501
|
Huang X, Zhu B, Wang X, Xiao R, Wang C. Three-dimensional co-culture of mesenchymal stromal cells and differentiated osteoblasts on human bio-derived bone scaffolds supports active multi-lineage hematopoiesis in vitro: Functional implication of the biomimetic HSC niche. Int J Mol Med 2016; 38:1141-51. [PMID: 27571775 PMCID: PMC5029969 DOI: 10.3892/ijmm.2016.2712] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 06/29/2016] [Indexed: 01/08/2023] Open
Abstract
Recent studies have indicated that the hematopoietic stem/progenitor cell (HSPC) niche, consisting of two major crucial components, namely osteoblasts (OBs) and mesenchymal stromal cells (MSCs), is responsible for the fate of HSPCs. Thus, closely mimicking the HSPC niche ex vivo may be an efficient strategy with which to develop new culture strategies to specifically regulate the balance between HSPC self-renewal and proliferation. The aim of this study was to establish a novel HSPC three-dimensional culture system by co-culturing bone marrow-derived MSCs and OBs differentiated from MSCs without any cytokines as feeder cells and applying bio-derived bone from human femoral metaphyseal portion as the scaffold. Scanning electron microscopy revealed the excellent biocompatibility of bio-derived bone with bone marrow-derived MSCs and OBs differentiated from MSCs. Western blot analysis revealed that many cytokines, which play key roles in HSPC regulation, were comprehensively secreted, while ELISA revealed that extracellular matrix molecules were also highly expressed. Hoechst 33342/propidium iodide fluorescence staining proved that our system could be used to supply a long-term culture of HSPCs. Flow cytometric analysis and qPCR of p21 expression demonstrated that our system significantly promoted the self-renewal and ex vivo expansion of HSPCs. Colony-forming unit (CFU) and long-term culture-initiating cell (LTC-IC) assays confirmed that our system has the ability for both the expansion of CD34+ hematopoietic stem cells (HPCs) and the maintenance of a primitive cell subpopulation of HSCs. The severe-combined immunodeficient mouse repopulating cell assay revealed the promoting effects of our system on the expansion of long-term primitive transplantable HSCs. In conclusion, our system may be a more comprehensive and balanced system which not only promotes the self-renewal and ex vivo expansion of HSPCs, but also maintains primitive HPCs with superior phenotypic and functional attributes.
Collapse
Affiliation(s)
- Xiaobing Huang
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Biao Zhu
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xiaodong Wang
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Rong Xiao
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Chunsen Wang
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
502
|
Liu Y, Yi L, Wang L, Chen L, Chen X, Wang Y. Ginsenoside Rg1 protects human umbilical cord blood-derived stromal cells against tert-Butyl hydroperoxide-induced apoptosis through Akt-FoxO3a-Bim signaling pathway. Mol Cell Biochem 2016; 421:75-87. [PMID: 27522666 DOI: 10.1007/s11010-016-2786-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022]
Abstract
Human umbilical cord blood-derived stromal cells (hUCBDSCs) possess strong capability of supporting hematopoiesis and immune regulation, whereas some stress conditions cause reactive oxygen species (ROS) accumulation and then lead to oxidative injury and cell apoptosis. Ginsenoside Rg1 (G-Rg1) has been demonstrated to exert antioxidative and prosurvival effects in many cell types. In this study, the tert-Butyl hydroperoxide (t-BHP), an analog of hydroperoxide, was utilized to mimic the oxidative damage to hUCBDSCs. We aimed to investigate the effects of Ginsenoside Rg1 on protecting hUCBDSCs from t-BHP-induced oxidative injury and apoptosis, as well as the possible signaling pathway involved. It was shown that the treatment of hUCBDSCs with G-Rg1 markedly restored the t-BHP-induced cell viability loss, promoted the CFU-F formation, and inhibited cell apoptosis. G-Rg1 also caused a reduced production of LDH and MDA while significantly enhancing the activity of SOD. Mechanistically, G-Rg1 promoted the phosphorylation of Akt and FoxO3a and led to the cytoplasmic translocation of FoxO3a, which in turn suppressed FoxO3a-modulated expression of proapoptotic Bim and elevated the ratio of Bcl-2 to Bax. All these results suggest that G-Rg1 enhances the survival of t-BHP-induced hUCBDSCs and protects them against apoptosis at least partially through Akt-FoxO3a-Bim signaling pathway.
Collapse
Affiliation(s)
- Ying Liu
- Department of Histology and Embryology, Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Long Yi
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, People's Republic of China
| | - Lu Wang
- Department of Histology and Embryology, Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Linbo Chen
- Department of Histology and Embryology, Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xiongbin Chen
- Department of Histology and Embryology, Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yaping Wang
- Department of Histology and Embryology, Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
503
|
Silberstein L, Goncalves KA, Kharchenko PV, Turcotte R, Kfoury Y, Mercier F, Baryawno N, Severe N, Bachand J, Spencer JA, Papazian A, Lee D, Chitteti BR, Srour EF, Hoggatt J, Tate T, Lo Celso C, Ono N, Nutt S, Heino J, Sipilä K, Shioda T, Osawa M, Lin CP, Hu GF, Scadden DT. Proximity-Based Differential Single-Cell Analysis of the Niche to Identify Stem/Progenitor Cell Regulators. Cell Stem Cell 2016; 19:530-543. [PMID: 27524439 DOI: 10.1016/j.stem.2016.07.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/08/2016] [Accepted: 07/07/2016] [Indexed: 01/17/2023]
Abstract
Physiological stem cell function is regulated by secreted factors produced by niche cells. In this study, we describe an unbiased approach based on the differential single-cell gene expression analysis of mesenchymal osteolineage cells close to, and further removed from, hematopoietic stem/progenitor cells (HSPCs) to identify candidate niche factors. Mesenchymal cells displayed distinct molecular profiles based on their relative location. We functionally examined, among the genes that were preferentially expressed in proximal cells, three secreted or cell-surface molecules not previously connected to HSPC biology-the secreted RNase angiogenin, the cytokine IL18, and the adhesion molecule Embigin-and discovered that all of these factors are HSPC quiescence regulators. Therefore, our proximity-based differential single-cell approach reveals molecular heterogeneity within niche cells and can be used to identify novel extrinsic stem/progenitor cell regulators. Similar approaches could also be applied to other stem cell/niche pairs to advance the understanding of microenvironmental regulation of stem cell function.
Collapse
Affiliation(s)
- Lev Silberstein
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kevin A Goncalves
- Graduate Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA; Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Peter V Kharchenko
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Raphael Turcotte
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02445, USA
| | - Youmna Kfoury
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Francois Mercier
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ninib Baryawno
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Nicolas Severe
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jacqueline Bachand
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joel A Spencer
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ani Papazian
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Dongjun Lee
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | - Jonathan Hoggatt
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Tiffany Tate
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | | | - Noriaki Ono
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephen Nutt
- Walter and Eliza Hall Research Institute, Parkville, VIC 3052, Australia
| | | | | | - Toshihiro Shioda
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Charles P Lin
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02445, USA
| | - Guo-Fu Hu
- Graduate Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA; Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA.
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02445, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
504
|
Lei K, Thi-Kim Vu H, Mohan RD, McKinney SA, Seidel CW, Alexander R, Gotting K, Workman JL, Sánchez Alvarado A. Egf Signaling Directs Neoblast Repopulation by Regulating Asymmetric Cell Division in Planarians. Dev Cell 2016; 38:413-29. [PMID: 27523733 DOI: 10.1016/j.devcel.2016.07.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 06/01/2016] [Accepted: 07/15/2016] [Indexed: 12/27/2022]
Abstract
A large population of proliferative stem cells (neoblasts) is required for physiological tissue homeostasis and post-injury regeneration in planarians. Recent studies indicate that survival of a few neoblasts after sublethal irradiation results in the clonal expansion of the surviving stem cells and the eventual restoration of tissue homeostasis and regenerative capacity. However, the precise mechanisms regulating the population dynamics of neoblasts remain largely unknown. Here, we uncovered a central role for epidermal growth factor (EGF) signaling during in vivo neoblast expansion mediated by Smed-egfr-3 (egfr-3) and its putative ligand Smed-neuregulin-7 (nrg-7). Furthermore, the EGF receptor-3 protein localizes asymmetrically on the cytoplasmic membrane of neoblasts, and the ratio of asymmetric to symmetric cell divisions decreases significantly in egfr-3(RNAi) worms. Our results not only provide the first molecular evidence of asymmetric stem cell divisions in planarians, but also demonstrate that EGF signaling likely functions as an essential regulator of neoblast clonal expansion.
Collapse
Affiliation(s)
- Kai Lei
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.
| | - Hanh Thi-Kim Vu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Ryan D Mohan
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Sean A McKinney
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Chris W Seidel
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Kirsten Gotting
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Jerry L Workman
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Alejandro Sánchez Alvarado
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA; Howard Hughes Medical Institute, Stowers Institute for Medical Research, Kansas City, MO 64110, USA.
| |
Collapse
|
505
|
Role of bone marrow macrophages in controlling homeostasis and repair in bone and bone marrow niches. Semin Cell Dev Biol 2016; 61:12-21. [PMID: 27521519 DOI: 10.1016/j.semcdb.2016.08.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/09/2016] [Accepted: 08/09/2016] [Indexed: 12/24/2022]
Abstract
Macrophages, named for their phagocytic ability, participate in homeostasis, tissue regeneration and inflammatory responses. Bone and adjacent marrow contain multiple functionally unique resident tissue macrophage subsets which maintain and regulate anatomically distinct niche environments within these interconnected tissues. Three subsets of bone-bone marrow resident tissue macrophages have been characterised; erythroblastic island macrophages, haematopoietic stem cell niche macrophages and osteal macrophages. The role of these macrophages in controlling homeostasis and repair in bone and bone marrow niches is reviewed in detail.
