151
|
Tosato G. Ephrin ligands and Eph receptors contribution to hematopoiesis. Cell Mol Life Sci 2017; 74:3377-3394. [PMID: 28589441 PMCID: PMC11107787 DOI: 10.1007/s00018-017-2566-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/12/2017] [Accepted: 06/01/2017] [Indexed: 12/12/2022]
Abstract
Hematopoietic stem and progenitor cells reside predominantly in the bone marrow. They supply billions of mature blood cells every day during life through maturation into multilineage progenitors and self-renewal. Newly produced mature cells serve to replenish the pool of circulating blood cells at the end of their life-span. These mature blood cells and a few hematopoietic progenitors normally exit the bone marrow through the sinusoidal vessels, a specialized venous vascular system that spreads throughout the bone marrow. Many signals regulate the coordinated mobilization of hematopoietic cells from the bone marrow to the circulation. In this review, we present recent advances on hematopoiesis and hematopoietic cell mobilization with a focus on the role of Ephrin ligands and their Eph receptors. These constitute a large family of transmembrane ligands and receptors that play critical roles in development and postnatally. New insights point to distinct roles of ephrin and Eph in different aspects of hematopoiesis.
Collapse
Affiliation(s)
- Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 4124, Bethesda, MD, 20892, USA.
| |
Collapse
|
152
|
Kumar S, Geiger H. HSC Niche Biology and HSC Expansion Ex Vivo. Trends Mol Med 2017; 23:799-819. [PMID: 28801069 DOI: 10.1016/j.molmed.2017.07.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 02/08/2023]
Abstract
Hematopoietic stem cell (HSC) transplantation can restore a new functional hematopoietic system in recipients in cases where the system of the recipient is not functional or for example is leukemic. However, the number of available donor HSCs is often too low for successful transplantation. Expansion of HSCs and thus HSC self-renewal ex vivo would greatly improve transplantation therapy in the clinic. In vivo, HSCs expand significantly in the niche, but establishing protocols that result in HSC expansion ex vivo remains challenging. In this review we discuss current knowledge of niche biology, the intrinsic regulators of HSC self-renewal in vivo, and introduce novel niche-informed strategies of HSC expansion ex vivo.
Collapse
Affiliation(s)
- Sachin Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA.
| | - Hartmut Geiger
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA; Institute of Molecular Medicine, Ulm University, Ulm, Germany; Aging Research Center, Ulm University, Ulm, Germany.
| |
Collapse
|
153
|
Bruserud Ø, Aasebø E, Hernandez-Valladares M, Tsykunova G, Reikvam H. Therapeutic targeting of leukemic stem cells in acute myeloid leukemia - the biological background for possible strategies. Expert Opin Drug Discov 2017; 12:1053-1065. [PMID: 28748730 DOI: 10.1080/17460441.2017.1356818] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is an aggressive malignancy, caused by the accumulation of immature leukemic blasts in blood and bone marrow. There is a relatively high risk of chemoresistant relapse even for the younger patients who can receive the most intensive antileukemic treatment. Treatment directed against the remaining leukemic and preleukemic stem cells will most likely reduce the risk of later relapse. Areas covered: Relevant publications were identified through literature searches. The authors searched for original articles and recent reviews describing (i) the characteristics of leukemic/preleukemic stem cells; (ii) the importance of the bone marrow stem cell niches in leukemogenesis; and (iii) possible therapeutic strategies to target the preleukemic/leukemic stem cells. Expert opinion: Leukemia relapse/progression seems to be derived from residual chemoresistant leukemic or preleukemic stem cells, and a more effective treatment directed against these cells will likely be important to improve survival both for patients receiving intensive treatment and leukemia-stabilizing therapy. Several possible strategies are now considered, including the targeting of the epigenetic regulation of gene expression, proapoptotic intracellular signaling, cell metabolism, telomere activity and the AML-supporting effects by neighboring stromal cells. Due to disease heterogeneity, the most effective stem cell-directed therapy will probably differ between individual patients.
Collapse
Affiliation(s)
- Øystein Bruserud
- a Division of Hematology, Institute of Clinical Science , University of Bergen , Bergen , Norway.,b Section of Hematology, Department of Medicine , Haukeland University Hospital , Bergen , Norway
| | - Elise Aasebø
- a Division of Hematology, Institute of Clinical Science , University of Bergen , Bergen , Norway.,c Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Maria Hernandez-Valladares
- a Division of Hematology, Institute of Clinical Science , University of Bergen , Bergen , Norway.,c Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Galina Tsykunova
- b Section of Hematology, Department of Medicine , Haukeland University Hospital , Bergen , Norway
| | - Håkon Reikvam
- b Section of Hematology, Department of Medicine , Haukeland University Hospital , Bergen , Norway
| |
Collapse
|
154
|
Yuan YH, Zhao SS, Wang XL, Teng ZP, Li DS, Zeng Y. HIV-1 p55-gag protein induces senescence of human bone marrow mesenchymal stem cells and reduces their capacity to support expansion of hematopoietic stem cells in vitro. Cell Biol Int 2017; 41:969-981. [PMID: 28544005 DOI: 10.1002/cbin.10791] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/09/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Ya-hong Yuan
- College of Life Science and Bioengineering; Beijing University of Technology; Beijing 100124 China
- Hubei Key Laboratory of Embryonic Stem Cell Research; Taihe Hospital; Hubei University of Medicine; Shiyan 442000 Hubei China
| | - Shan-shan Zhao
- Hubei Key Laboratory of Embryonic Stem Cell Research; Taihe Hospital; Hubei University of Medicine; Shiyan 442000 Hubei China
| | - Xiao-li Wang
- College of Life Science and Bioengineering; Beijing University of Technology; Beijing 100124 China
- Hubei Key Laboratory of Embryonic Stem Cell Research; Taihe Hospital; Hubei University of Medicine; Shiyan 442000 Hubei China
| | - Zhi-ping Teng
- Institute of Virology; Chinese Academy of Preventive Medicine; Beijing China
| | - Dong-sheng Li
- Hubei Key Laboratory of Embryonic Stem Cell Research; Taihe Hospital; Hubei University of Medicine; Shiyan 442000 Hubei China
| | - Yi Zeng
- College of Life Science and Bioengineering; Beijing University of Technology; Beijing 100124 China
| |
Collapse
|
155
|
Rao KS, Spees JL. Harnessing Epicardial Progenitor Cells and Their Derivatives for Rescue and Repair of Cardiac Tissue After Myocardial Infarction. ACTA ACUST UNITED AC 2017; 3:149-158. [PMID: 29057207 DOI: 10.1007/s40610-017-0066-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Ischemic heart disease and stroke lead to the greatest number of deaths worldwide. Despite decreased time to intervention and improvements in the standard of care, 1 out of 5 patients that survive a myocardial infarction (MI) still face long-term chronic heart failure and a 5-year mortality rate of about 50%. Based on their multi-potency for differentiation and paracrine activity, epicardial cells and their derivatives have potential to rescue jeopardized tissue and/or promote cardiac regeneration. Here we review the diagnosis and treatment of MI, basic epicardial cell biology, and potential treatment strategies designed to harness the reparative properties of epicardial cells. RECENT FINDINGS During cardiac development, epicardial cells covering the surface of the heart generate migratory progenitor cells that contribute to the coronary vasculature and the interstitial fibroblasts. Epicardial cells also produce paracrine signals required for myocardial expansion and cardiac growth. In adults with myocardial infarction, epicardial cells and their derivatives provide paracrine factors that affect myocardial remodeling and repair. At present, the intrinsic mechanisms and extrinsic signals that regulate epicardial cell fate and paracrine activity in adults remain poorly understood. SUMMARY Human diseases that result in heart failure due to negative remodeling or extensive loss of viable cardiac tissue require new, effective treatments. Improved understanding of epicardial cell function(s) and epicardial-mediated secretion of growth factors, cytokines and hormones during cardiac growth, homeostasis and injury may lead to new ways to treat patients with myocardial infarction.
Collapse
Affiliation(s)
- Krithika S Rao
- Department of Medicine, Stem Cell Core, University of Vermont, Colchester, VT 05446
- Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446
| | - Jeffrey L Spees
- Department of Medicine, Stem Cell Core, University of Vermont, Colchester, VT 05446
- Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446
| |
Collapse
|
156
|
Abstract
In response to myocardial infarction (MI), time-dependent leukocyte infiltration is critical to program the acute inflammatory response. Post-MI leukocyte density, residence time in the infarcted area, and exit from the infarcted injury predict resolving or nonresolving inflammation. Overactive or unresolved inflammation is the primary determinant in heart failure pathology post-MI. Here, our review describes supporting evidence that the acute inflammatory response also guides the generation of healing and regenerative mediators after cardiac damage. Time-dependent leukocyte density and diversity and the magnitude of myocardial injury is responsible for the resolving and nonresolving pathway in myocardial healing. Post MI, the diversity of leukocytes, such as neutrophils, macrophages, and lymphocytes, has been explored that regulate the clearance of deceased cardiomyocytes by using the classic and reparative pathways. Among the innovative factors and intermediates that have been recognized as essential in acute the self-healing and clearance mechanism, we highlight specialized proresolving mediators as the emerging factor for post-MI reparative mechanisms-translational leukocyte modifiers, such as aging, the source of leukocytes, and the milieu around the leukocytes. In the clinical setting, it is possible that leukocyte diversity is more prominent as a result of risk factors, such as obesity, diabetes, and hypertension. Pharmacologic agents are critical modifiers of leukocyte diversity in healing mechanisms that may impair or stimulate the clearance mechanism. Future research is needed, with a focused approach to understand the molecular targets, cellular effectors, and receptors. A clear understanding of resolving and nonresolving inflammation in myocardial healing will help to develop novel targets with major emphasis on the resolution of inflammation in heart failure pathology.-Tourki, B., Halade, G. Leukocyte diversity in resolving and nonresolving mechanisms of cardiac remodeling.
Collapse
Affiliation(s)
- Bochra Tourki
- Laboratoire des Venins et Biomolécules Thérapeutiques et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires, Institut Pasteur de Tunis, Université Carthage Tunis, Carthage, Tunisia
| | - Ganesh Halade
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
157
|
Maeda Y, Yonemochi Y, Nakajyo Y, Hidaka H, Ikeda T, Ando Y. CXCL12 and osteopontin from bone marrow-derived mesenchymal stromal cells improve muscle regeneration. Sci Rep 2017; 7:3305. [PMID: 28607396 PMCID: PMC5468354 DOI: 10.1038/s41598-017-02928-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/30/2017] [Indexed: 01/01/2023] Open
Abstract
Muscle satellite cells are essential for muscle regeneration. However, efficient regeneration does not occur without muscle-resident mesenchymal progenitor cells. We show here that bone marrow-derived mesenchymal stromal cells (Bm-MSCs) also facilitate muscle regeneration in Duchenne muscular dystrophy (DMD) model mice. Bm-MSCs transplanted into peritoneal cavities of DMD model mice with severe muscle degeneration strongly suppressed dystrophic pathology and improved death-related symptoms, which resulted in dramatic lifespan extension. Isolated single myofibers from Bm-MSC-transplanted mice manifested considerably less myofiber splitting compared with myofibers from non-transplanted mice, which indicated that transplantation significantly ameliorated abnormal regeneration. With regard to the number of satellite cells, several cells remained on myofibers from Bm-MSC-transplanted model mice, but satellite cells rarely occurred on myofibers from non-transplanted mice. Also, CXCL12 was crucial for muscle regeneration. CXCL12 facilitated muscle regeneration and paired box protein–7 (PAX7) expression after cardiotoxin-related muscle injury in vivo. The majority of primary muscle satellite cells sorted by integrin-α7 and CD34 expressed CXCR4, a receptor specific for CXCL12. CXCL12 strongly suppressed p-STAT3 expression in these sorted cells in vitro. CXCL12 may therefore influence muscle regeneration through STAT3 signaling in satellite cells. Targeting these proteins in or on muscle satellite cells may improve many degenerative muscle diseases.
