1
|
Borghini N, Lazzaretti M, Lunghi P, Malpeli G, Barbi S, Perris R. A translational perspective of the malignant hematopoietic proteoglycome. Cell Biosci 2025; 15:25. [PMID: 39980017 PMCID: PMC11844096 DOI: 10.1186/s13578-025-01360-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
Proteoglycans are an ample family of complex extracellular matrix/cell surface components known to impact on virtually all biological processes that take place during life of a human being, in its healthy and diseased conditions. They are consolidated multivalent regulators of the behaviour of normal and malignant hematopoietic cells because of being critical components of their membranes, because of their pivotal role as multifaceted factors of the hematopoietic niches and because of acting as pillars of the tumour microenvironment. Likewise, they act as promoters of the growth, spreading and therapeutic resistance of diseased hematopoietic cells, also by modulating intracellular processes through a dual utilization of core protein domains and their glycosaminoglycan side chains. The intricate pattern of expression of the myriads of proteoglycan isoforms generated by differential glycanations of the core proteins is differentiation- and cell activation-dependent and often associates with genomic aberrations and gene amplifications. Selected proteoglycans stand out as widely recognized, disease type-specific markers and as alluring but still unappreciated therapeutic targets. We therefore pose here a clinical-translational view on the hematopoietic proteoglycome to highlight its underestimated biological and pathological significance during normal and neoplastic hematopoiesis. We underscore the potential of several proteoglycans to be exploited as key markers for prognostication and therapeutic targeting of hematopoietic cancers.
Collapse
Affiliation(s)
- Naomi Borghini
- COMT- Centre for Molecular and Translational Oncology, University of Parma, Parco Area delle Scienze, 11/A, Parma, 43124, Italy
- Department of Chemical and Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/A, Parma, 43124, Italy
| | - Mirca Lazzaretti
- COMT- Centre for Molecular and Translational Oncology, University of Parma, Parco Area delle Scienze, 11/A, Parma, 43124, Italy
- Department of Chemical and Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/A, Parma, 43124, Italy
| | - Paolo Lunghi
- COMT- Centre for Molecular and Translational Oncology, University of Parma, Parco Area delle Scienze, 11/A, Parma, 43124, Italy
- Department of Chemical and Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/A, Parma, 43124, Italy
| | - Giorgio Malpeli
- Department of Life Science, Health, and Health Professions, Link Campus University, Via del Casale di San Pio V, 44, Roma, 00165, Italy
| | - Stefano Barbi
- Department of Diagnostics and Public Health, University and Hospital Trust of Verona, Piazzale L.A. Scuro, 10, Verona, 37134, Italy
| | - Roberto Perris
- COMT- Centre for Molecular and Translational Oncology, University of Parma, Parco Area delle Scienze, 11/A, Parma, 43124, Italy.
- Department of Chemical and Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/A, Parma, 43124, Italy.
| |
Collapse
|
2
|
Scodellaro C, Pina RR, Ferreira FC, Sanjuan-Alberte P, Fernandes TG. Unlocking the Potential of Stem Cell Microenvironments In Vitro. Bioengineering (Basel) 2024; 11:289. [PMID: 38534563 DOI: 10.3390/bioengineering11030289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/07/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
The field of regenerative medicine has recently witnessed groundbreaking advancements that hold immense promise for treating a wide range of diseases and injuries. At the forefront of this revolutionary progress are stem cells. Stem cells typically reside in specialized environments in vivo, known as microenvironments or niches, which play critical roles in regulating stem cell behavior and determining their fate. Therefore, understanding the complex microenvironments that surround stem cells is crucial for advancing treatment options in regenerative medicine and tissue engineering applications. Several research articles have made significant contributions to this field by exploring the interactions between stem cells and their surrounding niches, investigating the influence of biomechanical and biochemical cues, and developing innovative strategies for tissue regeneration. This review highlights the key findings and contributions of these studies, shedding light on the diverse applications that may arise from the understanding of stem cell microenvironments, thus harnessing the power of these microenvironments to transform the landscape of medicine and offer new avenues for regenerative therapies.
Collapse
Affiliation(s)
- Chiara Scodellaro
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Raquel R Pina
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Paola Sanjuan-Alberte
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
3
|
Zhang YW, Mess J, Aizarani N, Mishra P, Johnson C, Romero-Mulero MC, Rettkowski J, Schönberger K, Obier N, Jäcklein K, Woessner NM, Lalioti ME, Velasco-Hernandez T, Sikora K, Wäsch R, Lehnertz B, Sauvageau G, Manke T, Menendez P, Walter SG, Minguet S, Laurenti E, Günther S, Grün D, Cabezas-Wallscheid N. Hyaluronic acid-GPRC5C signalling promotes dormancy in haematopoietic stem cells. Nat Cell Biol 2022; 24:1038-1048. [PMID: 35725769 PMCID: PMC9276531 DOI: 10.1038/s41556-022-00931-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/02/2022] [Indexed: 12/11/2022]
Abstract
Bone marrow haematopoietic stem cells (HSCs) are vital for lifelong maintenance of healthy haematopoiesis. In inbred mice housed in gnotobiotic facilities, the top of the haematopoietic hierarchy is occupied by dormant HSCs, which reversibly exit quiescence during stress. Whether HSC dormancy exists in humans remains debatable. Here, using single-cell RNA sequencing, we show a continuous landscape of highly purified human bone marrow HSCs displaying varying degrees of dormancy. We identify the orphan receptor GPRC5C, which enriches for dormant human HSCs. GPRC5C is also essential for HSC function, as demonstrated by genetic loss- and gain-of-function analyses. Through structural modelling and biochemical assays, we show that hyaluronic acid, a bone marrow extracellular matrix component, preserves dormancy through GPRC5C. We identify the hyaluronic acid-GPRC5C signalling axis controlling the state of dormancy in mouse and human HSCs.
Collapse
Affiliation(s)
- Yu Wei Zhang
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,International Max Planck Research School for Molecular and Cellular Biology (IMPRS-MCB), Freiburg, Germany
| | - Julian Mess
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,Spemann Graduate School for Biology and Medicine (SGBM), Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), Freiburg, Germany
| | - Nadim Aizarani
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,International Max Planck Research School for Molecular and Cellular Biology (IMPRS-MCB), Freiburg, Germany
| | - Pankaj Mishra
- Pharmaceutical Bioinformatics, University of Freiburg, Freiburg, Germany
| | - Carys Johnson
- Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Mari Carmen Romero-Mulero
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Jasmin Rettkowski
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,Spemann Graduate School for Biology and Medicine (SGBM), Freiburg, Germany
| | - Katharina Schönberger
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,International Max Planck Research School for Molecular and Cellular Biology (IMPRS-MCB), Freiburg, Germany
| | - Nadine Obier
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Karin Jäcklein
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Nadine M Woessner
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Spemann Graduate School for Biology and Medicine (SGBM), Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), Freiburg, Germany.,Signalling Research Center BIOSS, Freiburg, Germany
| | | | - Talia Velasco-Hernandez
- Josep Carreras Leukemia Research Institute-Campus Clinic and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Katarzyna Sikora
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Ralph Wäsch
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medical, University of Freiburg, Freiburg, Germany
| | - Bernhard Lehnertz
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Canada
| | - Guy Sauvageau
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Canada
| | - Thomas Manke
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Pablo Menendez
- Signalling Research Center BIOSS, Freiburg, Germany.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.,Spanish Network for Cancer Research (CIBER-ONC)-ISCIII, Barcelona, Spain
| | | | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), Freiburg, Germany.,Signalling Research Center BIOSS, Freiburg, Germany
| | - Elisa Laurenti
- Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Stefan Günther
- Pharmaceutical Bioinformatics, University of Freiburg, Freiburg, Germany
| | - Dominic Grün
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), Freiburg, Germany.,Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität, Würzburg, Germany.,Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Nina Cabezas-Wallscheid
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany. .,Centre for Integrative Biological Signalling Studies (CIBSS), Freiburg, Germany.
| |
Collapse
|
4
|
Schmaus A, Rothley M, Schreiber C, Möller S, Roßwag S, Franz S, Garvalov BK, Thiele W, Spataro S, Herskind C, Prunotto M, Anderegg U, Schnabelrauch M, Sleeman J. Sulfated hyaluronic acid inhibits the hyaluronidase CEMIP and regulates the HA metabolism, proliferation and differentiation of fibroblasts. Matrix Biol 2022; 109:173-191. [DOI: 10.1016/j.matbio.2022.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/12/2022] [Accepted: 04/04/2022] [Indexed: 12/23/2022]
|
5
|
Otaegi-Ugartemendia M, Matheu A, Carrasco-Garcia E. Impact of Cancer Stem Cells on Therapy Resistance in Gastric Cancer. Cancers (Basel) 2022; 14:cancers14061457. [PMID: 35326607 PMCID: PMC8946717 DOI: 10.3390/cancers14061457] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/04/2022] Open
Abstract
Gastric cancer (GC) is the fourth leading cause of cancer death worldwide, with an average 5-year survival rate of 32%, being of 6% for patients presenting distant metastasis. Despite the advances made in the treatment of GC, chemoresistance phenomena arise and promote recurrence, dissemination and dismal prognosis. In this context, gastric cancer stem cells (gCSCs), a small subset of cancer cells that exhibit unique characteristics, are decisive in therapy failure. gCSCs develop different protective mechanisms, such as the maintenance in a quiescent state as well as enhanced detoxification procedures and drug efflux activity, that make them insusceptible to current treatments. This, together with their self-renewal capacity and differentiation ability, represents major obstacles for the eradication of this disease. Different gCSC regulators have been described and used to isolate and characterize these cell populations. However, at the moment, no therapeutic strategy has achieved the effective targeting of gCSCs. This review will focus on the properties of cancer stem cells in the context of therapy resistance and will summarize current knowledge regarding the impact of the gCSC regulators that have been associated with GC chemoradioresistance.