Collapse
|
506
|
De La Garza A, Sinha A, Bowman TV. Concise Review: Hematopoietic Stem Cell Origins: Lessons from Embryogenesis for Improving Regenerative Medicine. Stem Cells Transl Med 2016; 6:60-67. [PMID: 28170201 PMCID: PMC5442726 DOI: 10.5966/sctm.2016-0110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/16/2016] [Indexed: 12/04/2022] Open
Abstract
Hematopoietic stem cells (HSCs) have extensive regenerative capacity to replace all blood cell types, an ability that is harnessed in the clinic for bone marrow transplantation. Finding appropriate donors remains a major limitation to more extensive usage of HSC‐based therapies. Derivation of patient‐specific HSCs from pluripotent stem cells offers great promise to remedy this problem if scientists could crack the code on how to make robust, transplantable HSCs in a dish. Studies delving into the native origins of HSC production during embryonic development should supply the necessary playbook. This review presents recent discoveries from animal models, with a focus on zebrafish, and discusses the implications of these new advances in the context of prior knowledge. The focus is on the latest research exploring the role of epigenetic regulation, signaling pathways, and niche components needed for proper HSC formation. These studies provide new directions that should be explored for de novo generation and expansion of HSCs for regenerative therapies. Stem Cells Translational Medicine2017;6:60–67
Collapse
Affiliation(s)
- Adriana De La Garza
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Arpan Sinha
- Division of Pediatric Hematology/Oncology, Children's Hospital at Montefiore, Bronx, New York, USA
| | - Teresa V. Bowman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
507
|
Kusumbe AP. Vascular niches for disseminated tumour cells in bone. J Bone Oncol 2016; 5:112-116. [PMID: 27761369 PMCID: PMC5063228 DOI: 10.1016/j.jbo.2016.04.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 11/26/2022] Open
Abstract
The vasculature of the skeletal system regulates osteogenesis and hematopoiesis, in addition to its primary function as a transportation network. Recent studies suggest that the vasculature in bone regulates multiple steps involved in the metastatic cascade. Matrix and growth factor abundant vascular microenvironments in bone not only provide a fertile soil for the metastatic growth but also support the dormancy of Disseminated Tumour Cells (DTCs). Interestingly, vasculature also seems to direct the reactivation of dormant DTCs. Targeting such early steps of bone metastasis by directing therapies against vascular niches can lead to the development of effective therapeutic strategies that delay or even prevent the metastatic relapse. However, this would require a detailed understanding of the regulatory mechanisms that govern the interaction between endothelial cells and DTCs in the early stages of bone metastasis. This review aims to highlight the importance of vascular niches and outline their newly identified roles during bone metastasis.
Collapse
Affiliation(s)
- Anjali P. Kusumbe
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, UK
- Max-Planck-Institute for Molecular Biomedicine and University of Münster, D–48149 Münster, Germany
| |
Collapse
|
508
|
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: 2.3] [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
|
509
|
Satyamitra MM, DiCarlo AL, Taliaferro L. Understanding the Pathophysiology and Challenges of Development of Medical Countermeasures for Radiation-Induced Vascular/Endothelial Cell Injuries: Report of a NIAID Workshop, August 20, 2015. Radiat Res 2016; 186:99-111. [PMID: 27387859 DOI: 10.1667/rr14436.1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
After the events of September 11, 2001, a decade of research on the development of medical countermeasures (MCMs) to treat victims of a radiological incident has yielded two FDA-approved agents to mitigate acute radiation syndrome. These licensed agents specifically target the mitigation of radiation-induced neutropenia and infection potential, while the ramifications of the exposure event in a public health emergency incident could include the entire body, causing additional acute and/or delayed organ/tissue injuries. Anecdotal data as well as recent findings from both radiation accident survivors and animal experiments implicate radiation-induced injury or dysfunction of the vascular endothelium leading to tissue and organ injuries. There are significant gaps in our understanding of the disease processes and progression, as well as the optimum approaches to develop medical countermeasures to mitigate radiation vascular injury. To address this issue, the Radiation and Nuclear Countermeasures Program of the National Institute of Allergy and Infectious Diseases (NIAID) organized a one-day workshop to examine the current state of the science in radiation-induced vascular injuries and organ dysfunction, the natural history of the pathophysiology and the product development maturity of potential medical countermeasures to treat these injuries. Meeting presentations were followed by a NIAID-led open discussion among academic investigators, industry researchers and government agency representatives. This article provides a summary of these presentations and subsequent discussion from the workshop.
Collapse
Affiliation(s)
- Merriline M Satyamitra
- Division of Allergy Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852
| | - Andrea L DiCarlo
- Division of Allergy Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852
| | - Lanyn Taliaferro
- Division of Allergy Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852
| |
Collapse
|
510
|
Zewdu R, Risolino M, Barbulescu A, Ramalingam P, Butler JM, Selleri L. Spleen hypoplasia leads to abnormal stress hematopoiesis in mice with loss of Pbx homeoproteins in splenic mesenchyme. J Anat 2016; 229:153-69. [PMID: 27075259 PMCID: PMC5341595 DOI: 10.1111/joa.12479] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2016] [Indexed: 01/01/2023] Open
Abstract
The spleen plays critical roles in immunity and also provides a permissive microenvironment for hematopoiesis. Previous studies have reported that the TALE-class homeodomain transcription factor Pbx1 is essential in hematopoietic stem and progenitor cells (HSPCs) for stem cell maintenance and progenitor expansion. However, the role of Pbx1 in the hematopoietic niche has not been investigated. Here we explored the effects that genetic perturbation of the splenic mesenchymal niche has on hematopoiesis upon loss of members of the Pbx family of homeoproteins. Splenic mesenchyme-specific inactivation of Pbx1 (SKO) on a Pbx2- or Pbx3-deficient genetic background (DKO) resulted in abnormal development of the spleen, which is dysmorphic and severely hypoplastic. This phenotype, in turn, affected the number of HSPCs in the fetal and adult spleen at steady state, as well as markedly impairing the kinetics of hematopoietic regeneration in adult mice after sub-lethal and lethal myelosuppressive irradiation. Spleens of mice with compound Pyx deficiency 8 days following sublethal irradiation displayed significant downregulation of multiple cytokine-encoding genes, including KitL/SCF, Cxcl12/SDF-1, IL-3, IL-4, GM-CSF/Csf2 IL-10, and Igf-1, compared with controls. KitL/SCF and Cxcl12/SDF-1 were recently shown to play key roles in the splenic niche in response to various haematopoietic stresses such as myeloablation, blood loss, or pregnancy. Our results demonstrate that, in addition to their intrinsic roles in HSPCs, non-cell autonomous functions of Pbx factors within the splenic niche contribute to the regulation of hematopoiesis, at least in part via the control of KitL/SCF and Cxcl12/SDF-1. Furthermore, our study establishes that abnormal spleen development and hypoplasia have deleterious effects on the efficiency of hematopoietic recovery after bone marrow injury.
Collapse
Affiliation(s)
- Rediet Zewdu
- Department of Cell and Developmental BiologyWeill Cornell MedicineNew YorkNYUSA
- Present address: Huntsman Cancer Institute University of UtahSalt Lake CityUTUSA
| | - Maurizio Risolino
- Department of Cell and Developmental BiologyWeill Cornell MedicineNew YorkNYUSA
- Program in Craniofacial BiologyDepartment of Orofacial Sciences & Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | | | | | - Jason M. Butler
- Department of Genetic MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Licia Selleri
- Department of Cell and Developmental BiologyWeill Cornell MedicineNew YorkNYUSA
- Program in Craniofacial BiologyDepartment of Orofacial Sciences & Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| |
Collapse
|
511
|
Phosphoproteomic profiling of mouse primary HSPCs reveals new regulators of HSPC mobilization. Blood 2016; 128:1465-74. [PMID: 27365422 DOI: 10.1182/blood-2016-05-711424] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/28/2016] [Indexed: 12/14/2022] Open
Abstract
Protein phosphorylation is a central mechanism of signal transduction that both positively and negatively regulates protein function. Large-scale studies of the dynamic phosphorylation states of cell signaling systems have been applied extensively in cell lines and whole tissues to reveal critical regulatory networks, and candidate-based evaluations of phosphorylation in rare cell populations have also been informative. However, application of comprehensive profiling technologies to adult stem cell and progenitor populations has been challenging, due in large part to the scarcity of such cells in adult tissues. Here, we combine multicolor flow cytometry with highly efficient 3-dimensional high performance liquid chromatography/mass spectrometry to enable quantitative phosphoproteomic analysis from 200 000 highly purified primary mouse hematopoietic stem and progenitor cells (HSPCs). Using this platform, we identify ARHGAP25 as a novel regulator of HSPC mobilization and demonstrate that ARHGAP25 phosphorylation at serine 363 is an important modulator of its function. Our approach provides a robust platform for large-scale phosphoproteomic analyses performed with limited numbers of rare progenitor cells. Data from our study comprises a new resource for understanding the molecular signaling networks that underlie hematopoietic stem cell mobilization.
Collapse
|
512
|
|
513
|
Jalali S, Ansell SM. Bone marrow microenvironment in Waldenstrom's Macroglobulinemia. Best Pract Res Clin Haematol 2016; 29:148-155. [DOI: 10.1016/j.beha.2016.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/23/2016] [Indexed: 12/31/2022]
|
514
|
Abstract
PURPOSE OF REVIEW Cardiovascular disease is the leading cause of mortality worldwide. The underlying cause of the majority of cardiovascular disease is atherosclerosis. In the past, atherosclerosis was considered to be the result of passive lipid accumulation in the vessel wall. However, today's picture of the pathogenesis of atherosclerosis is much more complex, with a key role for immune cells and inflammation in conjunction with hyperlipidemia, especially elevated (modified) LDL levels. Knowledge on immune cells and immune responses in atherosclerosis has progressed tremendously over the past decades, and the same is true for the role of lipid metabolism and the different lipid components. However, it is largely unknown how lipids and the immune system interact. In this review, we will describe the effect of lipids on immune cell development and function, and the effects of immune cells on lipid metabolism. RECENT FINDINGS Recently, novel data have emerged that show that immune cells are affected, and behave differently in a hyperlipidemic environment. Moreover, immune cells have reported to be able to affect lipid metabolism. SUMMARY In this review, we will summarize the latest findings on the interactions between lipids and the immune system, and we will discuss the potential consequences of these novel insights for future therapies for atherosclerosis.
