial cells. These data indicate that a subset of cerebrovascular pericytes derive from mature macrophages in the very early phase of CNS vascular development, which in turn are recruited from sites of embryonic hematopoiesis such as the yolk sac by way of blood flow.
Collapse
Affiliation(s)
- Seiji Yamamoto
- Department of Pathology, University of Toyama, Toyama, Japan.
| | - Masashi Muramatsu
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Erika Azuma
- Department of Pathology, University of Toyama, Toyama, Japan.,Department of Technology Development, Astellas Pharma Tech Co., Ltd., Toyama, Japan
| | - Masashi Ikutani
- Department of Immunobiology and Pharmacological Genetics, University of Toyama, Toyama, Japan
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, University of Toyama, Toyama, Japan.,JST, PRESTO, Kawaguchi, Saitama, Japan
| | - Hiroshi Sagara
- Medical Proteomics Laboratory, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Bon-Nyeo Koo
- Department of Anesthesiology, Yonsei University College of Medicine, Seoul, Korea.,Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Maryland, USA
| | - Satomi Kita
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Erin O'Donnell
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Maryland, USA
| | - Tsuyoshi Osawa
- Laboratry for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Takahashi
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
| | - Ken-Ichi Takano
- Departments of Pharmacology, Weill Cornell Medical College, New York, USA
| | - Mitsuko Dohmoto
- Genome Biotechnology Laboratory, Kanazawa Institute of Technology, Ishikawa, Japan
| | - Michiya Sugimori
- Department of Integrative Neuroscience, University of Toyama, Toyama, Japan
| | - Isao Usui
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Yasuhide Watanabe
- Faculty of Medicine, School of Nursing, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Noboru Hatakeyama
- Department of Anesthesiology, Graduate School of Medicine, Aichi Medical University, Aichi, Japan
| | - Takahiro Iwamoto
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kiyoshi Takatsu
- Department of Immunobiology and Pharmacological Genetics, University of Toyama, Toyama, Japan.,Toyama Prefectural Institute for Pharmaceutical Research, Toyama, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Shumpei Niida
- Medical Genome Center, Center for Geriatrics and Gerontology, Aichi, Japan
| | - Naoyuki Matsuda
- Department of Emergency and Critical Care Medicine, Nagoya University, Nagoya, Japan
| | - Masabumi Shibuya
- Department of Research and Education, Jobu University, Gunma, Japan
| | | |
Collapse
|
192
|
Dwyer AR, Greenland EL, Pixley FJ. Promotion of Tumor Invasion by Tumor-Associated Macrophages: The Role of CSF-1-Activated Phosphatidylinositol 3 Kinase and Src Family Kinase Motility Signaling. Cancers (Basel) 2017; 9:E68. [PMID: 28629162 PMCID: PMC5483887 DOI: 10.3390/cancers9060068] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/08/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022] Open
Abstract
Macrophages interact with cells in every organ to facilitate tissue development, function and repair. However, the close interaction between macrophages and parenchymal cells can be subverted in disease, particularly cancer. Motility is an essential capacity for macrophages to be able to carry out their various roles. In cancers, the macrophage's interstitial migratory ability is frequently co-opted by tumor cells to enable escape from the primary tumor and metastatic spread. Macrophage accumulation within and movement through a tumor is often stimulated by tumor cell production of the mononuclear phagocytic growth factor, colony-stimulating factor-1 (CSF-1). CSF-1 also regulates macrophage survival, proliferation and differentiation, and its many effects are transduced by its receptor, the CSF-1R, via phosphotyrosine motif-activated signals. Mutational analysis of CSF-1R signaling indicates that the major mediators of CSF-1-induced motility are phosphatidyl-inositol-3 kinase (PI3K) and one or more Src family kinase (SFK), which activate signals to adhesion, actin polymerization, polarization and, ultimately, migration and invasion in macrophages. The macrophage transcriptome, including that of the motility machinery, is very complex and highly responsive to the environment, with selective expression of proteins and splice variants rarely found in other cell types. Thus, their unique motility machinery can be specifically targeted to block macrophage migration, and thereby, inhibit tumor invasion and metastasis.