Collapse
Affiliation(s)
- Yasushi Maeda
- Department of Neurology, National Hospital Organization Kumamoto Saishunso National Hospital, Kumamoto, Japan.
| | - Yasuhiro Yonemochi
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Nakajyo
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideaki Hidaka
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tokunori Ikeda
- Department of Clinical Research Center, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
158
|
Papy-Garcia D, Albanese P. Heparan sulfate proteoglycans as key regulators of the mesenchymal niche of hematopoietic stem cells. Glycoconj J 2017; 34:377-391. [PMID: 28577070 DOI: 10.1007/s10719-017-9773-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 12/21/2022]
Abstract
The complex microenvironment that surrounds hematopoietic stem cells (HSCs) in the bone marrow niche involves different coordinated signaling pathways. The stem cells establish permanent interactions with distinct cell types such as mesenchymal stromal cells, osteoblasts, osteoclasts or endothelial cells and with secreted regulators such as growth factors, cytokines, chemokines and their receptors. These interactions are mediated through adhesion to extracellular matrix compounds also. All these signaling pathways are important for stem cell fates such as self-renewal, proliferation or differentiation, homing and mobilization, as well as for remodeling of the niche. Among these complex molecular cues, this review focuses on heparan sulfate (HS) structures and functions and on the role of enzymes involved in their biosynthesis and turnover. HS associated to core protein, constitute the superfamily of heparan sulfate proteoglycans (HSPGs) present on the cell surface and in the extracellular matrix of all tissues. The key regulatory effects of major medullar HSPGs are described, focusing on their roles in the interactions between hematopoietic stem cells and their endosteal niche, and on their ability to interact with Heparin Binding Proteins (HBPs). Finally, according to the relevance of HS moieties effects on this complex medullar niche, we describe recent data that identify HS mimetics or sulfated HS signatures as new glycanic tools and targets, respectively, for hematopoietic and mesenchymal stem cell based therapeutic applications.
Collapse
Affiliation(s)
- Dulce Papy-Garcia
- CRRET Laboratory, Université Paris Est, EA 4397 Université Paris Est Créteil, ERL CNRS 9215, F-94010, Créteil, France
| | - Patricia Albanese
- CRRET Laboratory, Université Paris Est, EA 4397 Université Paris Est Créteil, ERL CNRS 9215, F-94010, Créteil, France.
| |
Collapse
|
159
|
Gvaramia D, Müller E, Müller K, Atallah P, Tsurkan M, Freudenberg U, Bornhäuser M, Werner C. Combined influence of biophysical and biochemical cues on maintenance and proliferation of hematopoietic stem cells. Biomaterials 2017; 138:108-117. [PMID: 28554007 DOI: 10.1016/j.biomaterials.2017.05.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 12/17/2022]
Abstract
Homeostasis of hematopoietic stem and progenitor cells (HSPC) is controlled by a combination of biochemical and biophysical environmental cues in the bone marrow (BM) niche, where a tight balance of quiescence and proliferation of HSPC is maintained. Specifically, alongside soluble factors and extracellular matrix (ECM) proteins, spatial confinement and ECM stiffness have been recognized to be critical for regulation of HSPC fate. Here we employ a modular, glycosaminoglycan (GAG)-based biohybrid hydrogel system to balance proliferation of human HSPC and maintenance of quiescent hematopoietic stem cells (HSC) through simultaneous regulation of exogenous biochemical and biophysical cues. Our results demonstrate that HSPC respond to increased spatial confinement with lowered proliferation and cell cycling, which results in higher frequency of quiescent LTC-IC (long-term culture initiating cells), while GAG-rich 3D environments further support maintenance of the cells.
Collapse
Affiliation(s)
- David Gvaramia
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | - Eike Müller
- Laboratory for Biointerfaces, Empa, Swiss Federal Laboratories for Material Science and Technology, CH-9014, St. Gallen, Switzerland
| | - Katrin Müller
- Medical Clinic I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Passant Atallah
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | - Mikhail Tsurkan
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany; Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307, Dresden, Germany.
| |
Collapse
|
160
|
Park MH, Kim N, Jin HK, Bae JS. Neuropeptide Y-based recombinant peptides ameliorate bone loss in mice by regulating hematopoietic stem/progenitor cell mobilization. BMB Rep 2017; 50:138-143. [PMID: 27998395 PMCID: PMC5422026 DOI: 10.5483/bmbrep.2017.50.3.191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Indexed: 12/30/2022] Open
Abstract
Ovariectomy-induced bone loss is related to an increased deposition of osteoclasts on bone surfaces. We reported that the 36-amino-acid-long neuropeptide Y (NPY) could mobilize hematopoietic stem/progenitor cells (HSPCs) from the bone marrow to the peripheral blood by regulating HSPC maintenance factors and that mobilization of HSPCs ameliorated low bone density in an ovariectomy-induced osteoporosis mouse model by reducing the number of osteoclasts. Here, we demonstrated that new NPY peptides, recombined from the cleavage of the full-length NPY, showed better functionality for HSPC mobilization than the full-length peptide. These recombinant peptides mediated HSPC mobilization with greater efficiency by decreasing HSPC maintenance factors. Furthermore, treatment with these peptides reduced the number of osteoclasts and relieved ovariectomy-induced bone loss in mice more effectively than treatment with full-length NPY. Therefore, these results suggest that peptides recombined from full-length NPY can be used to treat osteoporosis.
Collapse
Affiliation(s)
- Min Hee Park
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Namoh Kim
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Hee Kyung Jin
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Jae-Sung Bae
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
161
|
An N, Chen Y, Yin D, Zhang HJ, Liu Z, Feng F, Li N, Xin J, Yin W, Xu X, Hu X. Melanoma-Induced Anemia Could be Rescued by Sca-1 + Mesenchymal Stromal Cells in Mice. Stem Cells Dev 2017; 26:495-502. [PMID: 28052733 DOI: 10.1089/scd.2016.0139] [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] [Indexed: 01/09/2023] Open
Abstract
The intrinsic basis of cancer-related anemia (CRA) is erythropoiesis disorder, which is a common complication of cancer and exerts a negative influence on the life quality of cancer patients. Cell therapy using mesenchymal stromal cells (MSCs) is considered as a promising method in cancer treatment. Furthermore, MSCs have been used to cure few type of anemia and be considered as a potential strategy to recover anemia radically. However, none reports its application in CRA treatment. In CRA model mice, we found that the number of lin-c-kit+Sca-1+ and Sca-1+ MSCs was decreased. And CRA resulted in an increased number of proerythroblasts and basophilic erythroblasts and decreased number of orthochromatic erythroblasts. Furthermore, in CRA model mice transplanted with Sca-1+ MSCs and MSCs, the levels of red blood cell count and Hb in peripheral blood were obviously increased. And the accumulation of proerythroblasts and basophilic erythroblasts was inhibited. In addition, the expression patterns of GATA-1 and GATA-2, which is pivotal to anemia, were remarkably recovered. Our results demonstrated that either MSCs or its subpopulation could effectively recover CRA erythropoiesis through GATA-1/GATA-2 signaling, which outstrips the traditional symptomatic therapy.
Collapse
Affiliation(s)
- Ning An
- 1 Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University , Xi'an, China
| | - Yaozhen Chen
- 1 Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University , Xi'an, China
| | - Dandan Yin
- 2 Department of Hematology, Tangdu Hospital, The Fourth Military Medical University , Xi'an, China
| | - Hui-Jie Zhang
- 1 Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University , Xi'an, China
| | - Zheng Liu
- 3 Department of Biochemistry and Molecular Biophysics, Columbia University , New York, New York
| | - Fan Feng
- 4 Department of Digestive Surgery, Xijing Hospital, The Fourth Military Medical University , Xi'an, China
| | - Na Li
- 1 Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University , Xi'an, China
| | - Jiajia Xin
- 1 Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University , Xi'an, China
| | - Wen Yin
- 1 Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University , Xi'an, China
| | - Xueqing Xu
- 5 Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University , Chongqing, China
| | - Xingbin Hu
- 1 Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University , Xi'an, China
| |
Collapse
|
162
|
Hira VVV, Van Noorden CJF, Carraway HE, Maciejewski JP, Molenaar RJ. Novel therapeutic strategies to target leukemic cells that hijack compartmentalized continuous hematopoietic stem cell niches. Biochim Biophys Acta Rev Cancer 2017; 1868:183-198. [PMID: 28363872 DOI: 10.1016/j.bbcan.2017.03.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Acute myeloid leukemia and acute lymphoblastic leukemia cells hijack hematopoietic stem cell (HSC) niches in the bone marrow and become leukemic stem cells (LSCs) at the expense of normal HSCs. LSCs are quiescent and resistant to chemotherapy and can cause relapse of the disease. HSCs in niches are needed to generate blood cell precursors that are committed to unilineage differentiation and eventually production of mature blood cells, including red blood cells, megakaryocytes, myeloid cells and lymphocytes. Thus far, three types of HSC niches are recognized: endosteal, reticular and perivascular niches. However, we argue here that there is only one type of HSC niche, which consists of a periarteriolar compartment and a perisinusoidal compartment. In the periarteriolar compartment, hypoxia and low levels of reactive oxygen species preserve the HSC pool. In the perisinusoidal compartment, hypoxia in combination with higher levels of reactive oxygen species enables proliferation of progenitor cells and their mobilization into the circulation. Because HSC niches offer protection to LSCs against chemotherapy, we review novel therapeutic strategies to inhibit homing of LSCs in niches for the prevention of dedifferentiation of leukemic cells into LSCs and to stimulate migration of leukemic cells out of niches. These strategies enhance differentiation and proliferation and thus sensitize leukemic cells to chemotherapy. Finally, we list clinical trials of therapies that tackle LSCs in HSC niches to circumvent their protection against chemotherapy.
Collapse
Affiliation(s)
- Vashendriya V V Hira
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA.
| | - Cornelis J F Van Noorden
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| | - Hetty E Carraway
- Department of Translational Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA; Leukemia Program, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| | - Remco J Molenaar
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; Department of Translational Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|
163
|
In vivo longitudinal visualization of bone marrow engraftment process in mouse calvaria using two-photon microscopy. Sci Rep 2017; 7:44097. [PMID: 28276477 PMCID: PMC5343427 DOI: 10.1038/srep44097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/31/2017] [Indexed: 12/13/2022] Open
Abstract
Intravital microscopy of mouse calvarial bone marrow (BM) is a powerful method for studying hematopoietic stem cells (HSCs) and the BM microenvironment at the cellular level. However, the current method used to access the mouse calvaria allows for only a few imaging times in the same mouse because of scar formation and inflammation induced by multiple surgeries. Longitudinal imaging of the BM may help better understand its microenvironment. In this study, a mouse calvarial window model was developed for longitudinal imaging that involves attaching a cover glass window onto the mouse calvaria and sealing the surrounding exposed area with cyanoacrylate glue and dental cement. The model was used for the longitudinal two-photon microscopy (TPM) imaging of the BM engraftment process. The same BM cavity sites were imaged multiple times over 4 weeks after BM transplantation (BMT). Temporal changes in the BM microenvironment, such as the reconstitution of transplanted BM cells and the recovery of vasculature, were observed and analysed qualitatively and quantitatively. Longitudinal intravital microscopy using the mouse calvarial window model was successfully demonstrated and may be useful for further BM studies.