Collapse
Affiliation(s)
- Frank Schaftenaar
- aDivision of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden bDepartment of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands cInstitute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany
| | | | | | | |
Collapse
|
515
|
Regenerative approaches for the treatment of early OA. Knee Surg Sports Traumatol Arthrosc 2016; 24:1826-35. [PMID: 27120191 DOI: 10.1007/s00167-016-4125-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/29/2016] [Indexed: 01/08/2023]
Abstract
The diagnosis and the prompt treatment of early osteoarthritis (OA) represent vital steps for delaying the onset and progression of fully blown OA, which is the most common form of arthritis, involving more than 10 % of the world's population older than 60 years of age. Nonsurgical treatments such as physiotherapy, anti-inflammatory medications, and other disease-modifying drugs all have modest and short-lasting effect. In this context, the biological approaches have recently gained more and more attention. Growth factors, blood derivatives, such as platelet concentrates, and mesenchymal adult stem cells, either expanded or freshly isolated, are advocated amongst the most promising tool for the treatment of OA, especially in the early phases. Primarily targeted towards focal cartilage defects, these biological agents have indeed recently showed promising results to relieve pain and reduce inflammation in patients with more advanced OA as well, with the final aim to halt the progression of the disease and the need for joint replacement. However, despite of a number of satisfactory in vitro and pre-clinical studies, the evidences are still limited to support their clinical efficacy in OA setting.
Collapse
|
516
|
A humanized bone marrow ossicle xenotransplantation model enables improved engraftment of healthy and leukemic human hematopoietic cells. Nat Med 2016; 22:812-21. [PMID: 27213817 PMCID: PMC5549556 DOI: 10.1038/nm.4103] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 04/06/2016] [Indexed: 12/15/2022]
Abstract
Xenotransplantation models represent powerful tools for the investigation of healthy and malignant human hematopoiesis. However, current models do not fully mimic the components of the human bone marrow (BM) microenvironment, and they enable only limited engraftment of samples from some human malignancies. Here we show that a xenotransplantation model bearing subcutaneous humanized ossicles with an accessible BM microenvironment, formed by in situ differentiation of human BM-derived mesenchymal stromal cells, enables the robust engraftment of healthy human hematopoietic stem and progenitor cells, as well as primary acute myeloid leukemia (AML) samples, at levels much greater than those in unmanipulated mice. Direct intraossicle transplantation accelerated engraftment and resulted in the detection of substantially higher leukemia-initiating cell (LIC) frequencies. We also observed robust engraftment of acute promyelocytic leukemia (APL) and myelofibrosis (MF) samples, and identified LICs in these malignancies. This humanized ossicle xenotransplantation approach provides a system for modeling a wide variety of human hematological diseases.
Collapse
|
517
|
Chen JJ, Gao XT, Yang L, Fu W, Liang L, Li JC, Hu B, Sun ZJ, Huang SY, Zhang YZ, Liang YM, Qin HY, Han H. Disruption of Notch signaling aggravates irradiation-induced bone marrow injury, which is ameliorated by a soluble Dll1 ligand through Csf2rb2 upregulation. Sci Rep 2016; 6:26003. [PMID: 27188577 PMCID: PMC4870557 DOI: 10.1038/srep26003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/25/2016] [Indexed: 01/12/2023] Open
Abstract
Physical and chemical insult-induced bone marrow (BM) damage often leads to lethality resulting from the depletion of hematopoietic stem and progenitor cells (HSPCs) and/or a deteriorated BM stroma. Notch signaling plays an important role in hematopoiesis, but whether it is involved in BM damage remains unclear. In this study, we found that conditional disruption of RBP-J, the transcription factor of canonical Notch signaling, increased irradiation sensitivity in mice. Activation of Notch signaling with the endothelial cell (EC)-targeted soluble Dll1 Notch ligand mD1R promoted BM recovery after irradiation. mD1R treatment resulted in a significant increase in myeloid progenitors and monocytes in the BM, spleen and peripheral blood after irradiation. mD1R also enhanced hematopoiesis in mice treated with cyclophosphamide, a chemotherapeutic drug that induces BM suppression. Mechanistically, mD1R increased the proliferation and reduced the apoptosis of myeloid cells in the BM after irradiation. The β chain cytokine receptor Csf2rb2 was identified as a downstream molecule of Notch signaling in hematopoietic cells. mD1R improved hematopoietic recovery through up-regulation of the hematopoietic expression of Csf2rb2. Our findings reveal the role of Notch signaling in irradiation- and drug-induced BM suppression and establish a new potential therapy of BM- and myelo-suppression induced by radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Juan-Juan Chen
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.,Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiao-Tong Gao
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.,Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Lan Yang
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Fu
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.,Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Liang Liang
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Jun-Chang Li
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Bin Hu
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Zhi-Jian Sun
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Si-Yong Huang
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yi-Zhe Zhang
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Ying-Min Liang
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Hong-Yan Qin
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Hua Han
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.,Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
518
|
Nguyen H, Aum D, Mashkouri S, Rao G, Vega Gonzales-Portillo JD, Reyes S, Borlongan CV. Growth factor therapy sequesters inflammation in affording neuroprotection in cerebrovascular diseases. Expert Rev Neurother 2016; 16:915-26. [PMID: 27152762 DOI: 10.1080/14737175.2016.1184086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION In recent years, accumulating evidence has demonstrated the key role of inflammation in the progression of cerebrovascular diseases. Inflammation can persist over prolonged period of time after the initial insult providing a wider therapeutic window. Despite the acute endogenous upregulation of many growth factors after the injury, it is not sufficient to protect against inflammation and to regenerate the brain. Therapeutic approaches targeting both dampening inflammation and enhancing growth factors are likely to provide beneficial outcomes in cerebrovascular disease. AREAS COVERED In this mini review, we discuss major growth factors and their beneficial properties to combat the inflammation in cerebrovascular diseases. Emerging biotechnologies which facilitate the therapeutic effects of growth factors are also presented in an effort to provide insights into the future combination therapies incorporating both central and peripheral abrogation of inflammation. Expert commentary: Many studies discussed in this review have demonstrated the therapeutic effects of growth factors in treating cerebrovascular diseases. It is unlikely that one growth factor can be used to treat these complex diseases. Combination of growth factors and anti-inflammatory modulators may clinically improve outcomes for patients. In particular, transplantation of stem cells may be able to achieve both goals of modulating inflammation and upregulating growth factors. Large preclinical studies and multiple laboratory collaborations are needed to advance these findings from bench to bedside.
Collapse
Affiliation(s)
- Hung Nguyen
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - David Aum
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - Sherwin Mashkouri
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - Gautam Rao
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | | | - Stephanny Reyes
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - Cesario V Borlongan
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| |
Collapse
|
519
|
Oxidative stress and hypoxia in normal and leukemic stem cells. Exp Hematol 2016; 44:540-60. [PMID: 27179622 DOI: 10.1016/j.exphem.2016.04.012] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 12/20/2022]
Abstract
The main hematopoietic stem cell (HSC) functions, self-renewal and differentiation, are finely regulated by both intrinsic mechanisms such as transcriptional and epigenetic regulators and extrinsic signals originating in the bone marrow microenvironment (HSC niche) or in the body (humoral mediators). The interaction between regulatory signals and cellular metabolism is an emerging area. Several metabolic pathways function differently in HSCs compared with progenitors and differentiated cells. Hypoxia, acting through hypoxia-inducing factors, has emerged as a key regulator of stem cell biology and acts by maintaining HSC quiescence and a condition of metabolic dormancy based on anaerobic glycolytic energetic metabolism, with consequent low production reactive oxygen species (ROS) and high antioxidant defense. Hematopoietic cell differentiation is accompanied by changes in oxidative metabolism (decrease of anaerobic glycolysis and increase of oxidative phosphorylation) and increased levels of ROS. Leukemic stem cells, defined as the cells that initiate and maintain the leukemic process, show peculiar metabolic properties in that they are more dependent on oxidative respiration than on glycolysis and are more sensitive to oxidative stress than normal HSCs. Several mitochondrial abnormalities have been described in acute myeloid leukemia (AML) cells, explaining the shift to aerobic glycolysis observed in these cells and offering the unique opportunity for therapeutic metabolic targeting. Finally, frequent mutations of the mitochondrial isocitrate dehydrogenase-2 (IDH2) enzyme are observed in AML cells, in which the mutated enzyme acts as an oncogenic driver and can be targeted using specific inhibitors under clinical evaluation with promising results.
Collapse
|
520
|
Bernasconi P, Farina M, Boni M, Dambruoso I, Calvello C. Therapeutically targeting SELF-reinforcing leukemic niches in acute myeloid leukemia: A worthy endeavor? Am J Hematol 2016; 91:507-17. [PMID: 26822317 DOI: 10.1002/ajh.24312] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/21/2015] [Accepted: 01/16/2016] [Indexed: 12/17/2022]
Abstract
A tight relationship between the acute myeloid leukemia (AML) population and the bone marrow (BM) microenvironment has been convincingly established. The AML clone contains leukemic stem cells (LSCs) that compete with normal hematopoietic stem cells (HSCs) for niche occupancy and remodel the niche; whereas, the BM microenvironment might promote AML development and progression not only through hypoxia and homing/adhesion molecules, but also through genetic defects. Although it is still unknown whether the niche influences treatment results or contains any potential target for treatment, this dynamic AML-niche interaction might be a promising therapeutic objective to significantly improve the AML cure rate.