Collapse
Affiliation(s)
- Amy R Dwyer
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Eloise L Greenland
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Fiona J Pixley
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| |
Collapse
|
193
|
Corrado A, Maruotti N, Cantatore FP. Osteoblast Role in Rheumatic Diseases. Int J Mol Sci 2017; 18:ijms18061272. [PMID: 28617323 PMCID: PMC5486094 DOI: 10.3390/ijms18061272] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 06/07/2017] [Accepted: 06/12/2017] [Indexed: 12/11/2022] Open
Abstract
Alterations in osteoblast growth, differentiation and activity play a role in the pathogenesis of several rheumatic diseases, such as rheumatoid arthritis, spondyloarthritides, osteoarthritis, and osteoporosis. In fact, in these rheumatic diseases, abnormal activity of Wnt signaling, receptor activator of nuclear factor-κB (RANK)-RANK ligand (RANKL)-osteoprotegerin (OPG) signaling, bone morphogenetic proteins (BMPs) pathway and other mechanisms have been described in osteoblasts. This review article is focused on current knowledge on the role of osteoblast dysregulation occurring in rheumatic diseases.
Collapse
Affiliation(s)
- Addolorata Corrado
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia Medical School, 71122 Foggia, Italy.
| | - Nicola Maruotti
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia Medical School, 71122 Foggia, Italy.
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia Medical School, 71122 Foggia, Italy.
| |
Collapse
|
194
|
Nakamichi Y, Udagawa N, Horibe K, Mizoguchi T, Yamamoto Y, Nakamura T, Hosoya A, Kato S, Suda T, Takahashi N. VDR in Osteoblast-Lineage Cells Primarily Mediates Vitamin D Treatment-Induced Increase in Bone Mass by Suppressing Bone Resorption. J Bone Miner Res 2017; 32:1297-1308. [PMID: 28177161 DOI: 10.1002/jbmr.3096] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 02/01/2017] [Accepted: 02/03/2017] [Indexed: 01/01/2023]
Abstract
Long-term treatment with active vitamin D [1α,25(OH)2 D3 ] and its derivatives is effective for increasing bone mass in patients with primary and secondary osteoporosis. Derivatives of 1α,25(OH)2 D3 , including eldecalcitol (ELD), exert their actions through the vitamin D receptor (VDR). ELD is more resistant to metabolic degradation than 1α,25(OH)2 D3 . It is reported that ELD treatment causes a net increase in bone mass by suppressing bone resorption rather than by increasing bone formation in animals and humans. VDR in bone and extraskeletal tissues regulates bone mass and secretion of osteotropic hormones. Therefore, it is unclear what types of cells expressing VDR preferentially regulate the vitamin D-induced increase in bone mass. Here, we examined the effects of 4-week treatment with ELD (50 ng/kg/day) on bone using osteoblast lineage-specific VDR conditional knockout (Ob-VDR-cKO) and osteoclast-specific VDR cKO (Ocl-VDR-cKO) male mice aged 10 weeks. Immunohistochemically, VDR in bone was detected preferentially in osteoblasts and osteocytes. Ob-VDR-cKO mice showed normal bone phenotypes, despite no appreciable immunostaining of VDR in bone. Ob-VDR-cKO mice failed to increase bone mass in response to ELD treatment. Ocl-VDR-cKO mice also exhibited normal bone phenotypes, but normally responded to ELD. ELD-induced FGF23 production in bone was regulated by VDR in osteoblast-lineage cells. These findings suggest that the vitamin D treatment-induced increase in bone mass is mediated by suppressing bone resorption through VDR in osteoblast-lineage cells. © 2017 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Yuko Nakamichi
- Institute for Oral Science, Matsumoto Dental University, Nagano, Japan
| | - Nobuyuki Udagawa
- Department of Biochemistry, Matsumoto Dental University, Nagano, Japan
| | - Kanji Horibe
- Department of Oral Histology, Matsumoto Dental University, Nagano, Japan
| | | | - Yoko Yamamoto
- Department of Surgical Oncology, The University of Tokyo, Tokyo, Japan
| | - Takashi Nakamura
- Department of Biochemistry & Integrative Medical Biology, School of Medicine, Keio University, Tokyo, Japan