Collapse
Affiliation(s)
| | - Ander Matheu
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (M.O.-U.); (A.M.)
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Estefania Carrasco-Garcia
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (M.O.-U.); (A.M.)
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-943-006296
| |
Collapse
|
6
|
The extracellular matrix of hematopoietic stem cell niches. Adv Drug Deliv Rev 2022; 181:114069. [PMID: 34838648 PMCID: PMC8860232 DOI: 10.1016/j.addr.2021.114069] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 12/21/2022]
Abstract
Comprehensive overview of different classes of ECM molecules in the HSC niche. Overview of current knowledge on role of biophysics of the HSC niche. Description of approaches to create artificial stem cell niches for several application. Importance of considering ECM in drug development and testing.
Hematopoietic stem cells (HSCs) are the life-long source of all types of blood cells. Their function is controlled by their direct microenvironment, the HSC niche in the bone marrow. Although the importance of the extracellular matrix (ECM) in the niche by orchestrating niche architecture and cellular function is widely acknowledged, it is still underexplored. In this review, we provide a comprehensive overview of the ECM in HSC niches. For this purpose, we first briefly outline HSC niche biology and then review the role of the different classes of ECM molecules in the niche one by one and how they are perceived by cells. Matrix remodeling and the emerging importance of biophysics in HSC niche function are discussed. Finally, the application of the current knowledge of ECM in the niche in form of artificial HSC niches for HSC expansion or targeted differentiation as well as drug testing is reviewed.
Collapse
|
7
|
Matteini F, Mulaw MA, Florian MC. Aging of the Hematopoietic Stem Cell Niche: New Tools to Answer an Old Question. Front Immunol 2021; 12:738204. [PMID: 34858399 PMCID: PMC8631970 DOI: 10.3389/fimmu.2021.738204] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/11/2021] [Indexed: 12/31/2022] Open
Abstract
The hematopoietic stem cell (HSC) niche is a specialized microenvironment, where a complex and dynamic network of interactions across multiple cell types regulates HSC function. During the last years, it became progressively clearer that changes in the HSC niche are responsible for specific alterations of HSC behavior. The aging of the bone marrow (BM) microenvironment has been shown to critically contribute to the decline in HSC function over time. Interestingly, while upon aging some niche structures within the BM are degenerated and negatively affect HSC functionality, other niche cells and specific signals are preserved and essential to retaining HSC function and regenerative capacity. These new findings on the role of the aging BM niche critically depend on the implementation of new technical tools, developed thanks to transdisciplinary approaches, which bring together different scientific fields. For example, the development of specific mouse models in addition to coculture systems, new 3D-imaging tools, ossicles, and ex-vivo BM mimicking systems is highlighting the importance of new technologies to unravel the complexity of the BM niche on aging. Of note, an exponential impact in the understanding of this biological system has been recently brought by single-cell sequencing techniques, spatial transcriptomics, and implementation of artificial intelligence and deep learning approaches to data analysis and integration. This review focuses on how the aging of the BM niche affects HSCs and on the new tools to investigate the specific alterations occurring in the BM upon aging. All these new advances in the understanding of the BM niche and its regulatory function on HSCs have the potential to lead to novel therapeutical approaches to preserve HSC function upon aging and disease.
Collapse
Affiliation(s)
- Francesca Matteini
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], Barcelona, Spain
| | - Medhanie A. Mulaw
- Institute for Molecular Medicine and Internal Medicine I, Ulm University and University Hospital Ulm, Ulm, Germany
| | - M. Carolina Florian
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], Barcelona, Spain
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
8
|
The role of vascular niche and endothelial cells in organogenesis and regeneration. Exp Cell Res 2020; 398:112398. [PMID: 33271129 DOI: 10.1016/j.yexcr.2020.112398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/08/2023]
Abstract
The term vascular niche indicate the physical and biochemical microenvironment around blood vessel where endothelial cells, pericytes, and smooth muscle cells organize themselves to form blood vessels and release molecules involved in the recruitment of hematopoietic stem cells, endothelial progenitor cells and mesenchymal stem cells. The vascular niche creates a permissive environment that enables different cell types to realize their developmental or regenerative programs. In this context, the proximity between the endothelium and the new-forming cellular components of organs suggests an essential role of endothelial cells in the organs maturation. Dynamic interactions between specific organ endothelial cells and different cellular conponents are crucial for different organ morphogenesis and function. Conversely, organs provide cues shaping vascular network structure.
Collapse
|
9
|
Beiseigel M, Simon BT, Michalak C, Stickney MJ, Jeffery U. Effect of peri-operative crystalloid fluid rate on circulating hyaluronan in healthy dogs: A pilot study. Vet J 2020; 267:105578. [PMID: 33375957 DOI: 10.1016/j.tvjl.2020.105578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022]
Abstract
Hypervolemia can damage the endothelial glycocalyx, a key regulator of vascular permeability, coagulation and inflammation. A starting peri-operative crystalloid fluid rate of 5mL/kg/h is recommended for healthy dogs undergoing elective procedures but higher rates continue to be commonly used. This study aimed to determine if a higher starting perioperative fluid rate was associated with a greater increase in plasma concentrations of hyaluronan, a marker correlated with glycocalyx damage, in systemically healthy dogs undergoing elective surgical procedures. Based on a sample size calculation, 38 dogs undergoing ovariohysterectomy or castration were randomly assigned to receive lactated Ringer's at a starting perioperative fluid rate of 10mL/kg/h (n=19) or 5mL/kg/h (n=19). Plasma hyaluronan concentrations were measured by ELISA in pre- and post-fluid therapy samples. There were no significant differences between groups in hyaluronan values before (baseline, P=0.52) or after perioperative fluid administration (P=0.62). Compared to respective baseline values, hyaluronan values significantly increased following 5 and 10ml/kg/h fluid administration (P=0.02 for both comparisons). This preliminary study identified an increase in hyaluronan over the course of fluid therapy with both the low and high fluid rate. One possible explanation is that both fluid rates contribute to glycocalyx disruption, but it should be emphasized that hyaluronan is not specific to the glycocalyx. Further studies are needed to determine the origin of the increased circulating hyaluronan and its clinical significance in dogs undergoing elective surgical procedures.
Collapse
Affiliation(s)
- M Beiseigel
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - B T Simon
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - C Michalak
- Veterinary Medical Teaching Hospital, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - M J Stickney
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - U Jeffery
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
10
|
Nadra K, André M, Marchaud E, Kestemont P, Braccini F, Cartier H, Kéophiphath M, Fanian F. A hyaluronic acid-based filler reduces lipolysis in human mature adipocytes and maintains adherence and lipid accumulation of long-term differentiated human preadipocytes. J Cosmet Dermatol 2020; 20:1474-1482. [PMID: 33150734 PMCID: PMC8246837 DOI: 10.1111/jocd.13794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/22/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022]
Abstract
The beneficial role of subcutaneous adipose tissue in skin rejuvenation derived from its capacity to fill the under‐layer volumes but also from its ability to regulate the extracellular matrix production by dermis fibroblasts. Hyaluronic acid (HA), a major component of the extracellular matrix, is a commonly used injectable dermal filler showing excellent efficiencies to maintain tissue augmentation even after its biodegradation. To improve their stability, the HA molecules can also be “cross‐linked” to each other. The effects of cross‐linked HA‐based fillers on the dermal structure are well known. For safety reasons, most of the physicians prefer to use the blunt cannula for injections. However, evidences showed that the cannula could not be located in the dermis, but it passes through immediate hypodermis and the long‐lasting effect of cross‐linked HA‐based fillers may be related to its effects on adipose tissue. To test whether cross‐linked HA has a direct effect on human adipocytes, we treated isolated adipocytes and precursors cells from human skin donors with cross‐linked HA. Biochemical and cellular analysis demonstrated that treatment by cross‐linked HA showed beneficial effects on differentiated cell adherence and survival as well as reduced basal and induced lipolysis in fully mature adipocytes. Taken together, these data showed that cross‐linked HA promoted cell adherence and preserved the adipogenic capacity of preadipocytes during prolonged cell culture, bringing additional evidences of the beneficial role of cross‐linked HA‐based fillers in maintenance of the subcutaneous fat mass. This first study could defend a preventive approach to facial volume loss during natural aging.
Collapse
|
11
|
Abbonante V, Di Buduo CA, Malara A, Laurent PA, Balduini A. Mechanisms of platelet release: in vivo studies and in vitro modeling. Platelets 2020; 31:717-723. [PMID: 32522064 DOI: 10.1080/09537104.2020.1774532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mechanisms related to platelet release in the context of the bone marrow niche are not completely known. In this review we discuss what has been discovered about four critical aspects of this process: 1) the bone marrow niche organization, 2) the role of the extracellular matrix components, 3) the mechanisms by which megakaryocytes release platelets and 4) the novel approaches to mimic the bone marrow environment and produce platelets ex vivo.