Collapse
|
164
|
Liang SM, Lu YJ, Ko BS, Jan YJ, Shyue SK, Yet SF, Liou JY. Cordycepin disrupts leukemia association with mesenchymal stromal cells and eliminates leukemia stem cell activity. Sci Rep 2017; 7:43930. [PMID: 28266575 PMCID: PMC5339716 DOI: 10.1038/srep43930] [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: 08/09/2016] [Accepted: 01/31/2017] [Indexed: 12/19/2022] Open
Abstract
Maintaining stemness of leukemic stem cells (LSCs) and reciprocal interactions between leukemia and stromal cells support leukemic progression and resistance to chemotherapy. Targeting the niche-based microenvironment is thus a new approach for leukemia therapy. Cordycepin is an analogue of adenosine and has been suggested to possess anti-leukemia properties. However, whether cordycepin influences association of leukemia and mesenchymal stromal cells has never been investigated. Here we show that cordycepin reduces CD34+CD38− cells in U937 and K562 cells and induces Dkk1 expression via autocrine and paracrine regulation in leukemia and mesenchymal stromal/stem cells (MSCs). Cordycepin suppresses cell attachment of leukemia with MSCs and downregulates N-cadherin in leukemia and VCAM-1 in MSCs. Moreover, incubation with leukemic conditioned media (CM) significantly induces IL-8 and IL-6 expression in MSCs, which is abrogated by cordycepin. Suppression of leukemic CM-induced VCAM-1 and IL-8 by cordycepin in MSCs is mediated by impairing NFκB signaling. Finally, cordycepin combined with an adenosine deaminase inhibitor prolongs survival in a leukemic mouse model. Our results indicate that cordycepin is a potential anti-leukemia therapeutic adjuvant via eliminating LSCs and disrupting leukemia-stromal association.
Collapse
Affiliation(s)
- Shu-Man Liang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350, Taiwan
| | - Yi-Jhu Lu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350, Taiwan
| | - Bor-Sheng Ko
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Yee-Jee Jan
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350, Taiwan
| | - Jun-Yang Liou
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350, Taiwan.,Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
165
|
Mohammadi S, Nikbakht M, Sajjadi SM, Rad F, Chahardouli B, Sabour Takanlu J, Rostami S, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Reciprocal Interactions of Leukemic Cells with Bone Marrow Stromal Cells Promote Enrichment of Leukemic Stem Cell Compartments in Response to Curcumin and Daunorubicin. Asian Pac J Cancer Prev 2017; 18:831-840. [PMID: 28441794 PMCID: PMC5464507 DOI: 10.22034/apjcp.2017.18.3.831] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
A predominant challenge in developing curative leukemia therapy is interactions of leukemic cells with the bone marrow stromal microenvironment. We aimed to investigate the role of stromal cells, such as bone marrow mesenchymal stromal cells (BMSCs) and osteoblasts (OBs), in curcumin (CUR) and daunorubicin (DNR) induced apoptosis of acute myeloid leukemia (AML) cells. We used KG1 and U937 as leukemia cell line models and treated them with CUR and DNR. The cells were then co-cultured with BMSCs or a combination of BMSCs and OBs as feeders. After 24 hours of co-culture, BMSCs or OBs were sorted and separated from the leukemia cells and apoptosis levels were analyzed by annexin/propidium iodide (PI) staining on flow cytometry. Potentially involved molecular pathways were analyzed at gene and protein levels by Real time PCR and western blotting, respectively. The results showed AML cells co-cultured with BMSCs plus OBs to be more resistant to drug induced-apoptosis compared to co-culture with BMSCs alone or without co-culture. Expression levels of OPN, CXCL-12, IL-6, STAT-3 and VCAM-1 were also significantly up-regulated in OBs and AML cells, at both mRNA and protein levels after co-culture, with concurrent enrichment of CD34+ AML cells. Our data showed, in a stromal cell niche-based model, that OBs revoke the influence of BMSCs on leukemic cells and promote enrichment of both CD34+ and CD34- leukemic stem cell (LSC) compartments in response to CUR and DNR. Up-regulation of OPN, CXCL-12, IL-6, STAT-3 and VCAM-1 in OBs and AML cells in co-culture might be part of molecular mechanisms that block CUR or CUR+DNR-induced apoptosis and promote enrichment of CD34+ and CD34- LSCs.
Collapse
Affiliation(s)
- Saeed Mohammadi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Perucca S, Di Palma A, Piccaluga PP, Gemelli C, Zoratti E, Bassi G, Giacopuzzi E, Lojacono A, Borsani G, Tagliafico E, Scupoli MT, Bernardi S, Zanaglio C, Cattina F, Cancelli V, Malagola M, Krampera M, Marini M, Almici C, Ferrari S, Russo D. Mesenchymal stromal cells (MSCs) induce ex vivo proliferation and erythroid commitment of cord blood haematopoietic stem cells (CB-CD34+ cells). PLoS One 2017; 12:e0172430. [PMID: 28231331 PMCID: PMC5322933 DOI: 10.1371/journal.pone.0172430] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/03/2017] [Indexed: 12/30/2022] Open
Abstract
A human bone marrow-derived mesenchymal stromal cell (MSCs) and cord blood-derived CD34+ stem cell co-culture system was set up in order to evaluate the proliferative and differentiative effects induced by MSCs on CD34+ stem cells, and the reciprocal influences on gene expression profiles. After 10 days of co-culture, non-adherent (SN-fraction) and adherent (AD-fraction) CD34+ stem cells were collected and analysed separately. In the presence of MSCs, a significant increase in CD34+ cell number was observed (fold increase = 14.68), mostly in the SN-fraction (fold increase = 13.20). This was combined with a significant increase in CD34+ cell differentiation towards the BFU-E colonies and with a decrease in the CFU-GM. These observations were confirmed by microarray analysis. Through gene set enrichment analysis (GSEA), we noted a significant enrichment in genes involved in heme metabolism (e.g. LAMP2, CLCN3, BMP2K), mitotic spindle formation and proliferation (e.g. PALLD, SOS1, CCNA1) and TGF-beta signalling (e.g. ID1) and a down-modulation of genes participating in myeloid and lymphoid differentiation (e.g. PCGF2) in the co-cultured CD34+ stem cells. On the other hand, a significant enrichment in genes involved in oxygen-level response (e.g. TNFAIP3, SLC2A3, KLF6) and angiogenesis (e.g. VEGFA, IGF1, ID1) was found in the co-cultured MSCs. Taken together, our results suggest that MSCs can exert a priming effect on CD34+ stem cells, regulating their proliferation and erythroid differentiation. In turn, CD34+ stem cells seem to be able to polarise the BM-niche towards the vascular compartment by modulating molecular pathways related to hypoxia and angiogenesis.
Collapse
Affiliation(s)
- Simone Perucca
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Laboratorio CREA (Centro di Ricerca Emato-oncologica AIL), ASST Spedali Civili of Brescia, Brescia, Italy
| | - Andrea Di Palma
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Laboratorio CREA (Centro di Ricerca Emato-oncologica AIL), ASST Spedali Civili of Brescia, Brescia, Italy
| | - Pier Paolo Piccaluga
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), S. Orsola-Malpighi Hospital, Bologna University School of Medicine, Bologna, Italy
- Section of Genomics and Personalized Medicine, Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Claudia Gemelli
- Parco Scientifico e Tecnologico Materiali Innovativi e Ricerca Applicata del Mirandolese, Modena, Italy
| | - Elisa Zoratti
- Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Giulio Bassi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Edoardo Giacopuzzi
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine (DMTM), University of Brescia, Brescia, Italy
| | - Andrea Lojacono
- U.O. of Obstetrics and Gynecology I, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Giuseppe Borsani
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine (DMTM), University of Brescia, Brescia, Italy
| | - Enrico Tagliafico
- Centro di Ricerche Genomiche, Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Maria Teresa Scupoli
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy
| | - Simona Bernardi
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Laboratorio CREA (Centro di Ricerca Emato-oncologica AIL), ASST Spedali Civili of Brescia, Brescia, Italy
| | - Camilla Zanaglio
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- Laboratorio CREA (Centro di Ricerca Emato-oncologica AIL), ASST Spedali Civili of Brescia, Brescia, Italy
| | - Federica Cattina
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Valeria Cancelli
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Michele Malagola
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mirella Marini
- Department of Transfusion Medicine, Laboratory for Stem Cells Manipulation and Cryopreservation, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Camillo Almici
- Department of Transfusion Medicine, Laboratory for Stem Cells Manipulation and Cryopreservation, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Sergio Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
- * E-mail:
| |
Collapse
|
167
|
Agas D, Gusmão Silva G, Laus F, Marchegiani A, Capitani M, Vullo C, Catone G, Lacava G, Concetti A, Marchetti L, Sabbieti MG. INF-γ encoding plasmid administration triggers bone loss and disrupts bone marrow microenvironment. J Endocrinol 2017; 232:309-321. [PMID: 27908965 DOI: 10.1530/joe-16-0538] [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: 11/20/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
IFN-γ is a pleotropic cytokine produced in the bone microenvironment. Although IFN-γ is known to play a critical role on bone remodeling, its function is not fully elucidated. Consistently, outcomes on the effects of IFN-γ recombinant protein on bone loss are contradictory among reports. In our work we explored, for the first time, the role of IFN-γ encoding plasmid (pIFN-γ) in a mouse model of osteopenia induced by ovariectomy and in the sham-operated counterpart to estimate its effects in skeletal homeostasis. Ovariectomy produced a dramatic decrease of bone mineral density (BMD). pINF-γ injected mice showed a pathologic bone and bone marrow phenotype; the disrupted cortical and trabecular bone microarchitecture was accompanied by an increased release of pro-inflammatory cytokine by bone marrow cells. Moreover, mesenchymal stem cells' (MSCs) commitment to osteoblast was found impaired, as evidenced by the decline of osterix-positive (Osx+) cells within the mid-diaphyseal area of femurs. For instance, a reduction and redistribution of CXCL12 cells have been found, in accordance with bone marrow morphological alterations. As similar effects were observed both in sham-operated and in ovariectomized mice, our studies proved that an increased IFN-γ synthesis in bone marrow might be sufficient to induce inflammatory and catabolic responses even in the absence of pathologic predisposing substrates. In addition, the obtained data might raise questions about pIFN-γ's safety when it is used as vaccine adjuvant.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | - Guilherme Gusmão Silva
- Departamento de Bioquímica e ImunologiaUniversidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fulvio Laus
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | - Andrea Marchegiani
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | - Melania Capitani
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | - Cecilia Vullo
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | - Giuseppe Catone
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | - Giovanna Lacava
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | - Antonio Concetti
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | - Luigi Marchetti
- School of Bioscience and Veterinary MedicineUniversity of Camerino, Camerino, Italy
| | | |
Collapse
|
168
|
Numerous niches for hematopoietic stem cells remain empty during homeostasis. Blood 2017; 129:2124-2131. [PMID: 28130213 DOI: 10.1182/blood-2016-09-740563] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/18/2017] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cells (HSCs) reside in and are maintained by special microenvironments, termed niches. It is assumed that the HSC niche space remains occupied by endogenous cells and that myelosuppressive conditioning is required to achieve high levels of HSC engraftment. We herein demonstrate that upon the transplantation of very large numbers of purified HSCs into normal mice not exposed to myeloablation, donor HSCs engrafted in niches distant from filled HSC niches without replacing host HSCs and subsequently proliferated and generated hematopoietic progenitors, leading to marked increases in the overall HSC numbers in bone marrow. Additionally, stem cell factor that is produced by CXC chemokine ligand 12-abundant reticular cells is involved in HSC engraftment. In contrast, host granulocyte/macrophage progenitors (GMPs) were replaced by the progeny of transplanted donor HSCs, and overall GMP numbers remained unchanged. Thus, inconsistent with the classical concept, numerous empty HSC niches are available for engraftment and proliferation in bone marrow.