Collapse
Affiliation(s)
- Paolo Bernasconi
- Division of Hematology; Fondazione IRCCS Policlinico San Matteo, University of Pavia; Pavia Italy
| | - Mirko Farina
- Division of Hematology; Fondazione IRCCS Policlinico San Matteo, University of Pavia; Pavia Italy
| | - Marina Boni
- Division of Hematology; Fondazione IRCCS Policlinico San Matteo, University of Pavia; Pavia Italy
| | - Irene Dambruoso
- Division of Hematology; Fondazione IRCCS Policlinico San Matteo, University of Pavia; Pavia Italy
| | - Celeste Calvello
- Division of Hematology; Fondazione IRCCS Policlinico San Matteo, University of Pavia; Pavia Italy
| |
Collapse
|
521
|
Patenaude J, Perreault C. Thymic Mesenchymal Cells Have a Distinct Transcriptomic Profile. THE JOURNAL OF IMMUNOLOGY 2016; 196:4760-70. [DOI: 10.4049/jimmunol.1502499] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/28/2016] [Indexed: 12/22/2022]
|
522
|
Dhami SPS, Kappala SS, Thompson A, Szegezdi E. Three-dimensional ex vivo co-culture models of the leukaemic bone marrow niche for functional drug testing. Drug Discov Today 2016; 21:1464-1471. [PMID: 27130156 DOI: 10.1016/j.drudis.2016.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/04/2016] [Accepted: 04/20/2016] [Indexed: 10/21/2022]
Abstract
Acute myeloid leukaemia (AML) is a hierarchically structured malignancy in which aberrant leukemic stem cells drive the production of leukaemic blast cell clones. AML cells strictly depend on the bone marrow microenvironment (BMM) in which they reside. Classical AML cell cultures fail to mimic the BMM and, therefore, drug discovery studies are dominated by in vivo models. However, animal models are time consuming, labour intensive, provide limited mechanistic insight, and are unsuited for high-throughput studies, necessitating the development of novel AML models. The evolving ex vivo BMM-mimicking culture systems aim to fill this gap, with increasing success. Here, we discuss how AML-microenvironment co-culture models advance our understanding of this disease, and highlight their future potential for translational AML research.
Collapse
Affiliation(s)
- Sukhraj Pal S Dhami
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Shanthi S Kappala
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Alexander Thompson
- Centre for Cancer Research and Cell Biology, Queen's University, Belfast, United Kingdom
| | - Eva Szegezdi
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland.
| |
Collapse
|
523
|
Pietras EM, Mirantes-Barbeito C, Fong S, Loeffler D, Kovtonyuk LV, Zhang S, Lakshminarasimhan R, Chin CP, Techner JM, Will B, Nerlov C, Steidl U, Manz MG, Schroeder T, Passegué E. Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal. Nat Cell Biol 2016; 18:607-18. [PMID: 27111842 PMCID: PMC4884136 DOI: 10.1038/ncb3346] [Citation(s) in RCA: 504] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/24/2016] [Indexed: 02/08/2023]
Abstract
Haematopoietic stem cells (HSC) maintain lifelong blood production and increase blood cell numbers in response to chronic and acute injury. However, the mechanism(s) by which inflammatory insults are communicated to HSCs and their consequences for HSC activity remain largely unknown. Here, we demonstrate that interleukin-1 (IL-1), which functions as a key pro-inflammatory ‘emergency’ signal, directly accelerates cell division and myeloid differentiation of HSCs via precocious activation of a PU.1-dependent gene program. While this effect is essential for rapid myeloid recovery following acute injury to the bone marrow (BM), chronic IL-1 exposure restricts HSC lineage output, severely erodes HSC self-renewal capacity, and primes IL-1-exposed HSCs to fail massive replicative challenges like transplantation. Importantly, these damaging effects are transient and fully reversible upon IL-1 withdrawal. Our results identify a critical regulatory circuit that tailors HSC responses to acute needs, and likely underlies deregulated blood homeostasis in chronic inflammation conditions.
Collapse
Affiliation(s)
- Eric M Pietras
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California 94143, USA
| | - Cristina Mirantes-Barbeito
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California 94143, USA
| | - Sarah Fong
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California 94143, USA
| | - Dirk Loeffler
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Larisa V Kovtonyuk
- Division of Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - SiYi Zhang
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California 94143, USA
| | - Ranjani Lakshminarasimhan
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California 94143, USA
| | - Chih Peng Chin
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California 94143, USA
| | - José-Marc Techner
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California 94143, USA
| | - Britta Will
- Department of Cell Biology, Albert Einstein Medical College, Queens, New York 10461, USA
| | - Claus Nerlov
- Weatherall Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, UK
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein Medical College, Queens, New York 10461, USA
| | - Markus G Manz
- Division of Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Emmanuelle Passegué
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
524
|
Torisawa YS, Mammoto T, Jiang E, Jiang A, Mammoto A, Watters AL, Bahinski A, Ingber DE. Modeling Hematopoiesis and Responses to Radiation Countermeasures in a Bone Marrow-on-a-Chip. Tissue Eng Part C Methods 2016; 22:509-15. [PMID: 26993746 DOI: 10.1089/ten.tec.2015.0507] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Studies on hematopoiesis currently rely on animal models because in vitro culture methods do not accurately recapitulate complex bone marrow physiology. We recently described a bone marrow-on-a-chip microfluidic device that enables the culture of living hematopoietic bone marrow and mimics radiation toxicity in vitro. In the present study, we used this microdevice to demonstrate continuous blood cell production in vitro and model bone marrow responses to potential radiation countermeasure drugs. The device maintained mouse hematopoietic stem and progenitor cells in normal proportions for at least 2 weeks in culture. Increases in the number of leukocytes and red blood cells into the microfluidic circulation also could be detected over time, and addition of erythropoietin induced a significant increase in erythrocyte production. Exposure of the bone marrow chip to gamma radiation resulted in reduction of leukocyte production, and treatment of the chips with two potential therapeutics, granulocyte-colony stimulating factor or bactericidal/permeability-increasing protein (BPI), induced significant increases in the number of hematopoietic stem cells and myeloid cells in the fluidic outflow. In contrast, BPI was not found to have any effect when analyzed using static marrow cultures, even though it has been previously shown to accelerate recovery from radiation-induced toxicity in vivo. These findings demonstrate the potential value of the bone marrow-on-a-chip for modeling blood cell production, monitoring responses to hematopoiesis-modulating drugs, and testing radiation countermeasures in vitro.
Collapse
Affiliation(s)
- Yu-Suke Torisawa
- 1 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts.,2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Tadanori Mammoto
- 2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Elisabeth Jiang
- 2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Amanda Jiang
- 2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Akiko Mammoto
- 2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Alexander L Watters
- 1 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts
| | - Anthony Bahinski
- 1 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts
| | - Donald E Ingber
- 1 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts.,2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts.,3 School of Engineering and Applied Science, Harvard University , Cambridge, Massachusetts
| |
Collapse
|
525
|
Abstract
Adult or somatic stem cells are tissue-resident cells with the ability to proliferate, exhibit self-maintenance as well as to generate new cells with the principal phenotypes of the tissue in response to injury or disease. Due to their easy accessibility and their potential use in regenerative medicine, adult stem cells raise the hope for future personalisable therapies. After infection or during injury, they are exposed to broad range of pathogen or damage-associated molecules leading to changes in their proliferation, migration and differentiation. The sensing of such damage and infection signals is mostly achieved by Toll-Like Receptors (TLRs) with Toll-like receptor 4 being responsible for recognition of bacterial lipopolysaccharides (LPS) and endogenous danger-associated molecular patterns (DAMPs). In this review, we examine the current state of knowledge on the TLR4-mediated signalling in different adult stem cell populations. Specifically, we elaborate on the role of TLR4 and its ligands on proliferation, differentiation and migration of mesenchymal stem cells, hematopoietic stem cells as well as neural stem cells. Finally, we discuss conceptual and technical pitfalls in investigation of TLR4 signalling in stem cells.
Collapse
|
526
|
Tan MT, Hong Y, Han J, Jiang X. Expression of Hes1 during transdifferentiation of hUMSCs into islet progenitor cells. Shijie Huaren Xiaohua Zazhi 2016; 24:1357-1365. [DOI: 10.11569/wcjd.v24.i9.1357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To detect the expression of Hes1 during the transdifferentiation of human umbilical cord mesenchyreal stem cells (hUMSCs) into islet progenitor cells.
METHODS: After hUMSCs were isolated, cultivated and identified, hUMSCs at passage 5 were subjected staged induction to differentiate into islet precursor cells. Cell morphology was observed using an inverted phase contrast microscope. The expressions of insulin, neurogenin 3 (Ngn3) and glucagon after induction were detected by immunocytochemistry. The expression of Hes1 and Ngn3 was evaluated by immunocytochemistry and Western blot on 7 d, 14 d, and 21 d after induction.
RESULTS: After induction, HUMSCs became larger and colony-like, which is the characteristic of pancreatic progenitor cells. The expression of Ngn3, insulin and glucagon was positive. The level of Ngn3 increased gradually in the process of induction, peaked on 14 d (E2) and fell down on 21 d (E3). However, Hes1 remained unchanged from 7 d to 14 d, but was reduced on 21 d (E3).
CONCLUSION: The Notch signaling pathways' node molecule Hes1 may play an important role in the transdifferentiation of hUMSCs into islet progenitor cells.
Collapse
|
527
|
Birbrair A, Frenette PS. Niche heterogeneity in the bone marrow. Ann N Y Acad Sci 2016; 1370:82-96. [PMID: 27015419 DOI: 10.1111/nyas.13016] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/08/2016] [Accepted: 01/12/2016] [Indexed: 12/15/2022]
Abstract
In adult mammals, hematopoietic stem cells (HSCs) are defined by their abilities to self-renew and to differentiate to form all blood cell lineages. These rare multipotent cells occupy specific locations in the bone marrow (BM) microenvironment. The specific microenvironment regulating HSCs, commonly referred to as the niche, comprises multiple cell types whose exact contributions are under active investigation. Understanding cellular cross talk involving HSCs in the BM microenvironment is of fundamental importance for harnessing therapies against benign and malignant blood diseases. In this review, we summarize and evaluate recent advances in our understanding of niche heterogeneity and its influence on HSC function.