Collapse
Affiliation(s)
| | | | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia , Pavia, Italy
| | | | | |
Collapse
|
12
|
Reiprich S, Hofbauer E, Kiderlen S, Clausen-Schaumann H, Böcker W, Aszódi A, Schönitzer V. Adhesive Properties of the Hyaluronan Pericellular Coat in Hyaluronan Synthases Overexpressing Mesenchymal Stem Cells. Int J Mol Sci 2020; 21:ijms21113827. [PMID: 32481561 PMCID: PMC7312511 DOI: 10.3390/ijms21113827] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 05/26/2020] [Indexed: 12/24/2022] Open
Abstract
Hyaluronan (HA), a natural component of the extracellular matrix, is supposed to have a regulatory function in the stem cell niche. Bone marrow-derived human mesenchymal stem cells (hMSCs) are known to express all three hyaluronan synthases (HASes), which are responsible for HA production. HA is extruded into the extracellular matrix, but also stays bound to the plasma membrane forming a pericellular coat, which plays a key role during early cell adhesion. Since HAS isoenzymes, HAS1, HAS2 and HAS3, produce HA with different molecular weights, a difference in their role for cell adhesion is expected. Here, we transduced the immortalized hMSC cell line SCP1 to constitutively express eGFP-tagged HASes (SCP1-HAS-eGFP) by lentiviral gene transfer. The overexpression of the HAS-eGFP was shown on RNA and protein levels, HA was determined by ELISA and the stained HA-coat was analyzed using confocal microscopy. Time-lapse microscopy, spreading assay and single cell force spectroscopy using atomic force microscopy were applied to characterize adhesion of the different HAS transduced SCP1 cells. We showed in this study that HAS3 overexpressing cells formed the thickest pericellular coat compared with control or HAS1 and HAS2 transduced cells. Furthermore, SCP1-HAS3-eGFP displayed faster and stronger adhesion compared to cells overexpressing the other synthases or control cells. We conclude that overexpression of HASes in hMSCs differentially modulates their initial adhesive interactions with the substrate. This observation might be helpful in regenerative medicine goals.
Collapse
Affiliation(s)
- Sebastian Reiprich
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.R.); (E.H.); (W.B.); (A.A.)
| | - Eva Hofbauer
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.R.); (E.H.); (W.B.); (A.A.)
| | - Stefanie Kiderlen
- Center for Applied Tissue Engineering and Regenerative Medicine, Munich University of Applied Sciences, 80533 Munich, Germany; (S.K.); (H.C.-S.)
- Center for NanoScience, Ludwig-Maximilians-University, 80799 Munich, Germany
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine, Munich University of Applied Sciences, 80533 Munich, Germany; (S.K.); (H.C.-S.)
- Center for NanoScience, Ludwig-Maximilians-University, 80799 Munich, Germany
| | - Wolfgang Böcker
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.R.); (E.H.); (W.B.); (A.A.)
| | - Attila Aszódi
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.R.); (E.H.); (W.B.); (A.A.)
| | - Veronika Schönitzer
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.R.); (E.H.); (W.B.); (A.A.)
- Correspondence: ; Tel.: +49-89-4400-53147
| |
Collapse
|
13
|
Uslu M, Albayrak E, Kocabaş F. Temporal modulation of calcium sensing in hematopoietic stem cells is crucial for proper stem cell expansion and engraftment. J Cell Physiol 2020; 235:9644-9666. [PMID: 32394484 DOI: 10.1002/jcp.29777] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/25/2020] [Accepted: 04/29/2020] [Indexed: 12/11/2022]
Abstract
Hematopoietic stem cells (HSCs) are known to reside in a bone marrow (BM) niche, which is associated with relatively higher calcium content. HSCs sense and respond to calcium changes. However, how calcium-sensing components modulate HSC function and expansion is largely unknown. We investigated temporal modulation of calcium sensing and Ca2+ homeostasis during ex vivo HSC culture and in vivo. Murine BM-HSCs, human BM, and umbilical cord blood (UCB) mononuclear cells (MNCs) were treated with store-operated calcium entry (SOCE) inhibitors SKF 96365 hydrochloride (abbreviated as SKF) and 2-aminoethoxydiphenyl borate (2-APB). Besides, K+ channel inhibitor TEA chloride (abbreviated as TEA) was used to compare the relationship between calcium-activated potassium channel activities. Seven days of SKF treatment induced mouse and human ex vivo BM-HSC expansion as well as UCB-derived primitive HSC expansion. SKF treatment induced the surface expression of CaSR, CXCR4, and adhesion molecules on human hematopoietic stem and progenitor cells. HSCs expanded with SKF successfully differentiated into blood lineages in recipient animals and demonstrated a higher repopulation capability. Furthermore, modulation of SOCE in the BM-induced HSC content and differentially altered niche-related gene expression profile in vivo. Intriguingly, treatments with SOCE inhibitors SKF and 2-APB boosted the mouse BM mesenchymal stem cell (MSC) and human adipose-derived MSCs proliferation, whereas they did not affect the endothelial cell proliferation. These findings suggest that temporal modulation of calcium sensing is crucial in expansion and maintenance of murine HSCs, human HSCs, and mouse BM-MSCs function.
Collapse
Affiliation(s)
- Merve Uslu
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.,Graduate School of Natural and Applied Sciences, Yeditepe University, Istanbul, Turkey
| | - Esra Albayrak
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.,Graduate School of Natural and Applied Sciences, Yeditepe University, Istanbul, Turkey
| | - Fatih Kocabaş
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.,Graduate School of Natural and Applied Sciences, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
14
|
|
15
|
Lee-Sayer SSM, Dougan MN, Cooper J, Sanderson L, Dosanjh M, Maxwell CA, Johnson P. CD44-mediated hyaluronan binding marks proliferating hematopoietic progenitor cells and promotes bone marrow engraftment. PLoS One 2018; 13:e0196011. [PMID: 29684048 PMCID: PMC5912764 DOI: 10.1371/journal.pone.0196011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/04/2018] [Indexed: 12/15/2022] Open
Abstract
CD44 is a widely expressed cell adhesion molecule that binds to the extracellular matrix component, hyaluronan. However, this interaction is not constitutive in most immune cells at steady state, as the ability of CD44 to engage hyaluronan is highly regulated. While activated T cells and macrophages gain the ability to bind hyaluronan by CD44, the status in other immune cells is less studied. Here we found a percentage of murine eosinophils, natural killer and natural killer T cells were capable of interacting with hyaluronan at steady state. To further investigate the consequences of hyaluronan binding by CD44 in the hematopoietic system, point mutations of CD44 that either cannot bind hyaluronan (LOF-CD44) or have an increased affinity for hyaluronan (GOF-CD44) were expressed in CD44-deficient bone marrow. Competitive bone marrow reconstitution of irradiated mice revealed an early preference for GOF-CD44 over WT-CD44 expressing cells, and for WT-CD44 over LOF-CD44 expressing cells, in the hematopoietic progenitor cell compartment. The advantage of the hyaluronan-binding cells was observed in the hematopoietic stem and progenitor populations, and was maintained throughout the immune system. Hematopoietic stem cells bound minimal hyaluronan at steady state, and this was increased when the cells were induced to proliferate whereas multipotent progenitors had an increased ability to bind hyaluronan at steady state. In vitro, the addition of hyaluronan promoted their proliferation. Thus, proliferating hematopoietic progenitors bind hyaluronan, and hyaluronan binding cells have a striking competitive advantage in bone marrow engraftment.
Collapse
Affiliation(s)
- Sally S. M. Lee-Sayer
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
| | - Meghan N. Dougan
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
- Department of Pediatrics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, B.C., Canada
| | - Jesse Cooper
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
| | - Leslie Sanderson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
| | - Manisha Dosanjh
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
| | - Christopher A. Maxwell
- Department of Pediatrics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, B.C., Canada
| | - Pauline Johnson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, B.C., Canada
- * E-mail:
| |
Collapse
|
16
|
Mokhtari S, Colletti EJ, Atala A, Zanjani ED, Porada CD, Almeida-Porada G. Boosting Hematopoietic Engraftment after in Utero Transplantation through Vascular Niche Manipulation. Stem Cell Reports 2017; 6:957-969. [PMID: 27304918 PMCID: PMC4912311 DOI: 10.1016/j.stemcr.2016.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 02/07/2023] Open
Abstract
In utero hematopoietic stem/progenitor cell transplantation (IUHSCT) has only been fully successful in the treatment of congenital immunodeficiency diseases. Using sheep as a large animal model of IUHSCT, we demonstrate that administration of CD146+CXCL12+VEGFR2+ or CD146+CXCL12+VEGFR2− cells prior to, or in combination with, hematopoietic stem/progenitor cells (HSC), results in robust CXCL12 production within the fetal marrow environment, and significantly increases the levels of hematopoietic engraftment. While in the fetal recipient, donor-derived HSC were found to reside within the trabecular bone, the increased expression of VEGFR2 in the microvasculature of CD146+CXCL12+VEGFR2+ transplanted animals enhanced levels of donor-derived hematopoietic cells in circulation. These studies provide important insights into IUHSCT biology, and demonstrate the feasibility of enhancing HSC engraftment to levels that would likely be therapeutic in many candidate diseases for IUHSCT. After IUHSCT, HSC engraft in the trabecular bone of the metaphysis CD146++(+/−) cells engraft in diaphysis and make hematopoiesis-supporting cytokines Donor cell-derived CXCL12 and VEGFR2 significantly increase HSC engraftment IUHSCT of CD146+CXCL12+VEGFR2+ cells prior to HSC could be curative in several diseases
Collapse
Affiliation(s)
- Saloomeh Mokhtari
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, USA
| | - Evan J Colletti
- Experimental Station, University of Nevada Reno, Reno, NV 89503, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, USA
| | - Esmail D Zanjani
- Experimental Station, University of Nevada Reno, Reno, NV 89503, USA
| | - Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, USA
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, USA.