| | - Akihiro Hosoya
- Department of Oral Histology, Matsumoto Dental University, Nagano, Japan
| | - Shigeaki Kato
- Jyoban Hospital, Tokiwa Foundation, Fukushima, Japan
| | - Tatsuo Suda
- Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Naoyuki Takahashi
- Institute for Oral Science, Matsumoto Dental University, Nagano, Japan
| |
Collapse
|
195
|
Yoshimura Y, Kikuiri T, Hasegawa T, Matsuno M, Minamikawa H, Deyama Y, Suzuki K. How much medium do you use for cell culture? Medium volume influences mineralization and osteoclastogenesis in vitro. Mol Med Rep 2017; 16:429-434. [PMID: 28535008 DOI: 10.3892/mmr.2017.6611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/15/2017] [Indexed: 11/06/2022] Open
Abstract
Bone is maintained by a balance between bone formation and resorption. This remodeling is controlled by a wide variety of systemic and local factors including hormones, cytokines and mechanical stresses. The present in vitro study examined the impact of medium volume, using 0.4, 0.6, 0.8, 1.0, 1.5 and 2.0 ml/well in a 24‑well plate, on the differentiation of osteoblasts and osteoclasts. There were no differences in the alkaline phosphatase activity of osteoblasts amongst the groups; however, the area of mineral deposition was decreased in a media volume‑dependent manner. A co‑culture of osteoblastic cells with bone marrow cells revealed a reduction in the total number of osteoclastic tartrate‑resistant acid phosphatase (TRAP)‑positive multinuclear cells (≥2 nuclei), whereas the formation of large osteoclastic TRAP‑positive multinuclear cells (≥8 nuclei) was increased, in a media volume‑dependent manner. There were also no differences in receptor activator of nuclear factor‑κB ligand mRNA and total osteoprotegerin (OPG) protein expression levels amongst the groups, however the concentration of OPG decreased in a media volume‑dependent manner. In conclusion, the present study demonstrated that the suppression of mineralization in osteoblastic cells and the stimulation of osteoclast fusion are dependent on the medium volume, indicating that media volume is an important factor in in vitro cell culture systems.
Collapse
Affiliation(s)
- Yoshitaka Yoshimura
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Kita‑ku, Sapporo, Hokkaido 060‑8586, Japan
| | - Takashi Kikuiri
- Department of Pediatric Dentistry, Hokkaido University Graduate School of Dental Medicine, Kita‑ku, Sapporo, Hokkaido 060‑8586, Japan
| | - Tomokazu Hasegawa
- Department of Pediatric Dentistry, Faculty of Dentistry, Tokushima University, Tokushima, Tokushima 770‑8504, Japan
| | - Mino Matsuno
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Kita‑ku, Sapporo, Hokkaido 060‑8586, Japan
| | - Hajime Minamikawa
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Kita‑ku, Sapporo, Hokkaido 060‑8586, Japan
| | - Yoshiaki Deyama
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Kita‑ku, Sapporo, Hokkaido 060‑8586, Japan
| | - Kuniaki Suzuki
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Kita‑ku, Sapporo, Hokkaido 060‑8586, Japan
| |
Collapse
|
196
|
Murakami A, Matsuda M, Harada Y, Hirata M. Phospholipase C-related, but catalytically inactive protein (PRIP) up-regulates osteoclast differentiation via calcium-calcineurin-NFATc1 signaling. J Biol Chem 2017; 292:7994-8006. [PMID: 28341745 PMCID: PMC5427276 DOI: 10.1074/jbc.m117.784777] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 03/23/2017] [Indexed: 01/08/2023] Open
Abstract
Phospholipase C-related, but catalytically inactive protein (PRIP) was previously identified as a novel inositol 1,4,5-trisphosphate-binding protein with a domain organization similar to that of phospholipase C-δ but lacking phospholipase activity. We recently showed that PRIP gene knock-out (KO) in mice increases bone formation and concomitantly decreases bone resorption, resulting in increased bone mineral density and trabecular bone volume. However, the role of PRIP in osteoclastogenesis has not yet been fully elucidated. Here, we investigated the effects of PRIP on bone remodeling by investigating dynamic tooth movement in mice fitted with