Collapse
Affiliation(s)
- Alexander Birbrair
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York.,Departments of Medicine and Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York.,Departments of Medicine and Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
528
|
Böhm AM, Dirckx N, Maes C. Recruitment of osteogenic cells to bone formation sites during development and fracture repair. Z Rheumatol 2016; 76:5-9. [PMID: 27001056 DOI: 10.1007/s00393-015-1574-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recruitment of osteoblast lineage cells to their bone-forming locations is essential for skeletal development and fracture healing. In developing bones, osteoprogenitor cells invade the cartilage mold to establish the primary ossification center. Similarly, osteogenic cells infiltrate and populate the callus tissue that is formed following an injury. Proper bone development and successful fracture repair must, therefore, rely on controlled temporal and spatial navigation cues guiding the cells to the sites where new bone formation is needed. Some cellular mechanisms and molecular pathways involved have been elucidated.
Collapse
Affiliation(s)
- A-M Böhm
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, Gasthuisberg O&N 1, KU Leuven, Herestraat 49, box 813, B-3000, Leuven, Belgium
| | - N Dirckx
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, Gasthuisberg O&N 1, KU Leuven, Herestraat 49, box 813, B-3000, Leuven, Belgium
| | - C Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, Gasthuisberg O&N 1, KU Leuven, Herestraat 49, box 813, B-3000, Leuven, Belgium.
| |
Collapse
|
529
|
Böhm AM, Dirckx N, Maes C. [Recruitment of osteogenic cells to bone formation sites during development and fracture repair - German Version]. Z Rheumatol 2016; 75:316-21. [PMID: 27003859 DOI: 10.1007/s00393-016-0065-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recruitment of osteoblast lineage cells to their bone-forming locations is essential for skeletal development and fracture healing. In developing bones, osteoprogenitor cells invade the cartilage mold to establish the primary ossification center. Similarly, osteogenic cells infiltrate and populate the callus tissue that is formed following an injury. Proper bone development and successful fracture repair must, therefore, rely on controlled temporal and spatial navigation cues guiding the cells to the sites where new bone formation is needed. Some cellular mechanisms and molecular pathways involved have been elucidated.
Collapse
Affiliation(s)
- A-M Böhm
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Gasthuisberg O&N 1, Herestraat 49, Box 813, 3000, Leuven, Belgien
| | - N Dirckx
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Gasthuisberg O&N 1, Herestraat 49, Box 813, 3000, Leuven, Belgien
| | - C Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Gasthuisberg O&N 1, Herestraat 49, Box 813, 3000, Leuven, Belgien.
| |
Collapse
|
530
|
Cong Q, Jia H, Biswas S, Li P, Qiu S, Deng Q, Guo X, Ma G, Ling Chau JF, Wang Y, Zhang ZL, Jiang X, Liu H, Li B. p38α MAPK Regulates Lineage Commitment and OPG Synthesis of Bone Marrow Stromal Cells to Prevent Bone Loss under Physiological and Pathological Conditions. Stem Cell Reports 2016; 6:566-578. [PMID: 26947973 PMCID: PMC4834033 DOI: 10.1016/j.stemcr.2016.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 02/01/2016] [Accepted: 02/01/2016] [Indexed: 01/06/2023] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are capable of differentiating into osteoblasts, chondrocytes, and adipocytes. Skewed differentiation of BM-MSCs contributes to the pathogenesis of osteoporosis. Yet how BM-MSC lineage commitment is regulated remains unclear. We show that ablation of p38α in Prx1+ BM-MSCs produced osteoporotic phenotypes, growth plate defects, and increased bone marrow fat, secondary to biased BM-MSC differentiation from osteoblast/chondrocyte to adipocyte and increased osteoclastogenesis and bone resorption. p38α regulates BM-MSC osteogenic commitment through TAK1-NF-κB signaling and osteoclastogenesis through osteoprotegerin (OPG) production by BM-MSCs. Estrogen activates p38α to maintain OPG expression in BM-MSCs to preserve the bone. Ablation of p38α in BM-MSCs positive for Dermo1, a later BM-MSC marker, only affected osteogenic differentiation. Thus, p38α mitogen-activated protein kinase (MAPK) in Prx1+ BM-MSCs acts to preserve the bone by promoting osteogenic lineage commitment and sustaining OPG production. This study thus unravels previously unidentified roles for p38α MAPK in skeletal development and bone remodeling. p38α deletion in Prx1+ BM-MSCs led to osteoporosis and cartilage anomaly p38α controls proliferation and tri-lineage differentiation of Prx1+ BM-MSCs p38α regulates osteoclastogenesis through OPG production by BM-MSCs The BM-MSC p38-OPG axis participates in estrogen deficiency-induced osteoporosis
Collapse
Affiliation(s)
- Qian Cong
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Jia
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China; Faculty of Basic Medicine, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Soma Biswas
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ping Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shoutao Qiu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Deng
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xizhi Guo
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Gang Ma
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | | | - Yibin Wang
- Division of Molecular Medicine, Departments of Anesthesiology, Medicine and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Zhen-Lin Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University affiliated the Sixth People's Hospital, Shanghai 200233, China
| | - Xinquan Jiang
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Research Institute of Stomatology, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | - Huijuan Liu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Baojie Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China; Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
531
|
Gur-Cohen S, Kollet O, Graf C, Esmon CT, Ruf W, Lapidot T. Regulation of long-term repopulating hematopoietic stem cells by EPCR/PAR1 signaling. Ann N Y Acad Sci 2016; 1370:65-81. [PMID: 26928241 DOI: 10.1111/nyas.13013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 01/18/2023]
Abstract
The common developmental origin of endothelial and hematopoietic cells is manifested by coexpression of several cell surface receptors. Adult murine bone marrow (BM) long-term repopulating hematopoietic stem cells (LT-HSCs), endowed with the highest repopulation and self-renewal potential, express endothelial protein C receptor (EPCR), which is used as a marker to isolate them. EPCR/protease-activated receptor-1 (PAR1) signaling in endothelial cells has anticoagulant and anti-inflammatory roles, while thrombin/PAR1 signaling induces coagulation and inflammation. Recent studies define two new PAR1-mediated signaling cascades that regulate EPCR(+) LT-HSC BM retention and egress. EPCR/PAR1 signaling facilitates LT-HSC BM repopulation, retention, survival, and chemotherapy resistance by restricting nitric oxide (NO) production, maintaining NO(low) LT-HSC BM retention with increased VLA4 expression, affinity, and adhesion. Conversely, acute stress and clinical mobilization upregulate thrombin generation and activate different PAR1 signaling that overcomes BM EPCR(+) LT-HSC retention, inducing their recruitment to the bloodstream. Thrombin/PAR1 signaling induces NO generation, TACE-mediated EPCR shedding, and upregulation of CXCR4 and PAR1, leading to CXCL12-mediated stem and progenitor cell mobilization. This review discusses new roles for factors traditionally viewed as coagulation related, which independently act in the BM to regulate PAR1 signaling in bone- and blood-forming progenitor cells, navigating their fate by controlling NO production.
Collapse
Affiliation(s)
- Shiri Gur-Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Orit Kollet
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Claudine Graf
- Center for Thrombosis and Hemostasis and Johannes Gutenberg University Medical Center, Mainz, Germany.,Third Medical Department, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Charles T Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation and Departments of Pathology and Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis and Johannes Gutenberg University Medical Center, Mainz, Germany.,Department of Immunology and Microbial Science, the Scripps Research Institute, La Jolla, California
| | - Tsvee Lapidot
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
532
|
Choi JS, Harley BAC. Challenges and Opportunities to Harnessing the (Hematopoietic) Stem Cell Niche. CURRENT STEM CELL REPORTS 2016; 2:85-94. [PMID: 27134819 PMCID: PMC4845958 DOI: 10.1007/s40778-016-0031-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In our body, stem cells reside in a microenvironment termed the niche. While the exact composition and therefore the level of complexity of a stem cell niche can vary significantly tissue-to-tissue, the stem cell niche microenvironment is dynamic, typically containing spatial and temporal variations in both cellular, extracellular matrix, and biomolecular components. This complex flow of secreted or bound biomolecules, cytokines, extracellular matrix components, and cellular constituents all contribute to the regulation of stem cell fate specification events, making engineering approaches at the nano- and micro-scale of particular interest for creating an artificial niche environment in vitro. Recent advances in fabrication approaches have enabled biomedical researchers to capture and recreate the complexity of stem cell niche microenvironments in vitro. Such engineered platforms show promise as a means to enhance our understanding of the mechanisms underlying niche-mediated stem cell regulation as well as offer opportunities to precisely control stem cell expansion and differentiation events for clinical applications. While these principles generally apply to all adult stem cells and niches, in this review, we focus on recent developments in engineering synthetic niche microenvironments for one of the best-characterized stem cell populations, hematopoietic stem cells (HSC). Specifically, we highlight recent advances in platforms designed to facilitate the extrinsic control of HSC fate decisions.