| |
Collapse
|
17
|
Domingues MJ, Cao H, Heazlewood SY, Cao B, Nilsson SK. Niche Extracellular Matrix Components and Their Influence on HSC. J Cell Biochem 2017; 118:1984-1993. [PMID: 28112429 DOI: 10.1002/jcb.25905] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 12/16/2022]
Abstract
Maintenance of hematopoietic stem cells (HSC) takes place in a highly specialized microenvironment within the bone marrow. Technological improvements, especially in the field of in vivo imaging, have helped unravel the complexity of the niche microenvironment and have completely changed the classical concept from what was previously believed to be a static supportive platform, to a dynamic microenvironment tightly regulating HSC homeostasis through the complex interplay between diverse cell types, secreted factors, extracellular matrix molecules, and the expression of different transmembrane receptors. To add to the complexity, non-protein based metabolites have also been recognized as a component of the bone marrow niche. The objective of this review is to discuss the current understanding on how the different extracellular matrix components of the niche regulate HSC fate, both during embryonic development and in adulthood. Special attention will be provided to the description of non-protein metabolites, such as lipids and metal ions, which contribute to the regulation of HSC behavior. J. Cell. Biochem. 118: 1984-1993, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mélanie J Domingues
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Huimin Cao
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Shen Y Heazlewood
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Benjamin Cao
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Susan K Nilsson
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
18
|
Tamma R, Ribatti D. Bone Niches, Hematopoietic Stem Cells, and Vessel Formation. Int J Mol Sci 2017; 18:ijms18010151. [PMID: 28098778 PMCID: PMC5297784 DOI: 10.3390/ijms18010151] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/14/2016] [Accepted: 01/09/2017] [Indexed: 02/06/2023] Open
Abstract
Bone marrow (BM) is a source of hematopoietic stem cells (HSCs). HSCs are localized in both the endosteum, in the so-called endosteal niche, and close to thin-walled and fenestrated sinusoidal vessel in the center of BM, in the so-called vascular niche. HSCs give rise to all types of mature blood cells through a process finely controlled by numerous signals emerging from the bone marrow niches where HSCs reside. This review will focus on the description of the role of BM niches in the control of the fate of HSCs and will also highlight the role of the BM niches in the regulation of vasculogenesis and angiogenesis. Moreover, alterations of the signals in niche microenvironment are involved in many aspects of tumor progression and vascularization and further knowledge could provide the basis for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, 70124 Bari, Italy.
- National Cancer Institute Giovanni Paolo II, 70124 Bari, Italy.
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, 70124 Bari, Italy.
- National Cancer Institute Giovanni Paolo II, 70124 Bari, Italy.
| |
Collapse
|
19
|
Levesque JP, Winkler IG. Cell Adhesion Molecules in Normal and Malignant Hematopoiesis: from Bench to Bedside. CURRENT STEM CELL REPORTS 2016. [DOI: 10.1007/s40778-016-0066-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Liu RM, Sun RG, Zhang LT, Zhang QF, Chen DX, Zhong JJ, Xiao JH. Hyaluronic acid enhances proliferation of human amniotic mesenchymal stem cells through activation of Wnt/β-catenin signaling pathway. Exp Cell Res 2016; 345:218-229. [PMID: 27237096 DOI: 10.1016/j.yexcr.2016.05.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/15/2016] [Accepted: 05/23/2016] [Indexed: 01/11/2023]
Abstract
This study investigated the pro-proliferative effect of hyaluronic acid (HA) on human amniotic mesenchymal stem cells (hAMSCs) and the underlying mechanisms. Treatment with HA increased cell population growth in a dose- and time-dependent manner. Analyses by flow cytometry and immunocytochemistry revealed that HA did not change the cytophenotypes of hAMSCs. Additionally, the osteogenic, chondrogenic, and adipogenic differentiation capabilities of these hAMSCs were retained after HA treatment. Moreover, HA increased the mRNA expressions of wnt1, wnt3a, wnt8a, cyclin D1, Ki-67, and β-catenin as well as the protein level of β-catenin and cyclin D1 in hAMSCs; and the nuclear localization of β-catenin was also enhanced. Furthermore, the pro-proliferative effect of HA and up-regulated expression of Wnt/β-catenin pathway-associated proteins - wnt3a, β-catenin and cyclin D1 in hAMSCs were significantly inhibited upon pre-treatment with Wnt-C59, an inhibitor of the Wnt/β-catenin pathway. These results suggest that HA may positively regulate hAMSCs proliferation through regulation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Ru-Ming Liu
- Guizhou Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi 563000, PR China
| | - Ren-Gang Sun
- Guizhou Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi 563000, PR China
| | - Ling-Tao Zhang
- Guizhou Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi 563000, PR China
| | - Qing-Fang Zhang
- Guizhou Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi 563000, PR China
| | - Dai-Xiong Chen
- Guizhou Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi 563000, PR China
| | - Jian-Jiang Zhong
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Jian-Hui Xiao
- Guizhou Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi 563000, PR China.
| |
Collapse
|
21
|
Simpson RML, Hong X, Wong MM, Karamariti E, Bhaloo SI, Warren D, Kong W, Hu Y, Xu Q. Hyaluronan Is Crucial for Stem Cell Differentiation into Smooth Muscle Lineage. Stem Cells 2016; 34:1225-38. [PMID: 26867148 PMCID: PMC4864761 DOI: 10.1002/stem.2328] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/09/2015] [Accepted: 12/09/2015] [Indexed: 01/16/2023]
Abstract
Deciphering the extracellular signals that regulate SMC differentiation from stem cells is vital to further our understanding of the pathogenesis of vascular disease and for development of cell-based therapies and tissue engineering. Hyaluronan (HA) has emerged as an important component of the stem cell niche, however its role during stem cell differentiation is a complicated and inadequately defined process. This study aimed to investigate the role of HA in embryonic stem cell (ESC) differentiation toward a SMC lineage. ESCs were seeded on collagen-IV in differentiation medium to generate ESC-derived SMCs (esSMCs). Differentiation coincided with increased HA synthase (HAS) 2 expression, accumulation of extracellular HA and its assembly into pericellular matrices. Inhibition of HA synthesis by 4-methylumbelliferone (4MU), removal of the HA coat by hyaluronidase (HYAL) or HAS2 knockdown led to abrogation of SMC gene expression. HA activates ERK1/2 and suppresses EGFR signaling pathways via its principle receptor, CD44. EGFR inactivation coincided with increased binding to CD44, which was further augmented by addition of high molecular weight (HMW)-HA either exogenously or via HAS2 overexpression through adenoviral gene transfer. HMW-HA-stimulated esSMCs displayed a functional role in vascular tissue engineering ex vivo, vasculogenesis in a matrigel plug model and SMC accumulation in neointimal lesions of vein grafts in mice. These findings demonstrate that HAS2-induced HA synthesis and organization drives ESC-SMC differentiation. Thus, remodeling of the HA microenvironment is a critical step in directing stem cell differentiation toward a vascular lineage, highlighting HA as a potential target for treatment of vascular diseases. Stem Cells 2016;34:1225-1238.
Collapse
Affiliation(s)
- Russell M L Simpson
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Xuechong Hong
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Mei Mei Wong
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Eirini Karamariti
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Shirin Issa Bhaloo
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Derek Warren
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yanhua Hu
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| | - Qingbo Xu
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, United Kingdom
| |
Collapse
|
22
|
Progress and obstacles towards generating hematopoietic stem cells from pluripotent stem cells. Curr Opin Hematol 2016; 22:317-23. [PMID: 26049752 DOI: 10.1097/moh.0000000000000147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Human pluripotent stem cells (PSCs) have the potential to provide an inexhaustible source of hematopoietic stem cells (HSCs) that could be used in disease modeling and in clinical applications such as transplantation. Although the goal of deriving definitive HSCs from PSCs has not been achieved, recent studies indicate that progress is being made. This review will provide information on the current status of deriving HSCs from PSCs, and will highlight existing challenges and obstacles. RECENT FINDINGS Recent strides in HSC generation from PSCs has included derivation of developmental intermediates, identification of transcription factors and small molecules that support hematopoietic derivation, and the development of strategies to recapitulate niche-like conditions. SUMMARY Despite considerable progress in defining the molecular events driving derivation of hematopoietic progenitor cells from PSCs, the generation of robust transplantable HSCs from PSCs remains elusive. We propose that this goal can be facilitated by better understanding of the regulatory pathways governing HSC identity, development of HSC supportive conditions, and examining the marrow homing properties of PSC-derived HSCs.
Collapse
|
23
|
Liang J, Jiang D, Noble PW. Hyaluronan as a therapeutic target in human diseases. Adv Drug Deliv Rev 2016; 97:186-203. [PMID: 26541745 PMCID: PMC4753080 DOI: 10.1016/j.addr.2015.10.017] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/07/2023]
Abstract
Accumulation and turnover of extracellular matrix is a hallmark of tissue injury, repair and remodeling in human diseases. Hyaluronan is a major component of the extracellular matrix and plays an important role in regulating tissue injury and repair, and controlling disease outcomes. The function of hyaluronan depends on its size, location, and interactions with binding partners. While fragmented hyaluronan stimulates the expression of an array of genes by a variety of cell types regulating inflammatory responses and tissue repair, cell surface hyaluronan provides protection against tissue damage from the environment and promotes regeneration and repair. The interactions of hyaluronan and its binding proteins participate in the pathogenesis of many human diseases. Thus, targeting hyaluronan and its interactions with cells and proteins may provide new approaches to developing therapeutics for inflammatory and fibrosing diseases. This review focuses on the role of hyaluronan in biological and pathological processes, and as a potential therapeutic target in human diseases.