Collapse
|
169
|
De Luca L, Trino S, Laurenzana I, Simeon V, Calice G, Raimondo S, Podestà M, Santodirocco M, Di Mauro L, La Rocca F, Caivano A, Morano A, Frassoni F, Cilloni D, Del Vecchio L, Musto P. MiRNAs and piRNAs from bone marrow mesenchymal stem cell extracellular vesicles induce cell survival and inhibit cell differentiation of cord blood hematopoietic stem cells: a new insight in transplantation. Oncotarget 2017; 7:6676-92. [PMID: 26760763 PMCID: PMC4872742 DOI: 10.18632/oncotarget.6791] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/05/2015] [Indexed: 12/18/2022] Open
Abstract
Hematopoietic stem cells (HSC), including umbilical cord blood CD34+ stem cells (UCB-CD34+), are used for the treatment of several diseases. Although different studies suggest that bone marrow mesenchymal stem cells (BM-MSC) support hematopoiesis, the exact mechanism remains unclear. Recently, extracellular vesicles (EVs) have been described as a novel avenue of cell communication, which may mediate BM-MSC effect on HSC. In this work, we studied the interaction between UCB-CD34+ cells and BM-MSC derived EVs. First, by sequencing EV derived miRNAs and piRNAs we found that EVs contain RNAs able to influence UCB-CD34+ cell fate. Accordingly, a gene expression profile of UCB-CD34+ cells treated with EVs, identified about 100 down-regulated genes among those targeted by EV-derived miRNAs and piRNAs (e.g. miR-27b/MPL, miR-21/ANXA1, miR-181/EGR2), indicating that EV content was able to modify gene expression profile of receiving cells. Moreover, we demonstrated that UCB-CD34+ cells, exposed to EVs, significantly changed different biological functions, becoming more viable and less differentiated. UCB-CD34+ gene expression profile also identified 103 up-regulated genes, most of them codifying for chemokines, cytokines and their receptors, involved in chemotaxis of different BM cells, an essential function of hematopoietic reconstitution. Finally, the exposure of UCB-CD34+ cells to EVs caused an increased expression CXCR4, paralleled by an in vivo augmented migration from peripheral blood to BM niche in NSG mice. This study demonstrates the existence of a powerful cross talk between BM-MSC and UCB-CD34+ cells, mediated by EVs, providing new insight in the biology of cord blood transplantation.
Collapse
Affiliation(s)
- Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| | - Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| | - Vittorio Simeon
- Laboratory of Preclinical and Translational Research, IRCCS-Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| | - Giovanni Calice
- Laboratory of Preclinical and Translational Research, IRCCS-Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, University of Turin, Turin 10126, Italy
| | - Marina Podestà
- Stem Cell Center, S. Martino Hospital, Genova 16132, Italy
| | - Michele Santodirocco
- Transfusion Medicine Unit, Puglia Cord Blood Bank, IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 (FG), Italy
| | - Lazzaro Di Mauro
- Transfusion Medicine Unit, Puglia Cord Blood Bank, IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 (FG), Italy
| | - Francesco La Rocca
- Laboratory of Preclinical and Translational Research, IRCCS-Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| | - Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| | - Annalisa Morano
- Laboratory of Preclinical and Translational Research, IRCCS-Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| | - Francesco Frassoni
- Laboratorio Cellule Staminali Post Natali e Terapie Cellulari, Giannina Gaslini Institute, Genova 16148, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Turin 10126, Italy
| | - Luigi Del Vecchio
- CEINGE-Biotecnologie Avanzate S.C.a R.L., Naples, 80145, Italy.,Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples 80131, Italy
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Centro di Riferimento Oncologico Basilicata (CROB), Rionero in Vulture, 85028 (PZ), Italy
| |
Collapse
|
170
|
SANTRA LAKSHMAN, GUPTA SAURABH, KANNAN SANGEETHA, SINGH AJAYK, KUMAR GVPPSRAVI, NASKAR SOUMEN, GHOSH JYOTIRMOY, DHARA SUJOYK. Long bones, a slaughterhouse by-product, may serve as an excellent source for mesenchymal stem cells. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2017. [DOI: 10.56093/ijans.v87i1.66860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Mesenchymal stem cells (MSCs) are one of the rarest sub-populations of bone marrow resident cells having inherent ability to differentiate into mesenchyme tissues e.g. bone, cartilage and adipose tissues. The natural selfrenewal ability and potential for lineage specific differentiation have made these cells an excellent material for research and therapy in regenerative medicine. But, successful isolation and in vitro expansion of these cells still remain the pivotal steps for majority of stem cell based applications. Various techniques have been successfully used for isolation of MSCs from laboratory animals, but those are difficult to apply for domestic species. Hence, harvesting MSCs from most domestic animals remains a real challenge. Here we have demonstrated an easy, convenient, low cost method of MSCs isolation from slaughtered animals. As a proof of concept, MSCs were isolated from bone marrow of 3 different species, namely, sheep, pig and goat. These cells expressed multiple markers and also retained their self-renewal potential, exhibited by successful sub-culturing over 30 passages. Moreover, MSCs expressed many pluripotency factors e.g. OCT4, Nanog, c-Myc, KLF2 and KLF4. This indicated that the bone marrow derived MSCs were at very early stage of commitment and therefore, possibly retained high plasticity. Since these cells are available from slaughtered animals, this circumvents the bioethical issues associated with invasive method of MSC isolation from bone marrow. This invaluable and easily adoptable method for isolation of MSCs from large domestic animal would encourage isolation process in other animals and help in future cell based researches and therapies in the field of regenerative medicine.
Collapse
|
171
|
Vanegas NDP, Vernot JP. Loss of quiescence and self-renewal capacity of hematopoietic stem cell in an in vitro leukemic niche. Exp Hematol Oncol 2017; 6:2. [PMID: 28078190 PMCID: PMC5223333 DOI: 10.1186/s40164-016-0062-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 12/29/2016] [Indexed: 02/06/2023] Open
Abstract
Background Leukemic and mesenchymal stem cells interact in the leukemic microenvironment and affect each other differently. This interplay has also important implications for the hematopoietic stem cell (HSC) biology and function. This study evaluated human HSC self-renewal potential and quiescence in an in vitro leukemic niche without leukemic cells. Methods A leukemic niche was established by co-culturing mesenchymal stem cells with a fresh conditioned medium obtained from a leukemic (REH) cell line. After 3 days, the REH-conditioned medium was removed and freshly isolated CD34+ at a density of up to 100,000 cells/ml were added to the leukemic niche. CD34+ cell evaluations (cell cycle, self-renewal gene expression and migration capacity) were performed after 3 further days of co-culture. Additionally, we preliminary investigated the soluble factors present in the leukemic niche and their effect on the mesenchymal stem cells. Statistical significance was assessed by Student’s t test or the nonparametric test Kolmogorov–Smirnov. Results By co-culturing normal mesenchymal stem cells with the REH-conditioned medium we showed that hematopoietic stem cells, normally in a quiescent state, enter cell cycle and proliferate. This loss of quiescence was accompanied by an increased expression of Ki-67 and c-Myc, two well-known cell proliferation-associated markers. Two central regulators of quiescence GATA2 and p53 were also down regulated. Importantly, two genes involved in HSC self-renewal, Klf4 and the histone–lysine N-methyltransferase enzyme Ezh2, were severely affected. On the contrary, c-Kit expression, the stem cell factor receptor, was upregulated in hematopoietic stem cells when compared to the normal niche. Interestingly, mesenchymal stem cells incubated with the REH-conditioned medium stopped growing, showed a flattened morphology with the appearance of small vacuoles, and importantly, became positive for the senescence-associated beta-galactosidase activity. Evaluation of the leukemic-conditioned medium showed increased IL-6 and IL-8, suggesting that these cytokines could be responsible for the observed changes. Conclusions Our results showed that quiescence and self-renewal are severely affected in this leukemic niche. This in vitro leukemic niche, established without leukemic cells, will facilitate HSC gene expression evaluation and the development of therapeutic agents aimed to neutralize soluble factors and the cell signaling pathways involved in HSC alterations. Electronic supplementary material The online version of this article (doi:10.1186/s40164-016-0062-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Natalia-Del Pilar Vanegas
- Cellular and Molecular Physiology, Biomedical Research Institute, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, D.C. 111321 Colombia
| | - Jean-Paul Vernot
- Cellular and Molecular Physiology, Biomedical Research Institute, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, D.C. 111321 Colombia
| |
Collapse
|
172
|
Characterization of peritoneal leukemia-associated macrophages in Notch1-induced mouse T cell acute lymphoblastic leukemia. Mol Immunol 2017; 81:35-41. [DOI: 10.1016/j.molimm.2016.11.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 01/05/2023]
|
173
|
Choi JS, Harley BAC. Marrow-inspired matrix cues rapidly affect early fate decisions of hematopoietic stem and progenitor cells. SCIENCE ADVANCES 2017; 3:e1600455. [PMID: 28070554 PMCID: PMC5218514 DOI: 10.1126/sciadv.1600455] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 11/22/2016] [Indexed: 05/02/2023]
Abstract
Hematopoiesis is the physiological process where hematopoietic stem cells (HSCs) continuously generate the body's complement of blood and immune cells within unique regions of the bone marrow termed niches. Although previous investigations have revealed gradients in cellular and extracellular matrix (ECM) content across the marrow, and matrix elasticity and ligand type are believed to be strong regulators of stem cell fate, the impact of biophysical signals on HSC response is poorly understood. Using marrow-inspired ECM ligand-coated polyacrylamide substrates that present defined stiffness and matrix ligand cues, we demonstrate that the interplay between integrin engagement and myosin II activation processes affects the morphology, proliferation, and myeloid lineage specification of primary murine HSCs within 24 hours ex vivo. Notably, the impact of discrete biophysical signals on HSC fate decisions appears to be correlated to known microenvironmental transitions across the marrow. The combination of fibronectin and marrow matrix-associated stiffness was sufficient to maintain hematopoietic progenitor populations, whereas collagen and laminin enhanced proliferation and myeloid differentiation, respectively. Inhibiting myosin II-mediated contraction or adhesion to fibronectin via specific integrins (α5β1 and ανβ3) selectively abrogated the impact of the matrix environment on HSC fate decisions. Together, these findings indicate that adhesive interactions and matrix biophysical properties are critical design considerations in the development of biomaterials to direct HSC behavior in vitro.