Collapse
Affiliation(s)
- Ji Sun Choi
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
533
|
Maeda K, Enomoto A, Hara A, Asai N, Kobayashi T, Horinouchi A, Maruyama S, Ishikawa Y, Nishiyama T, Kiyoi H, Kato T, Ando K, Weng L, Mii S, Asai M, Mizutani Y, Watanabe O, Hirooka Y, Goto H, Takahashi M. Identification of Meflin as a Potential Marker for Mesenchymal Stromal Cells. Sci Rep 2016; 6:22288. [PMID: 26924503 PMCID: PMC4770287 DOI: 10.1038/srep22288] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 02/11/2016] [Indexed: 01/14/2023] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) in culture are derived from BM stromal cells or skeletal stem cells. Whereas MSCs have been exploited in clinical medicine, the identification of MSC-specific markers has been limited. Here, we report that a cell surface and secreted protein, Meflin, is expressed in cultured MSCs, fibroblasts and pericytes, but not other types of cells including epithelial, endothelial and smooth muscle cells. In vivo, Meflin is expressed by immature osteoblasts and chondroblasts. In addition, Meflin is found on stromal cells distributed throughout the BM, and on pericytes and perivascular cells in multiple organs. Meflin maintains the undifferentiated state of cultured MSCs and is downregulated upon their differentiation, consistent with the observation that Meflin-deficient mice exhibit increased number of osteoblasts and accelerated bone development. In the bone and BM, Meflin is more highly expressed in primitive stromal cells that express platelet-derived growth factor receptor α and Sca-1 than the Sca-1-negative adipo-osteogenic progenitors, which create a niche for hematopoiesis. Those results are consistent with a decrease in the number of clonogenic colony-forming unit-fibroblasts within the BM of Meflin-deficient mice. These preliminary data suggest that Meflin is a potential marker for cultured MSCs and their source cells in vivo.
Collapse
Affiliation(s)
- Keiko Maeda
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.,Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Atsushi Enomoto
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akitoshi Hara
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Naoya Asai
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takeshi Kobayashi
- Department of Physiology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Asuka Horinouchi
- Department of Nephrology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shoichi Maruyama
- Department of Nephrology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takahiro Nishiyama
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takuya Kato
- Tumour Cell Biology Laboratory, The Francis-Crick Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, United Kingdom
| | - Kenju Ando
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Liang Weng
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shinji Mii
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masato Asai
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yasuyuki Mizutani
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.,Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Osamu Watanabe
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshiki Hirooka
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hidemi Goto
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masahide Takahashi
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
534
|
Sympathetic stimulation facilitates thrombopoiesis by promoting megakaryocyte adhesion, migration, and proplatelet formation. Blood 2016; 127:1024-35. [DOI: 10.1182/blood-2015-07-660746] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/28/2015] [Indexed: 12/22/2022] Open
Abstract
Key Points
NE and EPI promote megakaryocyte adhesion, migration, and proplatelet formation via α2-adrenoceptor-ERK1/2 signaling. Sympathetic stimulation enhances platelet production, which may facilitate recovery of thrombocytopenia or aggravate atherosclerosis.
Collapse
|
535
|
Crisan M, Solaimani Kartalaei P, Neagu A, Karkanpouna S, Yamada-Inagawa T, Purini C, Vink CS, van der Linden R, van Ijcken W, Chuva de Sousa Lopes SM, Monteiro R, Mummery C, Dzierzak E. BMP and Hedgehog Regulate Distinct AGM Hematopoietic Stem Cells Ex Vivo. Stem Cell Reports 2016; 6:383-95. [PMID: 26923823 PMCID: PMC4788785 DOI: 10.1016/j.stemcr.2016.01.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 11/19/2022] Open
Abstract
Hematopoietic stem cells (HSC), the self-renewing cells of the adult blood differentiation hierarchy, are generated during embryonic stages. The first HSCs are produced in the aorta-gonad-mesonephros (AGM) region of the embryo through endothelial to a hematopoietic transition. BMP4 and Hedgehog affect their production and expansion, but it is unknown whether they act to affect the same HSCs. In this study using the BRE GFP reporter mouse strain that identifies BMP/Smad-activated cells, we find that the AGM harbors two types of adult-repopulating HSCs upon explant culture: One type is BMP-activated and the other is a non-BMP-activated HSC type that is indirectly controlled by Hedgehog signaling through the VEGF pathway. Transcriptomic analyses demonstrate that the two HSC types express distinct but overlapping genetic programs. These results revealing the bifurcation in HSC types at early embryonic stages in the AGM explant model suggest that their development is dependent upon the signaling molecules in the microenvironment. AGM explants contain two HSC types, BMP-activated and non-BMP-activated Non-BMP-activated HSCs are dependent on Hedgehog/VEGF Changes in the microenvironment ex vivo contribute to novel HSC composition
Collapse
Affiliation(s)
- Mihaela Crisan
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; BHF Centre for Cardiovascular Science, Scottish Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Parham Solaimani Kartalaei
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Alex Neagu
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Sofia Karkanpouna
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Tomoko Yamada-Inagawa
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Caterina Purini
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Chris S Vink
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Reinier van der Linden
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands
| | - Wilfred van Ijcken
- Center for Biomics, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | | | - Rui Monteiro
- Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Elaine Dzierzak
- Department of Cell Biology, Erasmus Medical Center, Erasmus MC Stem Cell Institute, 3000 CA Rotterdam, the Netherlands; Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
536
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells can self-renew and also give rise to the entire repertoire of hematopoietic cells. During acute infectious and inflammatory stresses, the hematopoietic system can quickly adapt to demand by increasing output of innate immune cells many-fold, often at the expense of lymphopoiesis and erythropoiesis. We review recent advances in understanding the regulation of stress-induced hematopoiesis with a specific focus on the direct effects of inflammatory signaling on hematopoietic stem and progenitor cells (HSPCs). RECENT FINDINGS Recent studies have highlighted several areas of exciting new developments in the field, including the complex interaction and crosstalk within HSPCs and between bone marrow mesenchymal stem cells and endothelial cells needed to achieve regulated myelopoiesis, identification of increased number of inflammatory and infectious molecules with direct effects on HSPCs, the critical role of inflammatory signaling on embryonic specification of hematopoietic stem cells, and the ability of cytokines to instruct lineage choice at the HSPC level. SUMMARY These exciting new findings will shape our fundamental understanding of how inflammatory signaling regulates hematopoiesis in health and disease, and facilitate the development of potential interventions to treat hematologic diseases associated with altered inflammatory signaling.
Collapse
|
537
|
Abstract
As part of the dynamic interactions between leukemic cells and cells of the bone marrow microenvironment, specific niches provide a sanctuary where subpopulations of leukemic cells evade chemotherapy-induced death and acquire a drug-resistant phenotype. This review focuses on the cellular and molecular biology of the leukemia stem cell (LSC) niche and of microenvironment/leukemia interactions. Key emerging therapeutic targets include chemokine receptors, adhesion molecules, the sympathetic nervous system, and hypoxia-related proteins, as well as the genetic and epigenetic abnormalities of the leukemia-associated stroma. The complex interplay between LSCs and microenvironment components provides a rationale for appropriately tailored molecular therapies designed to improve outcomes in leukemia. Further understanding of the contribution of the bone marrow niche to the process of leukemogenesis may provide new targets that allow destruction of LSCs without adversely affecting normal stem cell self-renewal.
Collapse
Affiliation(s)
- Yoko Tabe
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Unit 428 Holcombe Blvd 1515, Houston, TX, 77030, USA,
| | | |
Collapse
|
538
|
Klein C, Zwick A, Kissel S, Forster CU, Pfeifer D, Follo M, Illert AL, Decker S, Benkler T, Pahl H, Oostendorp RAJ, Aumann K, Duyster J, Dierks C. Ptch2 loss drives myeloproliferation and myeloproliferative neoplasm progression. J Exp Med 2016; 213:273-90. [PMID: 26834157 PMCID: PMC4749921 DOI: 10.1084/jem.20150556] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 12/21/2015] [Indexed: 01/12/2023] Open
Abstract
Klein et al. show that Ptch2 loss in either the niche or in hematopoietic cells drives myeloproliferation and accelerates JAK2V617F-driven pathogenesis, causing transformation of nonlethal chronic MPNs into aggressive lethal leukemias. JAK2V617F+ myeloproliferative neoplasms (MPNs) frequently progress into leukemias, but the factors driving this process are not understood. Here, we find excess Hedgehog (HH) ligand secretion and loss of PTCH2 in myeloproliferative disease, which drives canonical and noncanonical HH-signaling. Interestingly, Ptch2−/− mice mimic dual pathway activation and develop a MPN-phenotype with leukocytosis (neutrophils and monocytes), strong progenitor and LKS mobilization, splenomegaly, anemia, and loss of lymphoid lineages. HSCs exhibit increased cell cycling with improved stress hematopoiesis after 5-FU treatment, and this results in HSC exhaustion over time. Cytopenias, LKS loss, and mobilization are all caused by loss of Ptch2 in the niche, whereas hematopoietic loss of Ptch2 drives leukocytosis and promotes LKS maintenance and replating capacity in vitro. Ptch2−/− niche cells show hyperactive noncanonical HH signaling, resulting in reduced production of essential HSC regulators (Scf, Cxcl12, and Jag1) and depletion of osteoblasts. Interestingly, Ptch2 loss in either the niche or in hematopoietic cells dramatically accelerated human JAK2V617F-driven pathogenesis, causing transformation of nonlethal chronic MPNs into aggressive lethal leukemias with >30% blasts in the peripheral blood. Our findings suggest HH ligand inhibitors as possible drug candidates that act on hematopoiesis and the niche to prevent transformation of MPNs into leukemias.
Collapse
Affiliation(s)
- Claudius Klein
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany Institute for Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - Anabel Zwick
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Sandra Kissel
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Christine Ulrike Forster
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Marie Follo
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Anna Lena Illert
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Sarah Decker
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Thomas Benkler
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Heike Pahl
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Robert A J Oostendorp
- Klinikum rechts der Isar der Technischen Universität München, III. Medizinische Klinik, 81675 München, Germany
| | - Konrad Aumann
- Department of Pathology, University Medical Center Freiburg, D-79104 Freiburg, Germany
| | - Justus Duyster
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Christine Dierks
- Department of Hematology/Oncology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| |
Collapse
|
539
|
Abbonante V, Di Buduo CA, Gruppi C, Malara A, Gianelli U, Celesti G, Anselmo A, Laghi L, Vercellino M, Visai L, Iurlo A, Moratti R, Barosi G, Rosti V, Balduini A. Thrombopoietin/TGF-β1 Loop Regulates Megakaryocyte Extracellular Matrix Component Synthesis. Stem Cells 2016; 34:1123-33. [PMID: 26748484 DOI: 10.1002/stem.2285] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/13/2015] [Accepted: 12/01/2015] [Indexed: 11/11/2022]
Abstract
Extracellular matrix (ECM) components initiate crucial biochemical and biomechanical cues that are required for bone marrow homeostasis. In our research, we prove that a peri-cellular matrix composed primarily of type III and type IV collagens, and fibronectin surrounds human megakaryocytes in the bone marrow. The data we collected support the hypothesis that bone marrow megakaryocytes possess a complete mechanism to synthesize the ECM components, and that thrombopoietin is a pivotal regulator of this new function inducing transforming growth factor-β1 (TGF-β1) release and consequent activation of the downstream pathways, both in vitro and in vivo. This activation results in a dose dependent increase of ECM component synthesis by megakaryocytes, which is reverted upon incubation with JAK and TGF-β1 receptor specific inhibitors. These data are pivotal for understanding the central role of megakaryocytes in creating their own regulatory niche within the bone marrow environment.