orthodontic devices. Morphological analysis indicated that the extent of tooth movement was smaller in the PRIP-KO mice than in wild-type mice. Histological analysis revealed fewer osteoclasts on the bone-resorption side in maxillary bones of PRIP-KO mice, and osteoclast formation assays and flow cytometry indicated lower osteoclast differentiation in bone marrow cells isolated from these mice. The expression of genes implicated in bone resorption was lower in differentiated PRIP-KO cells, and genes involved in osteoclast differentiation, such as the transcription factor NFATc1, exhibited lower expression in immature PRIP-KO cells initiated by M-CSF. Moreover, calcineurin expression and activity were also lower in the PRIP-KO cells. The PRIP-KO cells also displayed fewer M-CSF-induced changes in intracellular Ca2+ and exhibited reduced nuclear localization of NFATc1. Up-regulation of intracellular Ca2+ restored osteoclastogenesis of the PRIP-KO cells. These results indicate that PRIP deficiency impairs osteoclast differentiation, particularly at the early stages, and that PRIP stimulates osteoclast differentiation through calcium-calcineurin-NFATc1 signaling via regulating intracellular Ca2.
Collapse
Affiliation(s)
- Ayako Murakami
- From the Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, and
| | - Miho Matsuda
- From the Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, and
| | - Yui Harada
- R&D Laboratory for Innovative Biotherapeutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan and
| | - Masato Hirata
- From the Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, and
- the Fukuoka Dental College, Fukuoka 814-0175, Japan
| |
Collapse
|
197
|
Immune microenvironment of gliomas. J Transl Med 2017; 97:498-518. [PMID: 28287634 DOI: 10.1038/labinvest.2017.19] [Citation(s) in RCA: 388] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/13/2022] Open
Abstract
High-grade gliomas are rapidly progressing tumors of the central nervous system (CNS) with a very poor prognosis despite extensive resection combined with radiation and/or chemotherapy. Histopathological and flow cytometry analyses of human and rodent experimental gliomas revealed heterogeneity of a tumor and its niche, composed of reactive astrocytes, endothelial cells, and numerous immune cells. Infiltrating immune cells consist of CNS resident (microglia) and peripheral macrophages, granulocytes, myeloid-derived suppressor cells (MDSCs), and T lymphocytes. Intratumoral density of glioma-associated microglia/macrophages (GAMs) and MDSCs is the highest in malignant gliomas and inversely correlates with patient survival. Although GAMs have a few innate immune functions intact, their ability to be stimulated via TLRs, secrete cytokines, and upregulate co-stimulatory molecules is not sufficient to initiate antitumor immune responses. Moreover, tumor-reprogrammed GAMs release immunosuppressive cytokines and chemokines shaping antitumor responses. Both GAMs and MDSCs have ability to attract T regulatory lymphocytes to the tumor, but MDSCs inhibit cytotoxic responses mediated by natural killer cells, and block the activation of tumor-reactive CD4+ T helper cells and cytotoxic CD8+ T cells. The presence of regulatory T cells may further contribute to the lack of effective immune activation against malignant gliomas. We review the immunological aspects of glioma microenvironment, in particular composition and various roles of the immune cells infiltrating malignant human gliomas and experimental rodent gliomas. We describe tumor-derived signals and mechanisms driving myeloid cell accumulation and reprogramming. Although, understanding the complexity of cell-cell interactions in glioma microenvironment is far from being achieved, recent studies demonstrated several glioma-derived factors that trigger migration, accumulation, and reprogramming of immune cells. Identification of these factors may facilitate development of immunotherapy for gliomas as immunomodulatory and immune evasion mechanisms employed by malignant gliomas pose an appalling challenge to brain tumor immunotherapy.