Collapse
Affiliation(s)
- Ji Sun Choi
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan A. C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Corresponding author.
| |
Collapse
|
174
|
Tvedt TH, Nepstad I, Bruserud Ø. Antileukemic effects of midostaurin in acute myeloid leukemia - the possible importance of multikinase inhibition in leukemic as well as nonleukemic stromal cells. Expert Opin Investig Drugs 2016; 26:343-355. [PMID: 28001095 DOI: 10.1080/13543784.2017.1275564] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Midostaurin is a multikinase inhibitor that inhibits receptor tyrosine kinases (Flt3, CD117/c-kit, platelet-derived growth factor receptor, vascular endothelial growth factor receptor 2) as well as non-receptor tyrosine kinases (Frg, Src, Syk, Protein kinase C). Combination of midostaurin with conventional intensive chemotherapy followed by one year maintenance monotherapy was recently reported to improve the survival of acute myeloid leukemia (AML) patients with Flt3 mutations. Areas covered: Relevant publications were identified through literature searches in the PubMed database. We searched for (i) original articles describing the results from clinical studies; (ii) published articles describing the importance of midostaurin-inhibited kinases for leukemogenesis and chemosensitivity. Expert opinion: Midostaurin monotherapy is well tolerated, combined with conventional chemotherapy gastrointestinal toxicity increases significantly. Midostaurin alters anthracycline pharmacokinetics. Furthermore, its antileukemic effects may not only be mediated through Flt3 inhibition alone; the inhibition of other kinases may also be important for the overall antileukemic effect. Midostaurin may then have direct effects on the leukemic cells but also indirect antileukemic effects through inhibition of the AML-supporting effects of neighboring stromal cells in the bone marrow microenvironment. Midostaurin may thus be used in combination with intensive chemotherapy, as maintenance treatment or as disease-stabilizing treatment for elderly unfit patients.
Collapse
Affiliation(s)
- Tor Henrik Tvedt
- a Section for Hematology, Department of Medicine , Haukeland University Hospital , Bergen , Norway
| | - Ina Nepstad
- b Section for Hematology , Institute of Clinical Science, University of Bergen , Bergen , Norway
| | - Øystein Bruserud
- a Section for Hematology, Department of Medicine , Haukeland University Hospital , Bergen , Norway.,b Section for Hematology , Institute of Clinical Science, University of Bergen , Bergen , Norway
| |
Collapse
|
175
|
Cathepsin K cleavage of SDF-1α inhibits its chemotactic activity towards glioblastoma stem-like cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:594-603. [PMID: 28040478 DOI: 10.1016/j.bbamcr.2016.12.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/22/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor with poor patient survival that is at least partly caused by malignant and therapy-resistant glioma stem-like cells (GSLCs) that are protected in GSLC niches. Previously, we have shown that the chemo-attractant stromal-derived factor-1α (SDF-1α), its C-X-C receptor type 4 (CXCR4) and the cysteine protease cathepsin K (CatK) are localized in GSLC niches in glioblastoma. Here, we investigated whether SDF-1α is a niche factor that through its interactions with CXCR4 and/or its second receptor CXCR7 on GSLCs facilitates their homing to niches. Furthermore, we aimed to prove that SDF-1α cleavage by CatK inactivates SDF-1α and inhibits the invasion of GSLCs. We performed mass spectrometric analysis of cleavage products of SDF-1α after proteolysis by CatK. We demonstrated that CatK cleaves SDF-1α at 3 sites in the N-terminus, which is the region of SDF-1α that binds to its receptors. Confocal imaging of human GBM tissue sections confirmed co-localization of SDF-1α and CatK in GSLC niches. In accordance, 2D and 3D invasion experiments using CXCR4/CXCR7-expressing GSLCs and GBM cells showed that SDF-1α had chemotactic activity whereas CatK cleavage products of SDF-1α did not. Besides, CXCR4 inhibitor plerixafor inhibited invasion of CXCR4/CXCR7-expressing GSLCs. In conclusion, CatK can cleave and inactivate SDF-1α. This implies that CatK activity facilitates migration of GSLCs out of niches. We propose that activation of CatK may be a promising strategy to prevent homing of GSLCs in niches and thus render these cells sensitive to chemotherapy and radiation.
Collapse
|
176
|
Digesting the role of bone marrow macrophages on hematopoiesis. Immunobiology 2016; 222:814-822. [PMID: 27890297 DOI: 10.1016/j.imbio.2016.11.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/27/2016] [Accepted: 11/12/2016] [Indexed: 01/07/2023]
Abstract
Tissue resident macrophages are found in various tissues like Langerhans cells in the skin or alveolar macrophages in the lung, and their main function is to regulate organ homeostasis. They have also been observed in the bone marrow and these cells in particular have been gaining importance in recent years as they are key players in hematopoiesis. However, as the characterization and classification of these putatively different bone marrow resident macrophages is far from established there is a need to generate an overview of tissue resident macrophages of the bone marrow. Here, we will review the current knowledge of bone marrow resident macrophages both in mouse and human. We will discuss the state of the art on the origin of bone marrow macrophages, specialized microenvironments where they reside and their unique characteristics. We will emphasize the two best studied examples of macrophage homeostatic function in the bone marrow, specifically within erythroblastic islands and the hematopoietic stem cell niche. Although increasing evidence shows that bone marrow resident macrophages are indispensable for hematopoietic stem cell function and bone marrow erythroid output, the field of bone marrow macrophages is in its infancy. This field is in dire need for a unified nomenclature to support functional experiments, model systems, and the identification of niches.
Collapse
|
177
|
Wharton’s Jelly Mesenchymal Stromal Cells as a Feeder Layer for the Ex Vivo Expansion of Hematopoietic Stem and Progenitor Cells: a Review. Stem Cell Rev Rep 2016; 13:35-49. [DOI: 10.1007/s12015-016-9702-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
178
|
Brenner AK, Andersson Tvedt TH, Bruserud Ø. The Complexity of Targeting PI3K-Akt-mTOR Signalling in Human Acute Myeloid Leukaemia: The Importance of Leukemic Cell Heterogeneity, Neighbouring Mesenchymal Stem Cells and Immunocompetent Cells. Molecules 2016; 21:molecules21111512. [PMID: 27845732 PMCID: PMC6273124 DOI: 10.3390/molecules21111512] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 12/11/2022] Open
Abstract
Therapeutic targeting of PI3K-Akt-mTOR is considered a possible strategy in human acute myeloid leukaemia (AML); the most important rationale being the proapoptotic and antiproliferative effects of direct PI3K/mTOR inhibition observed in experimental studies of human AML cells. However, AML is a heterogeneous disease and these effects caused by direct pathway inhibition in the leukemic cells are observed only for a subset of patients. Furthermore, the final effect of PI3K-Akt-mTOR inhibition is modulated by indirect effects, i.e., treatment effects on AML-supporting non-leukemic bone marrow cells. In this article we focus on the effects of this treatment on mesenchymal stem cells (MSCs) and monocytes/macrophages; both these cell types are parts of the haematopoietic stem cell niches in the bone marrow. MSCs have unique membrane molecule and constitutive cytokine release profiles, and mediate their support through bidirectional crosstalk involving both cell-cell contact and the local cytokine network. It is not known how various forms of PI3K-Akt-mTOR targeting alter the molecular mechanisms of this crosstalk. The effect on monocytes/macrophages is also difficult to predict and depends on the targeted molecule. Thus, further development of PI3K-Akt-mTOR targeting into a clinical strategy requires detailed molecular studies in well-characterized experimental models combined with careful clinical studies, to identify patient subsets that are likely to respond to this treatment.
Collapse
Affiliation(s)
- Annette K Brenner
- Section for Haematology, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
| | - Tor Henrik Andersson Tvedt
- Section for Haematology, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway.
| | - Øystein Bruserud
- Section for Haematology, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway.
| |
Collapse
|
179
|
MacLean AL, Lo Celso C, Stumpf MP. Concise Review: Stem Cell Population Biology: Insights from Hematopoiesis. Stem Cells 2016; 35:80-88. [DOI: 10.1002/stem.2508] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/19/2016] [Accepted: 08/21/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Adam L. MacLean
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| | - Cristina Lo Celso
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| | - Michael P.H. Stumpf
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| |
Collapse
|
180
|
Microengineered platforms for co-cultured mesenchymal stem cells towards vascularized bone tissue engineering. Tissue Eng Regen Med 2016; 13:465-474. [PMID: 30603428 DOI: 10.1007/s13770-016-9080-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/08/2015] [Accepted: 12/18/2015] [Indexed: 12/11/2022] Open
Abstract
Bone defects are common disease requiring thorough treatments since the bone is a complex vascularized tissue that is composed of multiple cell types embedded within an intricate extracellular matrix (ECM). For past decades, tissue engineering using cells, proteins, and scaffolds has been suggested as one of the promising approaches for effective bone regeneration. Recently, many researchers have been interested in designing effective platform for tissue regeneration by orchestrating factors involved in microenvironment around tissues. Among factors affecting bone formation, vascularization during bone development and after minor insults via endochondral and intramembranous ossification is especially critical for the long-term support for functional bone. In order to create vascularized bone constructs, the interactions between human mesenchymal stem cells (MSCs) and endothelial cells (ECs) have been investigated using both direct and indirect co-culture studies. Recently, various culture methods including micropatterning techniques, three dimensional scaffolds, and microfluidics have been developed to create micro-engineered platforms that mimic the nature of vascularized bone formation, leading to the creation of functional bone structures. This review focuses on MSCs co-cultured with endothelial cells and microengineered platforms to determine the underlying interplay between co-cultured MSCs and vascularized bone constructs, which is ultimately necessary for adequate regeneration of bone defects.
Collapse
|
181
|
Hu X, Garcia M, Weng L, Jung X, Murakami JL, Kumar B, Warden CD, Todorov I, Chen CC. Identification of a common mesenchymal stromal progenitor for the adult haematopoietic niche. Nat Commun 2016; 7:13095. [PMID: 27721421 PMCID: PMC5062560 DOI: 10.1038/ncomms13095] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/01/2016] [Indexed: 12/13/2022] Open
Abstract
Microenvironment cues received by haematopoietic stem cells (HSC) are important in regulating the choice between self-renewal and differentiation. On the basis of the differential expression of cell-surface markers, here we identify a mesenchymal stromal progenitor hierarchy, where CD45−Ter119−CD31−CD166−CD146−Sca1+(Sca1+) progenitors give rise to CD45−Ter119−CD31−CD166−CD146+(CD146+) intermediate and CD45−Ter119−CD31−CD166+CD146−(CD166+) mature osteo-progenitors. All three progenitors preserve HSC long-term multi-lineage reconstitution capability in vitro; however, their in vivo fates are different. Post-transplantation, CD146+ and CD166+ progenitors form bone only. While Sca1+ progenitors produce CD146+, CD166+ progenitors, osteocytes and CXCL12-producing stromal cells. Only Sca1+ progenitors are capable of homing back to the marrow post-intravenous infusion. Ablation of Sca1+ progenitors results in a decrease of all three progenitor populations as well as haematopoietic stem/progenitor cells. Moreover, suppressing production of KIT-ligand in Sca1+ progenitors inhibits their ability to support HSCs. Our results indicate that Sca1+ progenitors, through the generation of both osteogenic and stromal cells, provide a supportive environment for hematopoiesis. How the environment of the niche regulates haematopoietic stem cells (HSC) is unclear. Here, the authors identify a mesenchymal stromal progenitor hierarchy and identify Sca1+ cells as common progenitors for mesenchymal stromal cells in the adult niche that provide a supportive environment for hematopoiesis.