Collapse
Affiliation(s)
- Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Christian A Di Buduo
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Cristian Gruppi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Umberto Gianelli
- Hematopathology Service, Division of Pathology, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giuseppe Celesti
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Achille Anselmo
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Marco Vercellino
- Center for Tissue Engineering (CIT), INSTM UdR of Pavia, University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Ergonomics and Disability, Salvatore Maugeri Foundation (FSM), Laboratory of Nanotechnology, Pavia, Italy
| | - Livia Visai
- Center for Tissue Engineering (CIT), INSTM UdR of Pavia, University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Ergonomics and Disability, Salvatore Maugeri Foundation (FSM), Laboratory of Nanotechnology, Pavia, Italy
| | - Alessandra Iurlo
- Oncohematology of the Elderly Unit, Oncohematology Division, IRCCS Ca' Granda-Maggiore Policlinico Hospital Foundation, Milan, Italy
| | - Remigio Moratti
- Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Giovanni Barosi
- Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Vittorio Rosti
- Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy.,Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
540
|
van den Berg SM, Seijkens TTP, Kusters PJH, Beckers L, den Toom M, Smeets E, Levels J, de Winther MPJ, Lutgens E. Diet-induced obesity in mice diminishes hematopoietic stem and progenitor cells in the bone marrow. FASEB J 2016; 30:1779-88. [PMID: 26813974 DOI: 10.1096/fj.201500175] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/22/2015] [Indexed: 12/20/2022]
Abstract
Obesity is associated with chronic low-grade inflammation, characterized by leukocytosis and inflammation in the adipose tissue. Continuous activation of the immune system is a stressor for hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM). Here we studied how diet-induced obesity (DIO) affects HSPC population dynamics in the BM. Eight groups of age-matched C57Bl/6 mice received a high-fat diet (45% kilocalories from fat) ranging from 1 d up to 18 wk. The obesogenic diet caused decreased proliferation of lineage(-)Sca-1(+)c-Kit(+) (LSK) cells in the BM and a general suppression of progenitor cell populations including common lymphoid progenitors and common myeloid progenitors. Within the LSK population, DIO induced a shift in stem cells that are capable of self-renewal toward maturing multipotent progenitor cells. The higher differentiation potential resulted in increased lymphoid and myeloid ex vivo colony-forming capacity. In a competitive BM transplantation, BM from obese animals showed impaired multilineage reconstitution when transplanted into chow-fed mice. Our data demonstrate that obesity stimulates the differentiation and reduces proliferation of HSPCs in the BM, leading to a decreased HSPC population. This implies that the effects of obesity on HSPCs hampers proper functioning of the immune system.-Van den Berg, S. M., Seijkens, T. T. P., Kusters, P. J. H., Beckers, L., den Toom, M., Smeets, E., Levels, J., de Winther, M. P. J., Lutgens, E. Diet-induced obesity in mice diminishes hematopoietic stem and progenitor cells in the bone marrow.
Collapse
Affiliation(s)
- Susan M van den Berg
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom T P Seijkens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Pascal J H Kusters
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther Smeets
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Johannes Levels
- Department of Experimental Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Menno P J de Winther
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands; Institute for Cardiovascular Prevention, Ludwig Maximillians University, Munich, Germany
| |
Collapse
|
541
|
Kang HM, Huang S, Reidy K, Han SH, Chinga F, Susztak K. Sox9-Positive Progenitor Cells Play a Key Role in Renal Tubule Epithelial Regeneration in Mice. Cell Rep 2016; 14:861-871. [PMID: 26776520 DOI: 10.1016/j.celrep.2015.12.071] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 10/02/2015] [Accepted: 12/14/2015] [Indexed: 01/01/2023] Open
Abstract
The kidney has a tremendous capacity to regenerate following injury, but factors that govern this response are still largely unknown. We isolated cells from mouse kidneys with high proliferative and multi-lineage differentiation capacity. These cells expressed a high level of Sox9. In regenerating kidneys, Sox9 expression was induced early, and 89% of proliferating cells were Sox9 positive. In vitro, Sox9-positive cells showed unlimited proliferation and multi-lineage differentiation capacity. Using an inducible Sox9 Cre line and lineage-tagging methods, we show that Sox9-positive cells can generate new daughter cells, contributing to the regeneration of proximal tubule, loop of Henle, and distal tubule segments but not to collecting duct and glomerular cells. Furthermore, inducible deletion of Sox9 resulted in reduced epithelial proliferation, more severe injury, and fibrosis development. In summary, we demonstrate that, in the kidney, Sox9-positive cells show progenitor-like properties in vitro and contribute to epithelial regeneration following injury in vivo.
Collapse
Affiliation(s)
- Hyun Mi Kang
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shizheng Huang
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kimberly Reidy
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seung Hyeok Han
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frank Chinga
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
542
|
Affiliation(s)
- Nina Cabezas-Wallscheid
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
543
|
Moses BS, Slone WL, Thomas P, Evans R, Piktel D, Angel PM, Walsh CM, Cantrell PS, Rellick SL, Martin KH, Simpkins JW, Gibson LF. Bone marrow microenvironment modulation of acute lymphoblastic leukemia phenotype. Exp Hematol 2016; 44:50-9.e1-2. [PMID: 26407636 PMCID: PMC4684957 DOI: 10.1016/j.exphem.2015.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/11/2015] [Accepted: 09/12/2015] [Indexed: 01/25/2023]
Abstract
Acute lymphoblastic leukemia (ALL) treatment regimens have dramatically improved the survival of ALL patients. However, chemoresistant minimal residual disease that persists following cessation of therapy contributes to aggressive relapse. The bone marrow microenvironment (BMM) is an established "site of sanctuary" for ALL, as well as myeloid-lineage hematopoietic disease, with signals in this unique anatomic location contributing to drug resistance. Several models have been developed to recapitulate the interactions between the BMM and ALL cells. However, many in vitro models fail to accurately reflect the level of protection afforded to the most resistant subset of leukemic cells during coculture with BMM elements. Preclinical in vivo models have advantages, but can be costly, and are often not fully informed by optimal in vitro studies. We describe an innovative extension of 2-D coculture wherein ALL cells uniquely interact with bone marrow-derived stromal cells. Tumor cells in this model bury beneath primary human bone marrow-derived stromal cells or osteoblasts, termed "phase dim" ALL, and exhibit a unique phenotype characterized by altered metabolism, distinct protein expression profiles, increased quiescence, and pronounced chemotherapy resistance. Investigation focused on the phase dim subpopulation may more efficiently inform preclinical design and investigation of the minimal residual disease and relapse that arise from BMM-supported leukemic tumor cells.
Collapse
Affiliation(s)
- Blake S Moses
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV
| | - William L Slone
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV
| | - Patrick Thomas
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV
| | - Rebecca Evans
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV
| | - Debbie Piktel
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV
| | | | | | | | - Stephanie L Rellick
- Department of Physiology & Pharmacology, West Virginia University School of Medicine, Morgantown, WV
| | - Karen H Martin
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV; Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, WV
| | - James W Simpkins
- Department of Physiology & Pharmacology, West Virginia University School of Medicine, Morgantown, WV; Center for Basic and Translational Stroke Research, West Virginia University School of Medicine, Morgantown, WV; Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV
| | - Laura F Gibson
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV; Department of Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morganstown, WV.
| |
Collapse
|
544
|
Wei Y, Xu J, Zhang W, Wen Z, Liu F. RNA polymerase III component Rpc9 regulates hematopoietic stem and progenitor cell maintenance in zebrafish. Development 2016; 143:2103-10. [DOI: 10.1242/dev.126797] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 04/25/2016] [Indexed: 12/18/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are capable of self-renewal and replenishing all lineages of blood cells throughout the lifetime and thus critical for tissue homeostasis. However, the mechanism regulating HSPC development is still incompletely understood. Here, we isolate a zebrafish mutant with defective T lymphopoiesis and positional cloning identifies that Rpc9, a component of DNA-directed RNA polymerase III (Pol III) complex, is responsible for the mutant phenotype. Further analysis shows that rpc9-deficiency leads to the impairment of HSPCs and their derivatives in zebrafish embryos. Excessive apoptosis is observed in the caudal hematopoietic tissue (CHT, the equivalent of fetal liver in mammals) of rpc9−/− embryos and the hematopoietic defects in rpc9−/− embryos can be fully rescued by suppression of p53. Thus, our work illustrate that Rpc9, a component of Pol III, plays an important tissue-specific role in HSPC maintenance during zebrafish embryogenesis and that it might be conserved across vertebrates including mammals.