Collapse
Affiliation(s)
- Jiurong Liang
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W Noble
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
24
|
Nelson MR, Roy K. Bone-marrow mimicking biomaterial niches for studying hematopoietic stem and progenitor cells. J Mater Chem B 2016; 4:3490-3503. [DOI: 10.1039/c5tb02644j] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review discusses the considerations and approaches that have been employed for designing biomaterial based cultures for replicating the hematopoietic stem and progenitor cell niche.
Collapse
Affiliation(s)
- Michael R. Nelson
- Wallace H. Coulter Department of Biomedical Engineering at the Georgia Tech and Emory University
- The Parker H. Petit Institute for Bioengineering and Biosciences
- Georgia Institute of Technology
- Atlanta
- USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering at the Georgia Tech and Emory University
- The Parker H. Petit Institute for Bioengineering and Biosciences
- Georgia Institute of Technology
- Atlanta
- USA
| |
Collapse
|
25
|
Cao H, Heazlewood SY, Williams B, Cardozo D, Nigro J, Oteiza A, Nilsson SK. The role of CD44 in fetal and adult hematopoietic stem cell regulation. Haematologica 2015; 101:26-37. [PMID: 26546504 DOI: 10.3324/haematol.2015.135921] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/11/2015] [Indexed: 01/19/2023] Open
Abstract
Throughout development, hematopoietic stem cells migrate to specific microenvironments, where their fate is, in part, extrinsically controlled. CD44 standard as a member of the cell adhesion molecule family is extensively expressed within adult bone marrow and has been previously reported to play important roles in adult hematopoietic regulation via CD44 standard-ligand interactions. In this manuscript, CD44 expression and function are further assessed and characterized on both fetal and adult hematopoietic stem cells. Using a CD44(-/-) mouse model, conserved functional roles of CD44 are revealed throughout development. CD44 is critical in the maintenance of hematopoietic stem and progenitor pools, as well as in hematopoietic stem cell migration. CD44 expression on hematopoietic stem cells as well as other hematopoietic cells within the bone marrow microenvironment is important in the homing and lodgment of adult hematopoietic stem cells isolated from the bone/bone marrow interface. CD44 is also involved in fetal hematopoietic stem cell migration out of the liver, via a process involving stromal cell-derived factor-1α. The absence of CD44 in neonatal bone marrow has no impact on the size of the long-term reconstituting hematopoietic stem cell pool, but results in an enhanced long-term engraftment potential of hematopoietic stem cells.
Collapse
Affiliation(s)
- Huimin Cao
- Manufacturing, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Australia Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Shen Y Heazlewood
- Manufacturing, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Australia Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Brenda Williams
- Manufacturing, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Australia Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Daniela Cardozo
- Manufacturing, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Australia Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Julie Nigro
- Manufacturing, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Australia
| | - Ana Oteiza
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Norway
| | - Susan K Nilsson
- Manufacturing, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Australia Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| |
Collapse
|
26
|
Poon GFT, Dong Y, Marshall KC, Arif A, Deeg CM, Dosanjh M, Johnson P. Hyaluronan Binding Identifies a Functionally Distinct Alveolar Macrophage-like Population in Bone Marrow-Derived Dendritic Cell Cultures. THE JOURNAL OF IMMUNOLOGY 2015; 195:632-42. [PMID: 26085682 DOI: 10.4049/jimmunol.1402506] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 05/13/2015] [Indexed: 12/23/2022]
Abstract
Although classical dendritic cells (DCs) arise from distinct progenitors in the bone marrow, the origin of inflammatory DCs and the distinction between monocyte-derived DCs and macrophages is less clear. In vitro culture of mouse bone marrow cells with GM-CSF is a well-established method to generate DCs, but GM-CSF has also been used to generate bone marrow-derived macrophages. In this article, we identify a distinct subpopulation of cells within the GM-CSF bone marrow-derived DC culture based on their ability to bind hyaluronan (HA), a major component of the extracellular matrix and ligand for CD44. HA identified a morphologically distinct subpopulation of cells within the immature DC population (CD11c(+) MHC II(mid/low)) that were CCR5(+)/CCR7(-) and proliferated in response to GM-CSF, but, unlike immature DCs, did not develop into mature DCs expressing CCR7 and high levels of MHC II, even after stimulation with LPS. The majority of these cells produced TNF-α in response to LPS but were unable to activate naive T cells, whereas the majority of mature DCs produced IL-12 and activated naive T cells. This HA binding population shared many characteristics with alveolar macrophages and was retained in the alveolar space after lung instillation even after LPS stimulation, whereas the MHC II(high) mature DCs were found in the draining lymph node. Thus, HA binding in combination with MHC II expression can be used to identify alveolar-like macrophages from GM-CSF-treated bone marrow cultures, which provides a useful in vitro model to study alveolar macrophages.
Collapse
Affiliation(s)
- Grace F T Poon
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Yifei Dong
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Kelsey C Marshall
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Arif Arif
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Christoph M Deeg
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Manisha Dosanjh
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Pauline Johnson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| |
Collapse
|
27
|
Abstract
Blood is renewed throughout the entire life. The stem cells of the blood, called hematopoietic stem cells (HSCs), are responsible for maintaining a supply of all types of fresh blood cells. In contrast to other stem cells, the clinical application of these cells is well established and HSC transplantation is an established life-saving therapy for patients suffering from haematological disorders. Despite their efficient functionality throughout life in vivo, controlling HSC behaviour in vitro (including their proliferation and differentiation) is still a major task that has not been resolved with standard cell culture systems. Targeted HSC multiplication in vitro could be beneficial for many patients, because HSC supply is limited. The biology of these cells and their natural microenvironment - their niche - remain a matter of ongoing research. In recent years, evidence has come to light that HSCs are susceptible to physical stimuli. This makes the regulation of HSCs by engineering physical parameters a promising approach for the targeted manipulation of these cells for clinical applications. Nevertheless, the biophysical regulation of these cells is still poorly understood. This review sheds light on the role of biophysical parameters in HSC biology and outlines which knowledge on biophysical regulation identified in other cell types could be applied to HSCs.
Collapse
Affiliation(s)
- C Lee-Thedieck
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany.
| | | |
Collapse
|
28
|
Chomel JC, Aggoune D, Sorel N, Turhan AG. [Chronic myeloid leukemia stem cells: cross-talk with the niche]. Med Sci (Paris) 2014; 30:452-61. [PMID: 24801043 DOI: 10.1051/medsci/20143004022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The physiological hematopoietic niche located in bone marrow is a pluricellular structure whose components are now well identified. Within this microenvironment, hematopoietic stem cells are in direct contact with mesenchymal stromal cells, osteoblasts and sinusoidal endothelial cells. These close relationships drive specialized cellular functions (proliferation/quiescence, differentiation/self-renewal) ensuring an efficient hematopoiesis. Chronic myeloid leukemia (CML) is a major model of leukemic hematopoiesis. The BCR-ABL1 tyrosine kinase, constitutively activated in CML, plays a critical role in the pathogenesis of the disease. An intensive cross-talk between CML progenitors and the components of the hematopoietic niche has recently been demonstrated. Consequently, the occurrence of the so-called leukemic niche promotes both the proliferation of myeloid cells and the maintenance of quiescent leukemic stem cells. This bone marrow niche could also protect CML stem cells from tyrosine kinase inhibitors and probably contribute to their resistance towards targeted therapies.
Collapse
Affiliation(s)
- Jean-Claude Chomel
- Service de cancérologie biologique, CHU de Poitiers, Poitiers, France - Inserm U935, université de Poitiers, France
| | | | - Nathalie Sorel
- Service de cancérologie biologique, CHU de Poitiers, Poitiers, France - Inserm U935, université de Poitiers, France
| | - Ali G Turhan
- Inserm U935, université de Poitiers, France - hôpitaux universitaires Paris-Sud, le Kremlin Bicêtre, France - Inserm U935, université Paris-Sud 11, Paris, France
| |
Collapse
|
29
|
Qu C, Rilla K, Tammi R, Tammi M, Kröger H, Lammi MJ. Extensive CD44-dependent hyaluronan coats on human bone marrow-derived mesenchymal stem cells produced by hyaluronan synthases HAS1, HAS2 and HAS3. Int J Biochem Cell Biol 2014; 48:45-54. [DOI: 10.1016/j.biocel.2013.12.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/27/2013] [Accepted: 12/23/2013] [Indexed: 12/30/2022]
|
30
|
Heazlewood SY, Oteiza A, Cao H, Nilsson SK. Analyzing hematopoietic stem cell homing, lodgment, and engraftment to better understand the bone marrow niche. Ann N Y Acad Sci 2014; 1310:119-28. [PMID: 24428368 DOI: 10.1111/nyas.12329] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The existence of a bone marrow (BM) niche--the location in which hematopoietic stem cells (HSCs) reside--was proposed more than 30 years ago. Recent data suggest that the interaction of HSCs with cellular and extracellular components within the BM is critical for HSC regulation. The tracking of immunofluorescently labeled, prospectively isolated HSCs to and within the BM cavity allows the assessment of the regulatory processes involved in (1) homing, which involves transendothelial migration into the BM; (2) lodgment, including transmarrow migration through the extravascular space; and (3) BM reconstitution. Together, such analyses provide a better understanding of the cellular and extracellular components involved in the regulation of HSC quiescence and differentiation. Homing and lodgment of transplanted HSCs, the first critical steps in engraftment, involve multiple interactions between HSCs and the BM microenvironment. Herein, we describe a refined method of analyzing homing efficiency and spatial distribution of HSCs harvested from endosteal and/or central BM regions; we also review alternate methods. Using these techniques, microenvironment modifications within the recipient or surface protein-expression modifications on donor HSCs in animal models provide insights into components influencing the homing, lodgment, and engraftment processes.