Collapse
Affiliation(s)
- Xingbin Hu
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA.,Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 7100032, PR China
| | - Mayra Garcia
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Lihong Weng
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Xiaoman Jung
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Jodi L Murakami
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA.,Irell &Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010, USA
| | - Bijender Kumar
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Charles D Warden
- Bioinformatics Core, Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Ivan Todorov
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Ching-Cheng Chen
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA.,Irell &Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010, USA
| |
Collapse
|
182
|
Kan C, Vargas G, Pape FL, Clézardin P. Cancer Cell Colonisation in the Bone Microenvironment. Int J Mol Sci 2016; 17:ijms17101674. [PMID: 27782035 PMCID: PMC5085707 DOI: 10.3390/ijms17101674] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023] Open
Abstract
Bone metastases are a common complication of epithelial cancers, of which breast, prostate and lung carcinomas are the most common. The establishment of cancer cells to distant sites such as the bone microenvironment requires multiple steps. Tumour cells can acquire properties to allow epithelial-to-mesenchymal transition, extravasation and migration. Within the bone metastatic niche, disseminated tumour cells may enter a dormancy stage or proliferate to adapt and survive, interacting with bone cells such as hematopoietic stem cells, osteoblasts and osteoclasts. Cross-talk with the bone may alter tumour cell properties and, conversely, tumour cells may also acquire characteristics of the surrounding microenvironment, in a process known as osteomimicry. Alternatively, these cells may also express osteomimetic genes that allow cell survival or favour seeding to the bone marrow. The seeding of tumour cells in the bone disrupts bone-forming and bone-resorbing activities, which can lead to macrometastasis in bone. At present, bone macrometastases are incurable with only palliative treatment available. A better understanding of how these processes influence the early onset of bone metastasis may give insight into potential therapies. This review will focus on the early steps of bone colonisation, once disseminated tumour cells enter the bone marrow.
Collapse
Affiliation(s)
- Casina Kan
- National Institute of Health and Medical Research (INSERM), UMR 1033, Lyon 69372, France.
- Faculty of Medicine RTH Laennec, University of Lyon, Villeurbanne 69372, France.
| | - Geoffrey Vargas
- National Institute of Health and Medical Research (INSERM), UMR 1033, Lyon 69372, France.
- Faculty of Medicine RTH Laennec, University of Lyon, Villeurbanne 69372, France.
| | - François Le Pape
- National Institute of Health and Medical Research (INSERM), UMR 1033, Lyon 69372, France.
- Faculty of Medicine RTH Laennec, University of Lyon, Villeurbanne 69372, France.
| | - Philippe Clézardin
- National Institute of Health and Medical Research (INSERM), UMR 1033, Lyon 69372, France.
- Faculty of Medicine RTH Laennec, University of Lyon, Villeurbanne 69372, France.
| |
Collapse
|
183
|
Lewis EEL, Wheadon H, Lewis N, Yang J, Mullin M, Hursthouse A, Stirling D, Dalby MJ, Berry CC. A Quiescent, Regeneration-Responsive Tissue Engineered Mesenchymal Stem Cell Bone Marrow Niche Model via Magnetic Levitation. ACS NANO 2016; 10:8346-8354. [PMID: 27602872 DOI: 10.1021/acsnano.6b02841] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The bone marrow niche represents a specialized environment that regulates mesenchymal stem cell quiescence and self-renewal, yet fosters stem cell migration and differentiation upon demand. An in vitro model that embodies these features would open up the ability to perform detailed study of stem cell behavior. In this paper we present a simple bone marrow-like niche model, which comprises of nanomagnetically levitated stem cells cultured as multicellular spheroids within a type I collagen gel. The stem cells maintained are nestin positive and remain quiescent until regenerative demand is placed upon them. In response to coculture wounding, they migrate and appropriately differentiate upon engraftment. This tissue engineered regeneration-responsive bone marrow-like niche model will allow for greater understanding of stem cell response to injury and also facilitate as a testing platform for drug candidates in a multiwell plate format.
Collapse
Affiliation(s)
| | - Helen Wheadon
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow , Shelley Road, Glasgow G11 0YN, United Kingdom
| | | | | | | | - Andrew Hursthouse
- School of Science, University of the West of Scotland , Paisley PA1 2BE, United Kingdom
| | - David Stirling
- School of Science, University of the West of Scotland , Paisley PA1 2BE, United Kingdom
| | | | | |
Collapse
|
184
|
Barbieri F, Bajetto A, Thellung S, Würth R, Florio T. Drug design strategies focusing on the CXCR4/CXCR7/CXCL12 pathway in leukemia and lymphoma. Expert Opin Drug Discov 2016; 11:1093-1109. [DOI: 10.1080/17460441.2016.1233176] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
185
|
Herzig MC, Cap AP. Challenges in translating mesenchymal stem cell therapies for trauma and critical care. Transfusion 2016; 56:20S-5S. [PMID: 27079318 DOI: 10.1111/trf.13566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Maryanne C Herzig
- Coagulation and Blood Research Program, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, Texas
| | - Andrew P Cap
- Coagulation and Blood Research Program, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, Texas
| |
Collapse
|
186
|
Fortin PY, Lepetit-Coiffé M, Genevois C, Debeissat C, Quesson B, Moonen CTW, Konsman JP, Couillaud F. Spatiotemporal control of gene expression in bone-marrow derived cells of the tumor microenvironment induced by MRI guided focused ultrasound. Oncotarget 2016; 6:23417-26. [PMID: 26299614 PMCID: PMC4695127 DOI: 10.18632/oncotarget.4288] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/02/2015] [Indexed: 11/25/2022] Open
Abstract
The tumor microenvironment is an interesting target for anticancer therapies but modifying this compartment is challenging. Here, we demonstrate the feasibility of a gene therapy strategy that combined targeting to bone marrow-derived tumor microenvironment using genetically modified bone-marrow derived cells and control of transgene expression by local hyperthermia through a thermo-inducible promoter. Chimera were obtained by engraftment of bone marrow from transgenic mice expressing reporter genes under transcriptional control of heat shock promoter and inoculated sub-cutaneously with tumors cells. Heat shocks were applied at the tumor site using a water bath or magnetic resonance guided high intensity focused ultrasound device. Reporter gene expression was followed by bioluminescence and fluorescence imaging and immunohistochemistry. Bone marrow-derived cells expressing reporter genes were identified to be mainly tumor-associated macrophages. We thus provide the proof of concept for a gene therapy strategy that allows for spatiotemporal control of transgenes expression by macrophages targeted to the tumor microenvironment.
Collapse
Affiliation(s)
- Pierre-Yves Fortin
- Laboratoire d'Imagerie Moléculaire et Fonctionnelle (IMF), CNRS/UMR 5231, Université de Bordeaux, Bordeaux, France.,Institut de Bio-Imagerie (IBIO), CNRS/UMS 3428, Université de Bordeaux, Bordeaux, France
| | - Matthieu Lepetit-Coiffé
- Laboratoire d'Imagerie Moléculaire et Fonctionnelle (IMF), CNRS/UMR 5231, Université de Bordeaux, Bordeaux, France
| | - Coralie Genevois
- Laboratoire d'Imagerie Moléculaire et Fonctionnelle (IMF), CNRS/UMR 5231, Université de Bordeaux, Bordeaux, France.,Institut de Bio-Imagerie (IBIO), CNRS/UMS 3428, Université de Bordeaux, Bordeaux, France
| | - Christelle Debeissat
- Laboratoire d'Imagerie Moléculaire et Fonctionnelle (IMF), CNRS/UMR 5231, Université de Bordeaux, Bordeaux, France
| | - Bruno Quesson
- Laboratoire d'Imagerie Moléculaire et Fonctionnelle (IMF), CNRS/UMR 5231, Université de Bordeaux, Bordeaux, France
| | - Chrit T W Moonen
- Laboratoire d'Imagerie Moléculaire et Fonctionnelle (IMF), CNRS/UMR 5231, Université de Bordeaux, Bordeaux, France
| | - Jan Pieter Konsman
- Centre de Résonance Magnétique des Systèmes Biologiques (RMSB), CNRS/UMR 5536, Université de Bordeaux, Bordeaux, France
| | - Franck Couillaud
- Laboratoire d'Imagerie Moléculaire et Fonctionnelle (IMF), CNRS/UMR 5231, Université de Bordeaux, Bordeaux, France.,Centre de Résonance Magnétique des Systèmes Biologiques (RMSB), CNRS/UMR 5536, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
187
|
Yurino A, Takenaka K, Yamauchi T, Nunomura T, Uehara Y, Jinnouchi F, Miyawaki K, Kikushige Y, Kato K, Miyamoto T, Iwasaki H, Kunisaki Y, Akashi K. Enhanced Reconstitution of Human Erythropoiesis and Thrombopoiesis in an Immunodeficient Mouse Model with Kit(Wv) Mutations. Stem Cell Reports 2016; 7:425-438. [PMID: 27499200 PMCID: PMC5031955 DOI: 10.1016/j.stemcr.2016.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 07/04/2016] [Accepted: 07/04/2016] [Indexed: 01/18/2023] Open
Abstract
In human-to-mouse xenograft models, reconstitution of human hematopoiesis is usually B-lymphoid dominant. Here we show that the introduction of homozygous Kit(Wv) mutations into C57BL/6.Rag2(null)Il2rg(null) mice with NOD-Sirpa (BRGS) strongly promoted human multi-lineage reconstitution. After xenotransplantation of human CD34(+)CD38(-) cord blood cells, these newly generated C57BL/6.Rag2(null)Il2rg(null)NOD-Sirpa Kit(Wv/Wv) (BRGSK(Wv/Wv)) mice showed significantly higher levels of human cell chimerism and long-term multi-lineage reconstitution compared with BRGS mice. Strikingly, this mouse displayed a robust reconstitution of human erythropoiesis and thrombopoiesis with terminal maturation in the bone marrow. Furthermore, depletion of host macrophages by clodronate administration resulted in the presence of human erythrocytes and platelets in the circulation. Thus, attenuation of mouse KIT signaling greatly enhances the multi-lineage differentiation of human hematopoietic stem and progenitor cells (HSPCs) in mouse bone marrow, presumably by outcompeting mouse HSPCs to occupy suitable microenvironments. The BRGSK(Wv/Wv) mouse model is a useful tool to study human multi-lineage hematopoiesis.
Collapse
Affiliation(s)
- Ayano Yurino
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Katsuto Takenaka
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Takuji Yamauchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Takuya Nunomura
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yasufumi Uehara
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Fumiaki Jinnouchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Kohta Miyawaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yoshikane Kikushige
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Koji Kato
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Toshihiro Miyamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiromi Iwasaki
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Yuya Kunisaki
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan; Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka 812-8582, Japan.
| |
Collapse
|
188
|
Hur J, Baek SH, Kim HS. KAI1(CD82)-DARC(CD234) axis in the stem cell niche. Cell Cycle 2016; 15:1945-7. [PMID: 27163847 PMCID: PMC4968970 DOI: 10.1080/15384101.2016.1184514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 10/21/2022] Open
Affiliation(s)
- Jin Hur
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Hee Baek
- Creative Research Initiative Center for Chromatin Dynamics, School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Republic of Korea
- Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
189
|
Tunneling nanotubes mediate the transfer of stem cell marker CD133 between hematopoietic progenitor cells. Exp Hematol 2016; 44:1092-1112.e2. [PMID: 27473566 DOI: 10.1016/j.exphem.2016.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 12/24/2022]
Abstract
Deciphering all mechanisms of intercellular communication used by hematopoietic progenitors is important, not only for basic stem cell research, but also in view of their therapeutic relevance. Here, we investigated whether these cells can produce the thin F-actin-based plasma membrane protrusions referred to as tunneling nanotubes (TNTs), which are known to bridge cells over long distances without contact with the substratum and transfer cargo molecules along them in various biological processes. We found that human primary CD34+ hematopoietic progenitors and leukemic KG1a cells develop such structures upon culture on primary mesenchymal stromal cells or specific extracellular-matrix-based substrata. Time-lapse video microscopy revealed that cell dislodgement is the primary mechanism responsible for TNT biogenesis. Surprisingly, we found that, among various cluster of differentiation (CD) markers, only the stem cell antigen CD133 is transferred between cells. It is selectively and directionally transported along the surface of TNTs in small clusters, such as cytoplasmic phospho-myosin light chain 2, suggesting that the latter actin motor protein might be implicated in this process. Our data provide new insights into the biology of hematopoietic progenitors that can contribute to our understanding of all facets of intercellular communication in the bone marrow microenvironment under healthy or cancerous conditions.