Collapse
|
198
|
Enomoto T, Takami M, Yamamoto M, Kamijo R. LPS administration increases CD11b + c-Fms + CD14 + cell population that possesses osteoclast differentiation potential in mice. Cytotechnology 2017; 69:529-537. [PMID: 28429166 DOI: 10.1007/s10616-017-0094-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/04/2017] [Indexed: 01/09/2023] Open
Abstract
Osteoclasts are multinucleated giant cells that originate from a monocyte/macrophage lineage, and are involved in the inflammatory bone destruction accompanied by periodontitis. Recent studies have shown that osteoclast precursors reside not only in the bone marrow, but also in the peripheral blood and spleen, though the precise characteristics of each precursor have not been analyzed. We hypothesized that the number of osteoclast precursors in those tissues may increase under pathological conditions and contribute to osteoclast formation in vivo in a mouse model. To test this hypothesis, we attempted to identify cell populations that possess osteoclast differentiation potential in the bone marrow, spleen, and blood by analyzing macrophage/monocyte-related cell surface markers such as CD11b, CD14, and colony-stimulating factor-1 receptor (c-Fms). In the bone marrow, the CD11b- cell population, but not the CD11b+ cell population, differentiated into osteoclasts in the presence of receptor activator of nuclear factor-κB ligand and macrophage colony-stimulating factor. On the other hand, in the spleen and blood, CD11b+ cells differentiated into osteoclasts. Interestingly, lipopolysaccharide (LPS) administration to the mice dramatically increased the proportion of CD11b+ c-Fms+ CD14+ cells, which differentiated into osteoclasts, in the bone marrow and spleen. These results suggest that LPS administration increases the proportion of a distinct cell population expressing CD11b+, c-Fms+, and CD14+ in the bone marrow and spleen. Thus, these cell populations are considered to contribute to the increase in osteoclast number during inflammatory bone destruction such as periodontitis.
Collapse
Affiliation(s)
- Takuya Enomoto
- Department of Biochemistry, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
- Department of Periodontology, Showa University Dental Hospital, 2-1-1 Kitasenzoku, Ota, Tokyo, 145-8515, Japan
| | - Masamichi Takami
- Department of Biochemistry, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan.
- Department of Dental Pharmacology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan.