Collapse
Affiliation(s)
- Shen Y Heazlewood
- Materials Science and Engineering, Commonwealth Scientific and Industrial Research Organization (CSIRO), Melbourne, Australia
| | | | | | | |
Collapse
|
31
|
Turner LA, J. Dalby M. Nanotopography – potential relevance in the stem cell niche. Biomater Sci 2014; 2:1574-1594. [DOI: 10.1039/c4bm00155a] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nanotopographical cues observed in vivo (such as in the sinusoid and bone) closely resemble nanotopographies that in vitro have been shown to promote niche relevant stem cells behaviours; specifically, retention of multipotency and osteogenic differentiation on ordered and disordered nano-pits respectively. These and other observations highlight a potential role for nano topography in the stem cell niche.
Collapse
Affiliation(s)
- Lesley-Anne Turner
- Centre for Cell Engineering
- Institute of Molecular
- Cell and Systems Biology
- College of Medical
- Veterinary and Life Sciences
| | - Matthew J. Dalby
- Centre for Cell Engineering
- Institute of Molecular
- Cell and Systems Biology
- College of Medical
- Veterinary and Life Sciences
| |
Collapse
|
32
|
The role of Tenascin C in the lymphoid progenitor cell niche. Exp Hematol 2013; 41:1050-61. [PMID: 24084079 DOI: 10.1016/j.exphem.2013.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 09/20/2013] [Indexed: 11/21/2022]
Abstract
Hemopoietic stem cells (HSCs) are extrinsically controlled by the bone marrow (BM) microenvironment. Mice devoid of the extracellular matrix molecule Tenascin-C (TNC) were reported to develop normally. The current study explores the relationship between TNC and hemopoiesis, from HSCs within their niche to maturing progenitors in alternate niches. Although the absence of TNC did not alter the size of the BM stem cell pool, we report decreased thymic T cell progenitors with redistribution to other lymphoid organs, suggesting an anchoring role for TNC. TNC did not play an essential role in stem and progenitor cell homing to BM, but significantly altered lymphoid primed progenitor cell homing. These cells express the TNC receptor, integrin α9β1, with the same reduced homing evident in the absence of this integrin. The absence of TNC also resulted in an increased proportion and number of mature circulating T cells. In addition, the absence of TNC significantly impaired hemopoietic reconstitution after transplant and increased stem and progenitor cell mobilization. In summary, our analysis revealed unidentified roles for TNC in hemopoiesis: in lineage commitment of thymic T cell progenitors, peripheral T cell migration, and hemopoietic reconstitution.
Collapse
|
33
|
Cao H, Oteiza A, Nilsson SK. Understanding the role of the microenvironment during definitive hemopoietic development. Exp Hematol 2013; 41:761-8. [DOI: 10.1016/j.exphem.2013.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 06/17/2013] [Accepted: 06/17/2013] [Indexed: 01/29/2023]
|
34
|
Heazlewood SY, Neaves RJ, Williams B, Haylock DN, Adams TE, Nilsson SK. Megakaryocytes co-localise with hemopoietic stem cells and release cytokines that up-regulate stem cell proliferation. Stem Cell Res 2013; 11:782-92. [PMID: 23792434 DOI: 10.1016/j.scr.2013.05.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 04/23/2013] [Accepted: 05/14/2013] [Indexed: 12/20/2022] Open
Abstract
We report transplanted hemopoietic stem cells (HSC) preferentially lodge within two cells of mature megakaryocytes (MM). With both populations comprising ~0.2% of bone marrow cells, this strongly suggests a key functional interaction. HSC isolated from the endosteum (eLSKSLAM) showed significantly increased hemopoietic cell proliferation while in co-culture with MM. Furthermore, eLSKSLAM progeny retained HSC potential, maintaining long-term multi-lineage reconstitution capacity in lethally ablated recipients. Increased hemopoietic cell proliferation was not MM contact dependent and could be recapitulated with media supplemented with two factors identified in MM-conditioned media: insulin-like growth factor binding protein-3 (IGFBP-3) and insulin-like growth factor-1 (IGF-1). We demonstrate that HSC express the receptor for IGF-1 and that IGF-1/IGFBP-3 induced increased hemopoietic cell proliferation can be blocked by an anti-IGF-1 neutralising antibody. However, co-cultures of 8N, 16N or 32N MM with eLSKSLAM showed that MM of individual ploidy did not significantly increase hemopoietic cell proliferation. Our data suggests that MM are an important component of the HSC niche and regulate hemopoietic cell proliferation through cytokine release.
Collapse
Affiliation(s)
- Shen Y Heazlewood
- Materials Science and Engineering, Commonwealth Scientific and Industrial Research Organization, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
35
|
Hiraga T, Ito S, Nakamura H. Cancer stem-like cell marker CD44 promotes bone metastases by enhancing tumorigenicity, cell motility, and hyaluronan production. Cancer Res 2013; 73:4112-22. [PMID: 23633482 DOI: 10.1158/0008-5472.can-12-3801] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD44, an adhesion molecule that binds to the extracellular matrix, primarily to hyaluronan (HA), has been implicated in cancer cell migration, invasion, and metastasis. CD44 has also recently been recognized as a marker for stem cells of several types of cancer. However, the roles of CD44 in the development of bone metastasis are unclear. Here, we addressed this issue by using bone metastatic cancer cell lines, in which CD44 was stably knocked down. Tumor sphere formation and cell migration and invasion were significantly inhibited by CD44 knockdown. Furthermore, the downregulation of CD44 markedly suppressed tumorigenicity and bone metastases in nude mice. Of note, the number of osteoclasts decreased in the bone metastases. Microarray analysis revealed that the expression of HA synthase 2 was downregulated in CD44-knockdown cells. The localization of HA in the bone metastatic tumors was also markedly reduced. We then examined the roles of CD44-HA interaction in bone metastasis using 4-methylumbelliferone (4-MU), an inhibitor of HA synthesis. 4-MU decreased tumor sphere and osteoclast-like cell formation in vitro. Moreover, 4-MU inhibited bone metastases in vivo with reduced number of osteoclasts. These results collectively suggest that CD44 expression in cancer cells promotes bone metastases by enhancing tumorigenicity, cell migration and invasion, and HA production. Our results also suggest the possible involvement of CD44-expressing cancer stem cells in the development of bone metastases through interaction with HA. CD44-HA interaction could be a potential target for therapeutic intervention for bone metastases.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Nagano, Japan
| | | | | |
Collapse
|
36
|
Cavallo C, Desando G, Columbaro M, Ferrari A, Zini N, Facchini A, Grigolo B. Chondrogenic differentiation of bone marrow concentrate grown onto a hylauronan scaffold: Rationale for its use in the treatment of cartilage lesions. J Biomed Mater Res A 2012; 101:1559-70. [DOI: 10.1002/jbm.a.34460] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 09/07/2012] [Accepted: 09/14/2012] [Indexed: 01/22/2023]
|
37
|
Nishioka C, Ikezoe T, Furihata M, Yang J, Serada S, Naka T, Nobumoto A, Kataoka S, Tsuda M, Udaka K, Yokoyama A. CD34⁺/CD38⁻ acute myelogenous leukemia cells aberrantly express CD82 which regulates adhesion and survival of leukemia stem cells. Int J Cancer 2012; 132:2006-19. [PMID: 23055153 DOI: 10.1002/ijc.27904] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 09/25/2012] [Indexed: 12/22/2022]
Abstract
To identify molecular targets in leukemia stem cells (LSCs), this study compared the protein expression profile of freshly isolated CD34(+) /CD38(-) cells with that of CD34(+) /CD38(+) counterparts from individuals with acute myelogenous leukemia (n = 2, AML) using isobaric tags for relative and absolute quantitation (iTRAQ). A total of 98 proteins were overexpressed, while six proteins were underexpressed in CD34(+) /CD38(-) AML cells compared with their CD34(+) /CD38(+) counterparts. Proteins overexpressed in CD34(+) /CD38(-) AML cells included a number of proteins involved in DNA repair, cell cycle arrest, gland differentiation, antiapoptosis, adhesion, and drug resistance. Aberrant expression of CD82, a family of adhesion molecules, in CD34(+) /CD38(-) AML cells was noted in additional clinical samples (n = 12) by flow cytometry. Importantly, down-regulation of CD82 in CD34(+) /CD38(-) AML cells by a short hairpin RNA (shRNA) inhibited adhesion to fibronectin via up-regulation of matrix metalloproteinases 9 (MMP9) and colony forming ability of these cells as assessed by transwell assay, real-time RT-PCR, and colony forming assay, respectively. Moreover, we found that down-regulation of CD82 in CD34(+) /CD38(-) AML cells by an shRNA significantly impaired engraftment of these cells in severely immunocompromised mice. Taken together, aberrant expression of CD82 might play a role in adhesion of LSCs to bone marrow microenvironment and survival of LSCs. CD82 could be an attractive molecular target to eradicate LSCs.