Collapse
|
190
|
Mesenchymal Stem Cells and Myeloid Derived Suppressor Cells: Common Traits in Immune Regulation. J Immunol Res 2016; 2016:7121580. [PMID: 27529074 PMCID: PMC4978836 DOI: 10.1155/2016/7121580] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/08/2016] [Indexed: 02/08/2023] Open
Abstract
To protect host against immune-mediated damage, immune responses are tightly regulated. The regulation of immune responses is mediated by various populations of mature immune cells, such as T regulatory cells and B regulatory cells, but also by immature cells of different origins. In this review, we discuss regulatory properties and mechanisms whereby two distinct populations of immature cells, mesenchymal stem cells, and myeloid derived suppressor cells mediate immune regulation, focusing on their similarities, discrepancies, and potential clinical applications.
Collapse
|
191
|
Roberts J, Sahoo JK, McNamara LE, Burgess KV, Yang J, Alakpa EV, Anderson HJ, Hay J, Turner LA, Yarwood SJ, Zelzer M, Oreffo RC, Ulijn RV, Dalby MJ. Dynamic Surfaces for the Study of Mesenchymal Stem Cell Growth through Adhesion Regulation. ACS NANO 2016; 10:6667-79. [PMID: 27322014 PMCID: PMC4963921 DOI: 10.1021/acsnano.6b01765] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 06/20/2016] [Indexed: 05/18/2023]
Abstract
Out of their niche environment, adult stem cells, such as mesenchymal stem cells (MSCs), spontaneously differentiate. This makes both studying these important regenerative cells and growing large numbers of stem cells for clinical use challenging. Traditional cell culture techniques have fallen short of meeting this challenge, but materials science offers hope. In this study, we have used emerging rules of managing adhesion/cytoskeletal balance to prolong MSC cultures by fabricating controllable nanoscale cell interfaces using immobilized peptides that may be enzymatically activated to change their function. The surfaces can be altered (activated) at will to tip adhesion/cytoskeletal balance and initiate differentiation, hence better informing biological mechanisms of stem cell growth. Tools that are able to investigate the stem cell phenotype are important. While large phenotypical differences, such as the difference between an adipocyte and an osteoblast, are now better understood, the far more subtle differences between fibroblasts and MSCs are much harder to dissect. The development of technologies able to dynamically navigate small differences in adhesion are critical in the race to provide regenerative strategies using stem cells.
Collapse
Affiliation(s)
- Jemma
N. Roberts
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| | - Jugal Kishore Sahoo
- Department
of Pure & Applied Chemistry, WestCHEM, Thomas Graham Building, 295 Cathedral
Street, Glasgow G1 1XL, Scotland, U.K.
| | - Laura E. McNamara
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| | - Karl V. Burgess
- Glasgow
Polyomics Facility, Translational Cancer Research Centre, University of Glasgow Garscube Campus, Switchback Road, Glasgow G61 1QH, Scotland, U.K.
| | - Jingli Yang
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| | - Enateri V. Alakpa
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| | - Hilary J. Anderson
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| | - Jake Hay
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| | - Lesley-Anne Turner
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| | - Stephen J. Yarwood
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| | - Mischa Zelzer
- School
of Pharmacy, University of Nottingham, Boots Science Building, University Park, Nottingham NG7 2RD, U.K.
- National
Physical Laboratory, Teddington, Middlesex TW11 0LW, U.K.
| | - Richard
O. C. Oreffo
- Bone
&
Joint Research Group, Centre for Human Development, Stem Cells and
Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, U.K.
| | - Rein V. Ulijn
- Department
of Pure & Applied Chemistry, WestCHEM, Thomas Graham Building, 295 Cathedral
Street, Glasgow G1 1XL, Scotland, U.K.
- Advanced
Science Research Center (ASRC), City University
of New York, New York, New York 10031, United
States
- Department
of Chemistry and Biochemistry, City University
of New York—Hunter College, 695 Park Avenue, New York, New York 10065, United
States
| | - Matthew J. Dalby
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K.
| |
Collapse
|
192
|
Lister Z, Rayner KJ, Suuronen EJ. How Biomaterials Can Influence Various Cell Types in the Repair and Regeneration of the Heart after Myocardial Infarction. Front Bioeng Biotechnol 2016; 4:62. [PMID: 27486578 PMCID: PMC4948030 DOI: 10.3389/fbioe.2016.00062] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/01/2016] [Indexed: 12/15/2022] Open
Abstract
The healthy heart comprises many different cell types that work together to preserve optimal function. However, in a diseased heart the function of one or more cell types is compromised which can lead to many adverse events, one of which is myocardial infarction (MI). Immediately after MI, the cardiac environment is characterized by excessive cardiomyocyte death and inflammatory signals leading to the recruitment of macrophages to clear the debris. Proliferating fibroblasts then invade, and a collagenous scar is formed to prevent rupture. Better functional restoration of the heart is not achieved due to the limited regenerative capacity of cardiac tissue. To address this, biomaterial therapy is being investigated as an approach to improve regeneration in the infarcted heart, as they can possess the potential to control cell function in the infarct environment and limit the adverse compensatory changes that occur post-MI. Over the past decade, there has been considerable research into the development of biomaterials for cardiac regeneration post-MI; and various effects have been observed on different cell types depending on the biomaterial that is applied. Biomaterial treatment has been shown to enhance survival, improve function, promote proliferation, and guide the mobilization and recruitment of different cells in the post-MI heart. This review will provide a summary on the biomaterials developed to enhance cardiac regeneration and remodeling post-MI with a focus on how they control macrophages, cardiomyocytes, fibroblasts, and endothelial cells. A better understanding of how a biomaterial interacts with the different cell types in the heart may lead to the development of a more optimized biomaterial therapy for cardiac regeneration.
Collapse
Affiliation(s)
- Zachary Lister
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Katey J Rayner
- Atherosclerosis, Genomics and Cell Biology Group, University of Ottawa Heart Institute , Ottawa, ON , Canada
| | - Erik J Suuronen
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
193
|
de Lima KA, de Oliveira GLV, Yaochite JNU, Pinheiro DG, de Azevedo JTC, Silva WA, Covas DT, Couri CEB, Simões BP, Voltarelli JC, Oliveira MC, Malmegrim KCR. Transcriptional profiling reveals intrinsic mRNA alterations in multipotent mesenchymal stromal cells isolated from bone marrow of newly-diagnosed type 1 diabetes patients. Stem Cell Res Ther 2016; 7:92. [PMID: 27406064 PMCID: PMC4942931 DOI: 10.1186/s13287-016-0351-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/12/2016] [Accepted: 06/22/2016] [Indexed: 02/07/2023] Open
Abstract
Background Bone marrow multipotent mesenchymal stromal cells (MSCs) are a diverse subset of precursors that contribute to the homeostasis of the hematopoietic niche. MSCs can be isolated and expanded in vitro and have unique immunomodulatory and regenerative properties that make them attractive for the treatment of autoimmune diseases, including type 1 diabetes (T1D). Whether autologous or allogeneic MSCs are more suitable for therapeutic purposes has not yet been established. While autologous MSCs may present abnormal function, allogeneic cells may be recognized and rejected by the host immune system. Thus, studies that investigate biological characteristics of MSCs isolated from T1D patients are essential to guide future clinical applications. Methods Bone marrow-derived MSCs from recently diagnosed type 1 diabetes patients (T1D-MSCs) were compared with those from healthy individuals (C-MSCs) for morphological and immunophenotypic characteristics and for differentiation potential. Bioinformatics approaches allowed us to match absolute and differential gene expression of several adhesion molecules, immune mediators, growth factors, and their receptors involved with hematopoietic support and immunomodulatory properties of MSCs. Finally, the differentially expressed genes were collated for functional pathway enrichment analysis. Results T1D-MSCs and C-MSCs were similar for morphology, immunophenotype, and differentiation potential. Our absolute gene expression results supported previous literature reports, while also detecting new potential molecules related to bone marrow-derived MSC functions. T1D-MSCs showed intrinsic abnormalities in mRNA expression, including the immunomodulatory molecules VCAM-1, CXCL12, HGF, and CCL2. Pathway analyses revealed activation of sympathetic nervous system and JAK STAT signaling in T1D-MSCs. Conclusions Collectively, our results indicate that MSCs isolated from T1D patients present intrinsic transcriptional alterations that may affect their therapeutic potential. However, the implications of these abnormalities in T1D development as well as in the therapeutic efficacy of autologous MSCs require further investigation. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0351-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kalil A de Lima
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil. .,Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil. .,, Tenente Catao Roxo, 2501, Monte Alegre, 14051-140, Ribeirao Preto, Sao Paulo, Brazil.
| | - Gislane L V de Oliveira
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Juliana N U Yaochite
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, Ceara, Brazil
| | - Daniel G Pinheiro
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Júlia T C de Azevedo
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Wilson Araujo Silva
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Dimas T Covas
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Carlos E B Couri
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Belinda P Simões
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Julio C Voltarelli
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Maria C Oliveira
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Kelen C R Malmegrim
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
194
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
195
|
Rahmanifar F, Tamadon A, Mehrabani D, Zare S, Abasi S, Keshavarz S, Dianatpour M, Khodabandeh Z, Jahromi IRG, Koohi-Hoseinabadi O. Histomorphometric evaluation of treatment of rat azoosper-mic seminiferous tubules by allotransplantation of bone marrow-derived mesenchymal stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:653-61. [PMID: 27482347 PMCID: PMC4951605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVES Bone marrow-derived mesenchymal stem cells (BM-MSCs) potentials make them appropriate for cell therapy including ability of differentiation and release of anti-inflammatory cytokines and growth factors secreta. For treatment of azoospermia to induce proliferation and differentiation of germ cells, MSCs transplantation has been introduced. The aim of the present experimental case-control study was to histomorphometric evaluation of the germinal cells in seminiferous tubules of azoospermic rats before and after BM-MSCs allotransplantation. MATERIALS AND METHODS In the present study, BM-MSCs were isolated from six male rats and confirmed. Their testes also served as intact negative controls. The recipient rats (n=6) were received two doses of 10 mg/kg of busulfan with 21 days interval to induce azoospermia. After cessation of spermatogenesis, the rats were allotransplanted with the BM-MSCs into efferent duct of right testes. Thirty-five days later, the right cell-treated testes were compared to left azoospermic ones. RESULTS Histomorphometric analyses showed that the seminiferous tubules treated with BM-MSCs had normal morphology in comparison with azoospermic testes, which were without germinal layer. In most BM-MSCs-treated seminiferous tubules, spermatogenesis was observed. CONCLUSION The allotransplanted BM-MSCs could induce spermatogenesis in seminiferous tubules of azoospermic rats.