| | - Matsuo Yamamoto
- Department of Periodontology, Showa University Dental Hospital, 2-1-1 Kitasenzoku, Ota, Tokyo, 145-8515, Japan
| | - Ryutaro Kamijo
- Department of Biochemistry, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| |
Collapse
|
199
|
Urano-Morisawa E, Takami M, Suzawa T, Matsumoto A, Osumi N, Baba K, Kamijo R. Induction of osteoblastic differentiation of neural crest-derived stem cells from hair follicles. PLoS One 2017; 12:e0174940. [PMID: 28384239 PMCID: PMC5383073 DOI: 10.1371/journal.pone.0174940] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 03/17/2017] [Indexed: 12/12/2022] Open
Abstract
The neural crest (NC) arises near the neural tube during embryo development. NC cells migrate throughout the embryo and have potential to differentiate into multiple cell types, such as peripheral nerves, glial, cardiac smooth muscle, endocrine, and pigment cells, and craniofacial bone. In the present study, we induced osteoblast-like cells using whisker follicles obtained from the NC of mice. Hair follicle cells derived from the NC labeled with enhanced green fluorescent protein (EGFP) were collected from protein zero-Cre/floxed-EGFP double transgenic mice and cultured, then treated and cultured in stem cell growth medium. After growth for 14 days, results of flow cytometry analysis showed that 95% of the EGFP-positive (EGFP+) hair follicle cells derived from the NC had proliferated and 76.2% of those expressed mesenchymal stem cells markers, such as platelet-derived growth factor α and stem cell antigen-1, and also showed constitutive expression of Runx2 mRNA. Cells stimulated with bone morphogenetic protein-2 expressed osteocalcin, osterix, and alkaline phosphatase mRNA, resulting in production of mineralized matrices, which were detected by von Kossa and alizarin red staining. Moreover, EGFP+ hair follicle cells consistently expressed macrophage colony-stimulating factor and osteoprotegerin (OPG). Addition of 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3] (10-8 M) to the cultures suppressed OPG expression and induced RANKL production in the cells. Furthermore, multinucleated osteoclasts appeared within 6 days after starting co-cultures of bone marrow cells with EGFP+ cells in the presence of 1,25(OH)2D3 and PGE2. These results suggest that NC-derived hair follicle cells possess a capacity for osteoblastic differentiation and may be useful for developing new bone regenerative medicine therapies.
Collapse
Affiliation(s)
- Eri Urano-Morisawa
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
- Department of Prosthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Masamichi Takami
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
- Department of Pharmacology, School of Dentistry, Showa University, Tokyo, Japan
- * E-mail:
| | - Tetsuo Suzawa
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
| | - Akifumi Matsumoto
- Department of Prosthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Noriko Osumi
- Division of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuyoshi Baba
- Department of Prosthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Ryutaro Kamijo
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
| |
Collapse
|
200
|
Lee B, Iwaniec UT, Turner RT, Lin YW, Clarke BL, Gingery A, Wei LN. RIP140 in monocytes/macrophages regulates osteoclast differentiation and bone homeostasis. JCI Insight 2017; 2:e90517. [PMID: 28405613 DOI: 10.1172/jci.insight.90517] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Osteolytic bone diseases, such as osteoporosis, are characterized by diminished bone quality and increased fracture risk. The therapeutic challenge remains to maintain bone homeostasis with a balance between osteoclast-mediated resorption and osteoblast-mediated formation. Osteoclasts are formed by the fusion of monocyte/macrophage-derived precursors. Here we report, to our knowledge for the first time, that receptor-interacting protein 140 (RIP140) expression in osteoclast precursors and its protein regulation are crucial for osteoclast differentiation, activity, and coupled bone formation. In mice, monocyte/macrophage-specific knockdown of RIP140 (mϕRIP140KD) resulted in a cancellous osteopenic phenotype with significantly increased bone resorption and reduced bone formation. Osteoclast precursors isolated from mϕRIP140KD mice had significantly increased differentiation potential. Furthermore, conditioned media from mϕRIP140KD primary osteoclast cultures significantly suppressed osteoblast differentiation. This suppressive activity was effectively and rapidly terminated by specific Syk-stimulated RIP140 protein degradation. Mechanistic analysis revealed that RIP140 functions primarily by inhibiting osteoclast differentiation through forming a transcription-suppressor complex with testicular receptor 4 (TR4) to repress osteoclastogenic genes. These data reveal that monocyte/macrophage RIP140/TR4 complexes may serve as a critical transcription regulatory complex maintaining homeostasis of osteoclast differentiation, activity, and coupling with osteoblast formation. Accordingly, we propose a potentially novel therapeutic strategy, specifically targeting osteoclast precursor RIP140 protein in osteolytic bone diseases.
Collapse
Affiliation(s)
- Bomi Lee
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Russell T Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Yi-Wei Lin
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Bart L Clarke
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Anne Gingery
- Division of Orthopedic Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|