Collapse
Affiliation(s)
- Chie Nishioka
- Department of Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kallinikou K, Anjos-Afonso F, Blundell MP, Ings SJ, Watts MJ, Thrasher AJ, Linch DC, Bonnet D, Yong KL. Engraftment defect of cytokine-cultured adult human mobilized CD34+ cells is related to reduced adhesion to bone marrow niche elements. Br J Haematol 2012; 158:778-87. [DOI: 10.1111/j.1365-2141.2012.09219.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 05/29/2012] [Indexed: 12/17/2022]
Affiliation(s)
| | - Fernando Anjos-Afonso
- Haematopoietic Stem Cell Laboratory; Cancer Research UK; London Research Institute; London; UK
| | - Michael P. Blundell
- Molecular Immunology Unit; Wolfson Centre for Gene Therapy of Childhood Disease and Centre for Immunodeficiency; UCL Institute of Child Health; London; UK
| | | | | | - Adrian J. Thrasher
- Molecular Immunology Unit; Wolfson Centre for Gene Therapy of Childhood Disease and Centre for Immunodeficiency; UCL Institute of Child Health; London; UK
| | - David C. Linch
- Department of Haematology; Cancer Institute; University College of London; London; UK
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory; Cancer Research UK; London Research Institute; London; UK
| | - Kwee L. Yong
- Department of Haematology; Cancer Institute; University College of London; London; UK
| |
Collapse
|
39
|
Woeckel VJ, Eijken M, van de Peppel J, Chiba H, van der Eerden BCJ, van Leeuwen JPTM. IFNβ impairs extracellular matrix formation leading to inhibition of mineralization by effects in the early stage of human osteoblast differentiation. J Cell Physiol 2012; 227:2668-76. [PMID: 21898404 DOI: 10.1002/jcp.23009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Osteoimmunology is an emerging field of research focused on the interaction of the immune system and bone. In this study we demonstrate that human osteoblasts are sensitive to the immune cytokine interferon (IFN)β. Osteoblasts respond to IFNβ as shown by the induction of several known IFN target genes such as interferon-induced (IFI) proteins (IFIT1, IFI44L), interferon-stimulated gene factor 3 (ISGF3) complex and the induction of IFNβ itself. We demonstrated that IFNβ has severe inhibitory effects on mineralization of osteoblast-derived extracellular matrix (ECM). Analysis of the timing of the IFNβ effects revealed that committed osteoblasts in early stage of differentiation are most sensitive to IFNβ inhibition of mineralization. A single IFNβ treatment was as effective as multiple treatments. During the progress of differentiation osteoblasts become desensitized for IFNβ. This pinpoints to a complex pattern of IFNβ sensitivity in osteoblasts. Focusing on early osteoblasts, we showed that IFNβ decreased gene expression of ECM-related genes, such as type I Collagen (COL1A1), fibronectin (FN1), fibullin (FBLN1), fibrillin (FBN2), and laminin (LAMA1). Additionally, ECM produced by IFNβ-treated osteoblasts contained less collagen protein. IFNβ stimulated gene expression of osteopontin (OPN), annexin2 (ANXA2), and hyaluronan synthase 1 (HAS1), which are important factors in the adhesion of hematopoietic stem cells (HSC) in the HSC niche. In conclusion, IFNβ directly modifies human osteoblast function by inhibiting ECM synthesis eventually resulting in delayed bone formation and mineralization and induces a HSC niche supporting phenotype. These effects are highly dependent on timing of treatment in the early phase of osteoblast differentiation.
Collapse
Affiliation(s)
- V J Woeckel
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
40
|
Darzynkiewicz Z, Balazs EA. Genome integrity, stem cells and hyaluronan. Aging (Albany NY) 2012; 4:78-88. [PMID: 22383371 PMCID: PMC3314170 DOI: 10.18632/aging.100438] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Accepted: 02/24/2012] [Indexed: 12/12/2022]
Abstract
Faithful preservation of genome integrity is the critical mission of stem cells as well as of germ cells. Reviewed are the following mechanisms involved in protecting DNA in these cells: (a) The efflux machinery that can pump out variety of genotoxins in ATP-dependent manner; (b) the mechanisms maintaining minimal metabolic activity which reduces generation of reactive oxidants, by-products of aerobic respiration; (c) the role of hypoxic niche of stem cells providing a gradient of variable oxygen tension; (d)(e) the presence of hyaluronan (HA) and HA receptors on stem cells and in the niche; (f) the role of role of HA in protecting DNA from oxidative damage; (g) the specific role of HA that may play a role protecting DNA in stem cells; (h) the interactions of HA with sperm cells and oocytes that also may shield their DNA from oxidative damage, and (e) mechanisms by which HA exerts the anti-oxidant activity. While HA has multitude of functions its anti-oxidant capabilities are often overlooked but may be of significance in preservation of integrity of stem and germ cells genome.
Collapse
Affiliation(s)
- Zbigniew Darzynkiewicz
- Brander Cancer Research Institute & Department of Pathology, New York Medical College, Valhalla, NY 10595, USA.
| | | |
Collapse
|
41
|
Votteler M, Kluger PJ, Walles H, Schenke-Layland K. Stem cell microenvironments--unveiling the secret of how stem cell fate is defined. Macromol Biosci 2011; 10:1302-15. [PMID: 20715131 DOI: 10.1002/mabi.201000102] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stem cells are defined as unspecialized cells that are capable of long term self-renewal and differentiation into specialized cell types. These unique properties make them an attractive cell source for regenerative medicine applications. Although the functions of various stem cells have been extensively studied in the development of organisms and in diseases, the specific factors and conditions that control stem cell fate, specifically the conditions that allow them to remain unspecialized, are not well studied. It has been suggested that adult stem cell survival and maintenance, as well as proliferation and differentiation, are controlled by the three-dimensional (3D) microenvironment, the so-called niche. Major functional niche components include supporting niche cells, growth-modulating soluble factors stored within the niches, and the extracellular matrix (ECM). In this article, we review work highlighting the growing complexity of stem cell-ECM interactions and their impact on the fields of biomaterials research and regenerative medicine.
Collapse
Affiliation(s)
- Miriam Votteler
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Department of Cell and Tissue Engineering, Nobelstrasse 12, 70569 Stuttgart, Germany
| | | | | | | |
Collapse
|
42
|
Abstract
A large body of evidence suggests hemopoietic stem cells (HSCs) exist in an endosteal niche close to bone, whereas others suggest that the HSC niche is intimately associated with vasculature. In this study, we show that transplanted hemopoietic stem and progenitor cells (HSPCs) home preferentially to the trabecular-rich metaphysis of the femurs in nonablated mice at all time points from 15 minutes to 15 hours after transplantation. Within this region, they exist in an endosteal niche in close association with blood vessels. The preferential homing of HSPCs to the metaphysis occurs rapidly after transplantation, suggesting that blood vessels within this region may express a unique repertoire of endothelial adhesive molecules. One candidate is hyaluronan (HA), which is highly expressed on the blood vessel endothelium in the metaphysis. Analysis of the early stages of homing and the spatial dis-tribution of transplanted HSPCs at the single-cell level in mice devoid of Has3-synthesized HA, provides evidence for a previously undescribed role for HA expressed on endothelial cells in directing the homing of HSPCs to the metaphysis.
Collapse
|
43
|
Jiang D, Liang J, Noble PW. Hyaluronan as an immune regulator in human diseases. Physiol Rev 2011; 91:221-64. [PMID: 21248167 DOI: 10.1152/physrev.00052.2009] [Citation(s) in RCA: 800] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Accumulation and turnover of extracellular matrix components are the hallmarks of tissue injury. Fragmented hyaluronan stimulates the expression of inflammatory genes by a variety of immune cells at the injury site. Hyaluronan binds to a number of cell surface proteins on various cell types. Hyaluronan fragments signal through both Toll-like receptor (TLR) 4 and TLR2 as well as CD44 to stimulate inflammatory genes in inflammatory cells. Hyaluronan is also present on the cell surface of epithelial cells and provides protection against tissue damage from the environment by interacting with TLR2 and TLR4. Hyaluronan and hyaluronan-binding proteins regulate inflammation, tissue injury, and repair through regulating inflammatory cell recruitment, release of inflammatory cytokines, and cell migration. This review focuses on the role of hyaluronan as an immune regulator in human diseases.
Collapse
Affiliation(s)
- Dianhua Jiang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
44
|
Lam BS, Adams GB. Hematopoietic stem cell lodgment in the adult bone marrow stem cell niche. Int J Lab Hematol 2011; 32:551-8. [PMID: 20682000 DOI: 10.1111/j.1751-553x.2010.01250.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Treatment of malignant blood disorders, such as leukemia, that can provide a better chance of long-term remission involves myeloablation followed by transplantation of matched donor hematopoietic stem cells (HSCs). For successful engraftment and re-establishment of hematopoiesis to occur in the recipient, the transplanted HSCs must first migrate from the blood circulation to the bone marrow (BM), a process known as homing, then localize and anchor in suitable microenvironments within the BM, a process known as lodgment. After lodgment, the specific fate of the transplanted HSCs is determined through complex, bidirectional interactions with various stromal cell components in the niche. Ultimately, these interactions dictate the clinical outcome of the transplantation. Through the use of transgenic mouse models, considerable evidence has been accumulated in an attempt to unveil the possible underlying mechanisms that govern these processes. Here, we will emphasize the major factors that are involved in the regulation of lodgment of transplanted HSCs. Specifically, we will first introduce early observations on the spatial distribution of hematopoietic progenitors within the BM, then we will discuss the soluble factors, chemokines, cell-cell interactions, and cell-matrix interactions that have been studied and known to influence the site of HSC lodgment within the BM following transplantation.