Collapse
Affiliation(s)
- Farhad Rahmanifar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amin Tamadon
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Davood Mehrabani
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,Corresponding author: Davood Mehrabani. Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. Tel/Fax: +98-71 32341025; emails:
| | - Shahrokh Zare
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sorush Abasi
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeideh Keshavarz
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khodabandeh
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | |
Collapse
|
196
|
Scheller EL, Cawthorn WP, Burr AA, Horowitz MC, MacDougald OA. Marrow Adipose Tissue: Trimming the Fat. Trends Endocrinol Metab 2016; 27:392-403. [PMID: 27094502 PMCID: PMC4875855 DOI: 10.1016/j.tem.2016.03.016] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023]
Abstract
Marrow adipose tissue (MAT) is a unique fat depot, located in the skeleton, that has the potential to contribute to both local and systemic metabolic processes. In this review we highlight several recent conceptual developments pertaining to the origin and function of MAT adipocytes; consider the relationship of MAT to beige, brown, and white adipose depots; explore MAT expansion and turnover in humans and rodents; and discuss future directions for MAT research in the context of endocrine function and metabolic disease. MAT has the potential to exert both local and systemic effects on metabolic homeostasis, skeletal remodeling, hematopoiesis, and the development of bone metastases. The diversity of these functions highlights the breadth of the potential impact of MAT on health and disease.
Collapse
Affiliation(s)
- Erica L Scheller
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University, Saint Louis, MO 63110, USA.
| | - William P Cawthorn
- University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Aaron A Burr
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
197
|
Shipounova IN, Petinati NA, Bigildeev AE, Drize NJ, Sorokina TV, Kuzmina LA, Parovichnikova EN, Savchenko VG. Alterations of the bone marrow stromal microenvironment in adult patients with acute myeloid and lymphoblastic leukemias before and after allogeneic hematopoietic stem cell transplantation. Leuk Lymphoma 2016; 58:408-417. [PMID: 27244369 DOI: 10.1080/10428194.2016.1187277] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Bone marrow (BM) derived adult multipotent mesenchymal stromal cells (MMSCs) and fibroblast colony-forming units (CFU-Fs) of 20 patients with acute myeloid leukemia (AML) and 15 patients with acute lymphoblastic leukemia (ALL) before and during 1 year after receiving allogeneic hematopoietic stem cell transplantation (allo-HSCT) were studied. The growth characteristics of MMSCs of all patients before allo-HSCT were not altered; however, relative expression level (REL) of some genes in MMSCs, but not in CFU-Fs, from AML and ALL patients significantly changed. After allo-HSCT, CFU-F concentration and MMSC production were significantly decreased for 1 year; REL of several genes in MMSCs and CFU-F-derived colonies were also significantly downregulated. Thus, chemotherapy that was used for induction of remission did not impair the function of stromal precursors, but gene expression levels were altered. Allo-HSCT conditioning regimens significantly damaged MMSCs and CFU-Fs, and the effect lasted for at least 1 year.
Collapse
Affiliation(s)
- Irina N Shipounova
- a Physiology of Hematopoiesis Lab , Federal Government Budget Institution National Research Center for Hematology, Ministry of Health , Moscow , Russian Federation
| | - Nataliya A Petinati
- a Physiology of Hematopoiesis Lab , Federal Government Budget Institution National Research Center for Hematology, Ministry of Health , Moscow , Russian Federation
| | - Alexey E Bigildeev
- a Physiology of Hematopoiesis Lab , Federal Government Budget Institution National Research Center for Hematology, Ministry of Health , Moscow , Russian Federation
| | - Nina J Drize
- a Physiology of Hematopoiesis Lab , Federal Government Budget Institution National Research Center for Hematology, Ministry of Health , Moscow , Russian Federation
| | - Tamara V Sorokina
- b Department of High-Dose Chemotherapy , Depressions of Hematopoiesis and Bone Marrow Transplantation, Federal Government Budget Institution National Research Center for Hematology, Ministry of Health , Moscow , Russian Federation
| | - Larisa A Kuzmina
- b Department of High-Dose Chemotherapy , Depressions of Hematopoiesis and Bone Marrow Transplantation, Federal Government Budget Institution National Research Center for Hematology, Ministry of Health , Moscow , Russian Federation
| | - Elena N Parovichnikova
- b Department of High-Dose Chemotherapy , Depressions of Hematopoiesis and Bone Marrow Transplantation, Federal Government Budget Institution National Research Center for Hematology, Ministry of Health , Moscow , Russian Federation
| | - Valeri G Savchenko
- c Federal Government Budget Institution National Research Center for Hematology, Ministry of Health , Moscow , Russian Federation
| |
Collapse
|
198
|
Park MH, Lee JK, Kim N, Min WK, Lee JE, Kim KT, Akiyama H, Herzog H, Schuchman EH, Jin HK, Bae JS. Neuropeptide Y Induces Hematopoietic Stem/Progenitor Cell Mobilization by Regulating Matrix Metalloproteinase-9 Activity Through Y1 Receptor in Osteoblasts. Stem Cells 2016; 34:2145-56. [PMID: 27090492 DOI: 10.1002/stem.2383] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 03/04/2016] [Accepted: 03/26/2016] [Indexed: 12/15/2022]
Abstract
Hematopoietic stem/progenitor cell (HSPC) mobilization is an essential homeostatic process regulated by the interaction of cellular and molecular components in bone marrow niches. It has been shown by others that neurotransmitters released from the sympathetic nervous system regulate HSPC egress from bone marrow to peripheral blood. In this study, we investigate the functional role of neuropeptide Y (NPY) on this process. NPY deficient mice had significantly impaired HSPC mobilization due to increased expression of HSPC maintenance factors by reduction of matrix metalloproteinase-9 (MMP-9) activity in bone marrow. Pharmacological or endogenous elevation of NPY led to decrease of HSPC maintenance factors expression by activating MMP-9 in osteoblasts, resulting in HSPC mobilization. Mice in which the Y1 receptor was deleted in osteoblasts did not exhibit HSPC mobilization by NPY. Furthermore, NPY treatment in ovariectomized mice caused reduction of bone loss due to HSPC mobilization. These results suggest a new role of NPY on HSPC mobilization, as well as the potential therapeutic application of this neuropeptide for stem cell-based therapy. Stem Cells 2016;34:2145-2156.
Collapse
Affiliation(s)
- Min Hee Park
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Kyungpook National University, Daegu, Korea.,Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Korea.,Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Korea
| | - Jong Kil Lee
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Kyungpook National University, Daegu, Korea.,Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Korea.,Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Korea
| | - Namoh Kim
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Kyungpook National University, Daegu, Korea.,Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Korea.,Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Korea
| | - Woo-Kie Min
- Department of Orthopaedic Surgery, Kyungpook National University Hospital, Daegu, Korea
| | - Jeong Eun Lee
- Department of Radiation Oncology, Kyungpook National University Hospital, Daegu, Korea
| | - Kyoung-Tae Kim
- Department of Neurosurgery School of Medicine, Kyungpook National University, Daegu, Korea
| | | | - Herbert Herzog
- Neuroscience Research Program, Neuroscience Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Edward H Schuchman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hee Kyung Jin
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Kyungpook National University, Daegu, Korea.,Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Korea
| | - Jae-Sung Bae
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Kyungpook National University, Daegu, Korea.,Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Korea.,Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Korea
| |
Collapse
|
199
|
Anderson HJ, Sahoo JK, Ulijn RV, Dalby MJ. Mesenchymal Stem Cell Fate: Applying Biomaterials for Control of Stem Cell Behavior. Front Bioeng Biotechnol 2016; 4:38. [PMID: 27242999 PMCID: PMC4865671 DOI: 10.3389/fbioe.2016.00038] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/18/2016] [Indexed: 01/28/2023] Open
Abstract
The materials pipeline for biomaterials and tissue engineering applications is under continuous development. Specifically, there is great interest in the use of designed materials in the stem cell arena as materials can be used to manipulate the cells providing control of behavior. This is important as the ability to "engineer" complexity and subsequent in vitro growth of tissues and organs is a key objective for tissue engineers. This review will describe the nature of the materials strategies, both static and dynamic, and their influence specifically on mesenchymal stem cell fate.
Collapse
Affiliation(s)
| | - Jugal Kishore Sahoo
- Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow, UK
| | - Rein V. Ulijn
- Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow, UK
- Advanced Science Research Centre (ASRC), Hunter College, City University of New York, New York, NY, USA
| | | |
Collapse
|
200
|
Hur J, Choi JI, Lee H, Nham P, Kim TW, Chae CW, Yun JY, Kang JA, Kang J, Lee SE, Yoon CH, Boo K, Ham S, Roh TY, Jun JK, Lee H, Baek SH, Kim HS. CD82/KAI1 Maintains the Dormancy of Long-Term Hematopoietic Stem Cells through Interaction with DARC-Expressing Macrophages. Cell Stem Cell 2016; 18:508-21. [PMID: 26996598 DOI: 10.1016/j.stem.2016.01.013] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 12/10/2015] [Accepted: 01/20/2016] [Indexed: 12/14/2022]
Abstract
Hematopoiesis is regulated by crosstalk between long-term repopulating hematopoietic stem cells (LT-HSCs) and supporting niche cells in the bone marrow (BM). Here, we examine the role of CD82/KAI1 in niche-mediated LT-HSC maintenance. We found that CD82/KAI1 is expressed predominantly on LT-HSCs and rarely on other hematopoietic stem-progenitor cells (HSPCs). In Cd82(-/-) mice, LT-HSCs were selectively lost as they exited from quiescence and differentiated. Mechanistically, CD82-based TGF-β1/Smad3 signaling leads to induction of CDK inhibitors and cell-cycle inhibition. The CD82 binding partner DARC/CD234 is expressed on macrophages and stabilizes CD82 on LT-HSCs, promoting their quiescence. When DARC(+) BM macrophages were ablated, the level of surface CD82 on LT-HSCs decreased, leading to cell-cycle entry, proliferation, and differentiation. A similar interaction appears to be relevant for human HSPCs. Thus, CD82 is a functional surface marker of LT-HSCs that maintains quiescence through interaction with DARC-expressing macrophages in the BM stem cell niche.
Collapse
Affiliation(s)
- Jin Hur
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Jae-Il Choi
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Hwan Lee
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Pniel Nham
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Tae-Won Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Cheong-Whan Chae
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Ji-Yeon Yun
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Jin-A Kang
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Jeehoon Kang
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Sang Eun Lee
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Chang-Hwan Yoon
- Cardiovascular Center and Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do 463-707, Republic of Korea
| | - Kyungjin Boo
- Creative Research Initiative Center for Chromatin Dynamics, School of Biological Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Seokjin Ham
- BK21PLUS Fellowship Program, Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Tae-Young Roh
- BK21PLUS Fellowship Program, Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Jong Kwan Jun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea
| | - Ho Lee
- Division of Convergence Technology, National Cancer Center, Gyeonggi-do 410-769, Republic of Korea
| | - Sung Hee Baek
- Creative Research Initiative Center for Chromatin Dynamics, School of Biological Sciences, Seoul National University, Seoul 151-742, Republic of Korea.
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul 110-744, Republic of Korea; Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 110-744, Republic of Korea.
| |
Collapse
|