Collapse
Affiliation(s)
- Yonglong Wei
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Xu
- State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, Division of Life Science, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Wenqing Zhang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Cell Biology, Southern Medical University, Guangzhou 510515, China
| | - Zilong Wen
- State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, Division of Life Science, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
545
|
Philipp-Abbrederis K, Herrmann K, Knop S, Schottelius M, Eiber M, Lückerath K, Pietschmann E, Habringer S, Gerngroß C, Franke K, Rudelius M, Schirbel A, Lapa C, Schwamborn K, Steidle S, Hartmann E, Rosenwald A, Kropf S, Beer AJ, Peschel C, Einsele H, Buck AK, Schwaiger M, Götze K, Wester HJ, Keller U. In vivo molecular imaging of chemokine receptor CXCR4 expression in patients with advanced multiple myeloma. EMBO Mol Med 2015; 7:477-87. [PMID: 25736399 PMCID: PMC4403048 DOI: 10.15252/emmm.201404698] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
CXCR4 is a G-protein-coupled receptor that mediates recruitment of blood cells toward its ligand SDF-1. In cancer, high CXCR4 expression is frequently associated with tumor dissemination and poor prognosis. We evaluated the novel CXCR4 probe [(68)Ga]Pentixafor for in vivo mapping of CXCR4 expression density in mice xenografted with human CXCR4-positive MM cell lines and patients with advanced MM by means of positron emission tomography (PET). [(68)Ga]Pentixafor PET provided images with excellent specificity and contrast. In 10 of 14 patients with advanced MM [(68)Ga]Pentixafor PET/CT scans revealed MM manifestations, whereas only nine of 14 standard [(18)F]fluorodeoxyglucose PET/CT scans were rated visually positive. Assessment of blood counts and standard CD34(+) flow cytometry did not reveal significant blood count changes associated with tracer application. Based on these highly encouraging data on clinical PET imaging of CXCR4 expression in a cohort of MM patients, we conclude that [(68)Ga]Pentixafor PET opens a broad field for clinical investigations on CXCR4 expression and for CXCR4-directed therapeutic approaches in MM and other diseases.
Collapse
Affiliation(s)
- Kathrin Philipp-Abbrederis
- III. Medical Department of Hematology and Medical Oncology, Technische Universität München, Munich, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Stefan Knop
- Department of Internal Medicine II, Division of Hematology and Medical Oncology, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Margret Schottelius
- Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Katharina Lückerath
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Elke Pietschmann
- III. Medical Department of Hematology and Medical Oncology, Technische Universität München, Munich, Germany
| | - Stefan Habringer
- III. Medical Department of Hematology and Medical Oncology, Technische Universität München, Munich, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carlos Gerngroß
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Katharina Franke
- III. Medical Department of Hematology and Medical Oncology, Technische Universität München, Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Universitätsklinikum Würzburg and CCC Mainfranken, Würzburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | | | - Sabine Steidle
- III. Medical Department of Hematology and Medical Oncology, Technische Universität München, Munich, Germany
| | - Elena Hartmann
- Institute of Pathology, Universitätsklinikum Würzburg and CCC Mainfranken, Würzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Universitätsklinikum Würzburg and CCC Mainfranken, Würzburg, Germany
| | | | - Ambros J Beer
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Christian Peschel
- III. Medical Department of Hematology and Medical Oncology, Technische Universität München, Munich, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, Division of Hematology and Medical Oncology, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katharina Götze
- III. Medical Department of Hematology and Medical Oncology, Technische Universität München, Munich, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany Scintomics GmbH, Fürstenfeldbruck, Germany
| | - Ulrich Keller
- III. Medical Department of Hematology and Medical Oncology, Technische Universität München, Munich, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
546
|
Genetic and Epigenetic Mechanisms That Maintain Hematopoietic Stem Cell Function. Stem Cells Int 2015; 2016:5178965. [PMID: 26798358 PMCID: PMC4699043 DOI: 10.1155/2016/5178965] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/03/2015] [Accepted: 09/09/2015] [Indexed: 01/15/2023] Open
Abstract
All hematopoiesis cells develop from multipotent progenitor cells. Hematopoietic stem cells (HSC) have the ability to develop into all blood lineages but also maintain their stemness. Different molecular mechanisms have been identified that are crucial for regulating quiescence and self-renewal to maintain the stem cell pool and for inducing proliferation and lineage differentiation. The stem cell niche provides the microenvironment to keep HSC in a quiescent state. Furthermore, several transcription factors and epigenetic modifiers are involved in this process. These create modifications that regulate the cell fate in a more or less reversible and dynamic way and contribute to HSC homeostasis. In addition, HSC respond in a unique way to DNA damage. These mechanisms also contribute to the regulation of HSC function and are essential to ensure viability after DNA damage. How HSC maintain their quiescent stage during the entire life is still matter of ongoing research. Here we will focus on the molecular mechanisms that regulate HSC function.
Collapse
|
547
|
Dhawan J, Laxman S. Decoding the stem cell quiescence cycle--lessons from yeast for regenerative biology. J Cell Sci 2015; 128:4467-74. [PMID: 26672015 PMCID: PMC5695657 DOI: 10.1242/jcs.177758] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In the past decade, major advances have occurred in the understanding of mammalian stem cell biology, but roadblocks (including gaps in our fundamental understanding) remain in translating this knowledge to regenerative medicine. Interestingly, a close analysis of the Saccharomyces cerevisiae literature leads to an appreciation of how much yeast biology has contributed to the conceptual framework underpinning our understanding of stem cell behavior, to the point where such insights have been internalized into the realm of the known. This Opinion article focuses on one such example, the quiescent adult mammalian stem cell, and examines concepts underlying our understanding of quiescence that can be attributed to studies in yeast. We discuss the metabolic, signaling and gene regulatory events that control entry and exit into quiescence in yeast. These processes and events retain remarkable conservation and conceptual parallels in mammalian systems, and collectively suggest a regulated program beyond the cessation of cell division. We argue that studies in yeast will continue to not only reveal fundamental concepts in quiescence, but also leaven progress in regenerative medicine.
Collapse
Affiliation(s)
- Jyotsna Dhawan
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India CSIR Center for Cellular and Molecular Biology, Hyderabad, India
| | - Sunil Laxman
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| |
Collapse
|
548
|
Hayakawa Y, Ariyama H, Stancikova J, Sakitani K, Asfaha S, Renz BW, Dubeykovskaya ZA, Shibata W, Wang H, Westphalen CB, Chen X, Takemoto Y, Kim W, Khurana SS, Tailor Y, Nagar K, Tomita H, Hara A, Sepulveda AR, Setlik W, Gershon MD, Saha S, Ding L, Shen Z, Fox JG, Friedman RA, Konieczny SF, Worthley DL, Korinek V, Wang TC. Mist1 Expressing Gastric Stem Cells Maintain the Normal and Neoplastic Gastric Epithelium and Are Supported by a Perivascular Stem Cell Niche. Cancer Cell 2015; 28:800-814. [PMID: 26585400 PMCID: PMC4684751 DOI: 10.1016/j.ccell.2015.10.003] [Citation(s) in RCA: 244] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/26/2015] [Accepted: 10/08/2015] [Indexed: 12/12/2022]
Abstract
The regulation and stem cell origin of normal and neoplastic gastric glands are uncertain. Here, we show that Mist1 expression marks quiescent stem cells in the gastric corpus isthmus. Mist1(+) stem cells serve as a cell-of-origin for intestinal-type cancer with the combination of Kras and Apc mutation and for diffuse-type cancer with the loss of E-cadherin. Diffuse-type cancer development is dependent on inflammation mediated by Cxcl12(+) endothelial cells and Cxcr4(+) gastric innate lymphoid cells (ILCs). These cells form the perivascular gastric stem cell niche, and Wnt5a produced from ILCs activates RhoA to inhibit anoikis in the E-cadherin-depleted cells. Targeting Cxcr4, ILCs, or Wnt5a inhibits diffuse-type gastric carcinogenesis, providing targets within the neoplastic gastric stem cell niche.
Collapse
Affiliation(s)
- Yoku Hayakawa
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Hiroshi Ariyama
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jitka Stancikova
- Department of Cell and Developmental Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague 14220, Czech Republic
| | - Kosuke Sakitani
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Samuel Asfaha
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Bernhard W Renz
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA; Department of General, Visceral, Transplantation, Vascular, and Thoracic Surgery, Hospital of the University of Munich, Munich 81377, Germany
| | - Zinaida A Dubeykovskaya
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Wataru Shibata
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Hongshan Wang
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Christoph B Westphalen
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Xiaowei Chen
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yoshihiro Takemoto
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Woosook Kim
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Shradha S Khurana
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yagnesh Tailor
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Karan Nagar
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Antonia R Sepulveda
- Division of Clinical Pathology and Cell Biology, Department of Pathology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Wanda Setlik
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Michael D Gershon
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Subhrajit Saha
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Lei Ding
- Departments of Rehabilitation and Regenerative Medicine and Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Zeli Shen
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard A Friedman
- Herbert Irving Comprehensive Cancer Center Biomedical Informatics Shared Resource and Department of Biomedical Informatics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Stephen F Konieczny
- Department of Biological Sciences and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Daniel L Worthley
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Vladimir Korinek
- Department of Cell and Developmental Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague 14220, Czech Republic
| | - Timothy C Wang
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
549
|
Wang Z, Ema H. Mechanisms of self-renewal in hematopoietic stem cells. Int J Hematol 2015; 103:498-509. [DOI: 10.1007/s12185-015-1919-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 11/30/2015] [Indexed: 11/29/2022]
|
550
|
Plasmacytoid dendritic cells in allogeneic hematopoietic cell transplantation: benefit or burden? Bone Marrow Transplant 2015; 51:333-43. [PMID: 26642333 DOI: 10.1038/bmt.2015.301] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/25/2015] [Accepted: 10/27/2015] [Indexed: 11/09/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and have important roles in hematopoietic engraftment, GvHD and graft-versus-leukemia responses following allogeneic hematopoietic cell transplantation (HCT). In addition, pDCs mediate antiviral immunity, particularly as they are the body's primary cellular source of type I interferon. Given their pleiotropic roles, pDCs have emerged as cells that critically impact transplant outcomes, including overall survival. In this article, we will review the pre-clinical and clinical literature, supporting the crucial roles that pDCs assume as key immune effector cells during HCT.
Collapse
|