Collapse
Affiliation(s)
- B S Lam
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | |
Collapse
|
45
|
Sharma M, Afrin F, Satija N, Tripathi RP, Gangenahalli GU. Stromal-derived factor-1/CXCR4 signaling: indispensable role in homing and engraftment of hematopoietic stem cells in bone marrow. Stem Cells Dev 2011; 20:933-46. [PMID: 21186999 DOI: 10.1089/scd.2010.0263] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Homing and engraftment of hematopoietic stem/progenitor cells (HSPCs) in bone marrow is the major determining factor in success of hematopoietic stem cell transplantation. This is a complex, multistep process orchestrated by the coordinated interplay between adhesion molecules, cytokines, growth factors, and regulatory cofactors, many of which remain to be defined. Recent studies have highlighted the pivotal role of unique stromal-derived factor-1 (SDF-1)/CXCR4 signaling in the regulation of HSPC homing and subsequent engraftment. In addition, studies suggest that SDF-1/CXCR4 signaling acts as an essential survival-promoting factor of transplanted HSPCs as well as maintenance of quiescent HSCs in bone marrow niche. These pleiotropic effects exerted by SDF-1/CXCR4 axis make this unique signaling initiator very promising, not only for optimal hematopoietic reconstitution but also for the development of innovative approaches to achieve restoration, regeneration, or repair of other damaged tissues potentially amendable to reversal by stem cell transplantation. This goal can only be achieved when the role of SDF-1/CXCR4 axis in hematopoietic transplantation is clearly defined. Hence, this review presents current knowledge of the mechanisms through which SDF-1/CXCR4 signaling promotes restoration of hematopoiesis by regulating the homing and engraftment of HSPCs.
Collapse
Affiliation(s)
- Menka Sharma
- Stem Cell and Gene Therapy Research Group, Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Defense Research and Development Organization, New Delhi, India
| | | | | | | | | |
Collapse
|
46
|
Grassinger J, Nilsson SK. Methods to analyze the homing efficiency and spatial distribution of hematopoietic stem and progenitor cells and their relationship to the bone marrow endosteum and vascular endothelium. Methods Mol Biol 2011; 750:197-214. [PMID: 21618093 DOI: 10.1007/978-1-61779-145-1_14] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The tracking of immunofluorescent labeled hematopoietic stem and progenitor cells (HSC/HPC) within the bone marrow (BM) cavity allows the assessment of the regulatory processes involved in transendothelial migration, trans-marrow migration, and finally lodgement into the HSC niche. This is of interest as the extracellular and cellular components involved in the regulation of HSC quiescence and differentiation are still not completely understood. Homing of transplanted HSC is the first critical step in the interaction between HSC and the microenvironment of the BM. As a consequence, murine models allowing the evaluation of the structural relationship between migrating HSC, the endosteal bone surface, and the vascular components of the BM enhance our understanding of hematopoietic regulation.
Collapse
Affiliation(s)
- Jochen Grassinger
- CSIRO Molecular and Health Technologies, C/O Australian Stem Cell Centre, Monash University, Clayton, VIC, Australia
| | | |
Collapse
|
47
|
Pharmacologic modulation of the calcium-sensing receptor enhances hematopoietic stem cell lodgment in the adult bone marrow. Blood 2010; 117:1167-75. [PMID: 21076044 DOI: 10.1182/blood-2010-05-286294] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The ability of hematopoietic stem cells (HSCs) to undergo self-renewal is partly regulated by external signals originating from the stem cell niche. Our previous studies with HSCs obtained from fetal liver of mice deficient for the calcium-sensing receptor (CaR) have shown the crucial role of this receptor in HSC lodgment and engraftment in the bone marrow (BM) endosteal niche. Using a CaR agonist, Cinacalcet, we assessed the effects of stimulating the CaR on the function of murine HSCs. Our results show that CaR stimulation increases primitive hematopoietic cell activity in vitro, including growth in stromal cell cocultures, adhesion to extracellular matrix molecules such as collagen I and fibronectin, and migration toward the chemotactic stimulus, stromal cell-derived factor 1α. Receptor stimulation also led to augmented in vivo homing, CXCR4-mediated lodgment at the endosteal niche, and engraftment capabilities. These mechanisms by which stimulating the CaR dictates preferential localization of HSCs in the BM endosteal niche provide additional insights into the fundamental interrelationship between the stem cell and its niche. These studies also have implications in the area of clinical stem cell transplantation, where ex vivo modulation of the CaR may be envisioned as a strategy to enhance HSC engraftment in the BM.
Collapse
|
48
|
Lévesque JP, Helwani FM, Winkler IG. The endosteal 'osteoblastic' niche and its role in hematopoietic stem cell homing and mobilization. Leukemia 2010; 24:1979-92. [PMID: 20861913 DOI: 10.1038/leu.2010.214] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The concept of hematopoietic stem cell (HSC) niche was formulated in 1978, but HSC niches remained unidentified for the following two decades largely owing to technical limitations. Sophisticated live microscopy techniques and genetic manipulations have identified the endosteal region of the bone marrow (BM) as a preferential site of residence for the most potent HSC - able to reconstitute in serial transplants - with osteoblasts and their progenitors as critical cellular elements of these endosteal niches. This article reviews the path to the discovery of these endosteal niches (often called 'osteoblastic' niches) for HSC, what cell types contribute to these niches with their known physical and biochemical features. In the past decade, a first wave of research uncovered many mechanisms responsible for HSC homing to, and mobilization from, the whole BM tissue. However, the recent discovery of endosteal HSC niches has initiated a second wave of research focusing on the mechanisms by which most primitive HSC lodge into and migrate out of their endosteal niches. The second part of this article reviews the current knowledge of the mechanisms of HSC lodgment into, retention in and mobilization from osteoblastic niches.
Collapse
Affiliation(s)
- J-P Lévesque
- Biotherapies Program, Haematopoietic Stem Cell Laboratory, Mater Medical Research Institute, South Brisbane, Queensland, Australia.
| | | | | |
Collapse
|
49
|
Nair RR, Tolentino J, Hazlehurst LA. The bone marrow microenvironment as a sanctuary for minimal residual disease in CML. Biochem Pharmacol 2010; 80:602-12. [PMID: 20382130 PMCID: PMC3285111 DOI: 10.1016/j.bcp.2010.04.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 03/29/2010] [Accepted: 04/01/2010] [Indexed: 12/15/2022]
Abstract
Bcr-abl kinase inhibitors have provided proof of principal that targeted therapy holds great promise for the treatment of cancer. However, despite the success of these agents in treating chronic myelogenous leukemia (CML), the majority of patients continue to present with minimal residual disease contained within the bone marrow microenvironment. These clinical observations suggest that the bone marrow microenvironment may provide survival signals that contribute to the failure to eliminate minimal residual disease. The bone marrow microenvironment is comprised of multiple sub-domains which vary in cellular composition and gradients of soluble factors and matrix composition. Experimental evidence indicate that exposure of tumor cells to either bone marrow derived soluble factors or the extracellular matrix can confer a multi-drug resistance phenotype. Together, these data indicate that targeting such pathways may be a viable approach for increasing the efficacy of chemotherapy. Moreover, we propose that personalized medicine must go beyond understanding predictive models inherent to tumors but rather build predictive models that consider diversity in response due to interactions with the tumor microenvironment. Although review will focus on CML, understanding the contribution of the bone marrow microenvironment could contribute to rationale combination therapy in other types of leukemia, multiple myeloma and solid tumors which metastasize to the bone.
Collapse
Affiliation(s)
- Rajesh R. Nair
- Molecular Oncology Program, H Lee Moffitt Cancer Center, Tampa, FL 33612, United States
| | - Joel Tolentino
- Molecular Oncology Program, H Lee Moffitt Cancer Center, Tampa, FL 33612, United States
| | - Lori A. Hazlehurst
- Molecular Oncology Program, H Lee Moffitt Cancer Center, Tampa, FL 33612, United States
| |
Collapse
|
50
|
Phenotypically identical hemopoietic stem cells isolated from different regions of bone marrow have different biologic potential. Blood 2010; 116:3185-96. [PMID: 20631378 DOI: 10.1182/blood-2009-12-260703] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Hemopoietic stem cells (HSCs) reside within a specified area of the bone marrow (BM) cavity called a "niche" that modulates HSC quiescence, proliferation, differentiation, and migration. Our previous studies have identified the endosteal BM region as the site for the HSC niche and demonstrated that hemopoietic stem and progenitor populations (HSPCs, LSK) isolated from different BM regions exhibit significantly different hemopoietic potential. In this study, we have analyzed subpopulations of LSK cells isolated from different regions of the BM and showed that CD150(+)CD48(-)LSK HSCs within the endosteal BM region have superior proliferative capacity and homing efficiency compared with CD150(+)CD48(-)LSK HSCs isolated from the central BM. Furthermore, we show, for the first time, that a subset of CD150(+)CD48(+)LSK progenitor cells, previously defined as B-lymphoid primed hemopoietic cells, are capable of multilineage reconstitution, however, only when isolated from the endosteal region. In addition, we provide evidence for an unrecognized role of CD48 in HSC homing. Together, our data provide strong evidence that highly purified HSCs show functional differences depending on their origin within the BM and that the most primitive HSCs reside within the endosteal BM region.
Collapse
|